Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Thromb Haemost ; 2018 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-29883024

RESUMEN

Two basic carboxypeptidases, carboxypeptidase B2 (CPB2) and carboxypeptidase N (CPN) are present in plasma. CPN is constitutively active, whereas CPB2 circulates as a precursor, procarboxypeptidase B2 (proCPB2), that needs to be activated by the thrombin-thrombomodulin complex or plasmin bound to glycosaminoglycans. The substrate specificities of CPB2 and CPN are similar; they both remove C-terminal basic amino acids from bioactive peptides and proteins, thereby inactivating them. The complement cascade is a cascade of proteases and cofactors activated by pathogens or dead cells, divided into two phases, with the second phase only being triggered if sufficient C3b is present. Complement activation generates anaphylatoxins: C3a, which stimulates macrophages; and C5a, which is an activator and attractant for neutrophils. Pharmacological intervention with inhibitors has shown that CPB2 delays fibrinolysis, whereas CPN is responsible for systemic inactivation of C3a and C5a. Among mice genetically deficient in either CPB2 or CPN, in a model of hemolytic-uremic syndrome, Cpb2-/- mice had the worst disease, followed by Cpn-/- mice, with wild-type (WT) mice being the most protected. This model is driven by C5a, and shows that CPB2 is important in inactivating C5a. In contrast, when mice were challenged acutely with cobra venom factor, the reverse phenotype was observed; Cpn-/- mice had markedly worse disease than Cpb2-/- mice, and WT mice were resistant. These observations need to be confirmed in humans. Therefore, CPB2 and CPN have different roles. CPN inactivates C3a and C5a generated spontaneously, whereas proCPB2 is activated at specific sites, where it inactivates bioactive peptides that would overwhelm CPN.

2.
J Thromb Haemost ; 16(5): 991-1002, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29383821

RESUMEN

Essentials Two basic carboxypeptidases are present in plasma, B2 (CPB2) and N (CPN). Cpb2-/- and Cpn-/- mice were challenged in a hemolytic uremic syndrome (HUS) model vs. wild type. Cpb2-/- exacerbates HUS while Cpn-/- exacerbates cobra venom factor challenge vs. wild type mice. CPB2 and CPN have overlapping but non-redundant roles. SUMMARY: Background There are two basic carboxypeptidases in plasma. Carboxypeptidase B2 (CPB2) is activated from a circulating zymogen, proCPB2, and carboxypeptidase N (CPN) is constitutively active with both inactivating complement C3a and C5a. Aims To test the roles of CPB2 and CPN in complement-driven mouse models of cobra venom factor (CVF) challenge and hemolytic-uremic syndrome (HUS). Methods Cpb2-/- , Cpn-/- and wild-type (WT) mice were compared in an HUS model induced by Shiga toxin and lipopolysaccharide administration and following CVF administration. Results HUS was exacerbated in Cpb2-/- mice more than in Cpn-/- mice, compared with WT mice. Cpb2-/- mice developed the HUS clinical triad of microangiopathic hemolytic anemia, uremia and thrombocytopenia. Treatment with anti-C5 antibody improved survival of both Cpb2-/- and Cpn-/- mice. In contrast, when challenged acutely with CVF, the reverse phenotype was observed. Cpn-/- mice had markedly worse disease than Cpb2-/- mice, whereas the WT mice were resistant. Conclusions CPN and CPB2 play overlapping but non-redundant roles in regulating complement activation in vivo. The constitutively active CPN is key for inactivation of systemic C5a, whereas CPB2 functions as an on-demand supplementary anaphylatoxin inhibitor in inactivating excessive C5a formed locally.


Asunto(s)
Carboxipeptidasa B2/sangre , Activación de Complemento , Complemento C3/metabolismo , Complemento C5a/metabolismo , Síndrome Hemolítico-Urémico/enzimología , Lisina Carboxipeptidasa/sangre , Animales , Carboxipeptidasa B2/deficiencia , Carboxipeptidasa B2/genética , Activación de Complemento/efectos de los fármacos , Complemento C5a/antagonistas & inhibidores , Complemento C5a/inmunología , Inactivadores del Complemento/farmacología , Modelos Animales de Enfermedad , Venenos Elapídicos/toxicidad , Endotoxinas , Genotipo , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/inducido químicamente , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Lisina Carboxipeptidasa/deficiencia , Lisina Carboxipeptidasa/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteolisis , Toxina Shiga II
3.
J Thromb Haemost ; 13(6): 1090-102, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25851247

RESUMEN

BACKGROUND AND OBJECTIVES: Carboxypeptidase B2 (CPB2) is a basic carboxypeptidase with fibrin and complement C3a and C5a as physiological substrates. We hypothesized that in polymicrobial sepsis, CPB2-deficient mice would have sustained C5a activity, leading to disease exacerbation. METHODS: Polymicrobial sepsis was induced by cecal ligation and puncture (CLP). RESULTS: Contrary to our hypothesis, Cpb2(-/-) mice had significantly improved survival, with reduced lung edema, less liver and kidney damage, and less disseminated intravascular coagulation. Hepatic pro-CPB2 was induced by CLP, leading to increased pro-CPB2 levels. Thrombomodulin present on mesothelium supported thrombin activation of pro-CPB2. Both wild-type and Cpb2(-/-) animals treated with a C5a receptor antagonist had improved survival, demonstrating that C5a was detrimental in this model. Treatment with a fibrinolysis inhibitor, tranexamic acid, caused a decrease in survival in both genotypes; however, the Cpb2(-/-) animals retained their survival advantage. Administration of a C3a receptor antagonist exacerbated the disease in both wild-type and Cpb2(-/-) mice and eliminated the survival advantage of Cpb2(-/-) mice. C5a receptor is expressed in both peritoneal macrophages and neutrophils; in contrast, C3a receptor expression is restricted to peritoneal macrophages, and C3a induced signaling in macrophages but not neutrophils. CONCLUSIONS: While C5a exacerbates the peritonitis, resulting in a deleterious generalized inflammatory state, C3a activation of peritoneal macrophages may limit the initial infection following CLP, thereby playing a diametrically opposing protective role in this polymicrobial sepsis model.


Asunto(s)
Carboxipeptidasa B2/deficiencia , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Peritonitis/enzimología , Sepsis/enzimología , Animales , Antifibrinolíticos/farmacología , Trastornos de la Coagulación Sanguínea/enzimología , Trastornos de la Coagulación Sanguínea/genética , Trastornos de la Coagulación Sanguínea/inmunología , Trastornos de la Coagulación Sanguínea/microbiología , Carboxipeptidasa B2/genética , Ciego/microbiología , Ciego/cirugía , Células Cultivadas , Complemento C3a/antagonistas & inhibidores , Complemento C3a/inmunología , Complemento C5a/antagonistas & inhibidores , Complemento C5a/inmunología , Modelos Animales de Enfermedad , Activación Enzimática , Fibrina/metabolismo , Mediadores de Inflamación/sangre , Leucopenia/enzimología , Leucopenia/genética , Leucopenia/inmunología , Leucopenia/microbiología , Ligadura , Hígado/enzimología , Hígado/inmunología , Hígado/microbiología , Activación de Macrófagos , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/genética , Peritonitis/inmunología , Peritonitis/microbiología , Factores Protectores , Punciones , Factores de Riesgo , Sepsis/genética , Sepsis/inmunología , Sepsis/microbiología , Trombina/metabolismo , Trombomodulina/metabolismo , Factores de Tiempo
4.
J Thromb Haemost ; 8(5): 868-76, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20128866

RESUMEN

SUMMARY: Thrombin-activatable fibrinolysis inhibitor (TAFI) is a circulating zymogen that is activated physiologically by the thrombin/thrombomodulin complex to activated TAFI (TAFIa) which is a basic carboxypeptidase. Substrates include fibrin, leading to a reduction in rate of plasmin generation, and several proinflammatory mediators such as bradykinin, thrombin-cleaved osteopontin and complement factor C5a. TAFI-deficient mice have no phenotype without being challenged and TAFIa appears to play a limited role in physiological fibrinolysis in vivo. In several disease models, the TAFI-deficient mice have different outcomes from the wild type (WT), but whether the difference is beneficial or an exacerbation of the disease depends on the model. The consequences of TAFI deficiency include increased plasmin as a result of enhanced incorporation of plasminogen and tissue plasminogen activator into the fibrin clot, but also loss of its ability to degrade other substrates, with the resultant up-regulation of several proinflammatory mediators, including C5a. Criteria are recommended to demonstrate that a substrate is a physiological substrate of TAFIa.


Asunto(s)
Carboxipeptidasa B2/genética , Animales , Fibrinólisis , Ratones , Modelos Animales , Fenotipo , Trombosis , Cicatrización de Heridas
5.
J Thromb Haemost ; 8(3): 548-55, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20002544

RESUMEN

BACKGROUND: Polyphosphate (a linear polymer of inorganic phosphate) is secreted from platelet dense granules, and we recently showed that it accelerates factor V activation by thrombin. OBJECTIVE: To examine the interaction of polyphosphate with thrombin. METHODS AND RESULTS: Thrombin, but not prothrombin, altered the electrophoretic migration of polyphosphate in gel mobility assays. Thrombin binding to polyphosphate was influenced by ionic strength, and was evident even in plasma. Two positively charged exosites on thrombin mediate its interactions with other proteins and accessory molecules: exosite I (mainly with thrombin substrates), and exosite II (mainly with certain anionic polymers). Free thrombin, thrombin in complex with hirudin's C-terminal dodecapeptide and gamma-thrombin all bound polyphosphate similarly, excluding exosite I involvement. Mutations within exosite II, but not within exosite I, the Na(+)-binding site or hydrophobic pocket, weakened thrombin binding to polyphosphate as revealed by NaCl dependence. Surface plasmon resonance demonstrated tight interaction of polyphosphate with thrombin (K(d) approximately 5 nm) but reduced interaction with a thrombin exosite II mutant. Certain glycosaminoglycans, including heparin, only partially competed with polyphosphate for binding to thrombin, and polyphosphate did not reduce heparin-catalyzed inactivation of thrombin by antithrombin. CONCLUSION: Polyphosphate interacts with thrombin's exosite II at a site that partially overlaps with, but is not identical to, the heparin-binding site. Polyphosphate interactions with thrombin may be physiologically relevant, as the polyphosphate concentrations achievable following platelet activation are far above the approximately 5 nM K(d) for the polyphosphate-thrombin interaction.


Asunto(s)
Polifosfatos/metabolismo , Trombina/metabolismo , Antitrombinas/metabolismo , Sitios de Unión , Unión Competitiva , Ensayo de Cambio de Movilidad Electroforética , Heparina/metabolismo , Hirudinas/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Concentración Osmolar , Fragmentos de Péptidos/metabolismo , Polifosfatos/química , Unión Proteica , Conformación Proteica , Protrombina/metabolismo , Cloruro de Sodio/metabolismo , Resonancia por Plasmón de Superficie , Trombina/química , Trombina/genética
6.
J Thromb Haemost ; 5(7): 1486-92, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17635698

RESUMEN

BACKGROUND: Protein C inhibitor (PCI) and antithrombin (AT) are serine protease inhibitors (serpins) that inhibit a wide array of blood coagulation serine proteases including thrombin. OBJECTIVE: Fifty-five Ala-scanned recombinant thrombin mutants were used to determine thrombin residues important for inhibition by PCI with and without the cofactors heparin and thrombomodulin (TM) and compared with the prototypical serpin, AT. RESULTS: Residues around the active site (Tyr50 and Glu202) and the sodium-binding site (Glu229 and Arg233) were required for thrombin inhibition by PCI with and without cofactors. Exosite-2 residues (Arg89, Arg93, Glu94, Arg98, Arg245, Arg248, and Gln251) were critical for heparin-accelerated inhibition of thrombin by PCI. Exosite-1 residues (especially Lys65 and Tyr71) were required for enhanced PCI inhibition of thrombin-TM. Interestingly, we also found that the TM chondroitin sulfate moiety is not required for the approximately 150-fold enhanced rate of thrombin inhibition by PCI. Using the aforementioned thrombin exosite-2 mutants that were essential for heparin-catalyzed PCI-thrombin inhibition reactions we found no change in PCI inhibition rates for thrombin-TM. CONCLUSIONS: Collectively, these results show that (i) similar thrombin exosite-2 residues are critical for the heparin-catalyzed inhibition by PCI and AT, (ii) PCI and AT are different in their thrombin-TM inhibition properties, and (iii) PCI has a distinct advantage over AT in the regulation of the activity of thrombin-TM.


Asunto(s)
Heparina/metabolismo , Inhibidor de Proteína C/farmacología , Trombina/antagonistas & inhibidores , Trombina/genética , Trombomodulina/metabolismo , Sustitución de Aminoácidos , Sitios de Unión/genética , Heparina/farmacología , Humanos , Técnicas In Vitro , Modelos Moleculares , Complejos Multiproteicos , Mutagénesis Sitio-Dirigida , Inhibidor de Proteína C/genética , Conformación Proteica , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Trombina/química , Trombina/metabolismo
8.
J Biol Chem ; 276(27): 25143-9, 2001 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-11312264

RESUMEN

The interaction interface between human thrombin and human factor V (FV), necessary for complex formation and cleavage to generate factor Va, was investigated using a site-directed mutagenesis strategy. Fifty-three recombinant thrombins, with a total of 78 solvent-exposed basic and polar residues substituted with alanine, were used in a two-stage clotting assay with human FV. Seventeen mutants with less than 50% of wild-type (WT) thrombin FV activation were identified and mapped to anion-binding exosite I (ABE-I), anion-binding exosite II (ABE-II), the Leu(45)-Asn(57) insertion loop, and the Na(+) binding loop of thrombin. Three ABE-I mutants (R68A, R70A, and Y71A) and the ABE-II mutant R98A had less than 30% of WT activity. The thrombin Na(+) binding loop mutants, E229A and R233A, and the Leu(45)-Asn(57) insertion loop mutant, W50A, had a major effect on FV activation with 5, 15, and 29% of WT activity, respectively. The K52A mutant, which maps to the S' specificity pocket, had 29% of WT activity. SDS-polyacrylamide gel electrophoresis analysis of cleavage reactions using the thrombin ABE mutants R68A, Y71A, and R98A, the Na(+) binding loop mutant E229A, and the Leu(45)-Asn(57) insertion loop mutant W50A showed a requirement for both ABEs and the Na(+)-bound form of thrombin for efficient cleavage at the FV residue Arg(709). Several basic residues in both ABEs have moderate decreases in FV activation (40-60% of WT activity), indicating a role for the positive electrostatic fields generated by both ABEs in enhancing complex formation with complementary negative electrostatic fields generated by FV. The data show that thrombin activation of FV requires an extensive interaction interface with thrombin. Both ABE-I and ABE-II and the S' subsite are required for optimal cleavage, and the Na(+)-bound form of thrombin is important for its procoagulant activity.


Asunto(s)
Factor V/metabolismo , Trombina/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Humanos , Modelos Moleculares , Trombina/genética
9.
Proc Natl Acad Sci U S A ; 98(4): 1823-8, 2001 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-11172035

RESUMEN

Glycoprotein (GP) V is a major substrate cleaved by the protease thrombin during thrombin-induced platelet activation. Previous analysis of platelets from GP V-null mice suggested a role for GP V as a negative modulator of platelet activation by thrombin. We now report the mechanism by which thrombin activates GP V -/- platelets. We show that proteolytically inactive forms of thrombin induce robust stimulatory responses in GP V null mouse platelets, via the platelet GP Ib--IX--V complex. Because proteolytically inactive thrombin can activate wild-type mouse and human platelets after treatment with thrombin to cleave GP V, this mechanism is involved in thrombin-induced platelet aggregation. Platelet activation through GP Ib-IX depends on ADP secretion, and specific inhibitors demonstrate that the recently cloned P2Y(12) ADP receptor (G(i)-coupled ADP receptor) is involved in this pathway, and that the P2Y(1) receptor (G(q)-coupled ADP receptor) may play a less significant role. Thrombosis was generated in GP V null mice only in response to catalytically inactive thrombin, whereas thrombosis occurred in both genotypes (wild type and GP V null) in response to active thrombin. These data support a thrombin receptor function for the platelet membrane GP Ib--IX--V complex, and describe a novel thrombin signaling mechanism involving an initiating proteolytic event followed by stimulation of the GP Ib--IX via thrombin acting as a ligand, resulting in platelet activation.


Asunto(s)
Plaquetas/fisiología , Agregación Plaquetaria/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Trombina/metabolismo , Animales , Células CHO , Cricetinae , Heparina/metabolismo , Humanos , Ratones , Ratones Noqueados , Activación Plaquetaria/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transducción de Señal/fisiología
10.
Thromb Haemost ; 86(6): 1466-74, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11776315

RESUMEN

Thrombin binding to fibrin may be important in localizing thrombin to the site of vascular injury. However, fibrin-bound thrombin retains its catalytic activity toward fibrinogen, and may be prothrombotic under certain conditions. A collection of 52 purified thrombin mutants was used to identify those residues mediating the thrombin-fibrin interaction. Comparison of fibrinogen clotting activity with fibrin binding activity identified twenty residues involved in fibrinogen recognition with four of these residues important in fibrin binding (Lys65, His66, Tyr71, Arg73). No mutant was identified with normal clotting activity and deficient fibrin binding, suggesting that these two properties are not readily dissociable. A DNA thrombin aptamer that binds to these residues was able to inhibit the thrombin-fibrin interaction, and displace thrombin that was already bound. Mapping of these fibrin-binding residues on thrombin revealed that they are localized within exosite I, and comprise a subset of the residues important in fibrinogen recognition.


Asunto(s)
Fibrina/metabolismo , Trombina/metabolismo , Regulación Alostérica , Sustitución de Aminoácidos , Coagulación Sanguínea , Codón/genética , Relación Dosis-Respuesta a Droga , Fibrina/química , Fibrinógeno/metabolismo , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación Missense , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Mutación Puntual , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Trombina/química
11.
Trends Cardiovasc Med ; 10(2): 89-92, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11150736

RESUMEN

Thrombin is an allosteric enzyme that interacts with multiple procoagulant substrates such as specific clotting factors and cell surface thrombin receptors, as well as the anticoagulant substrate protein C. Functional mapping of thrombin's interactions with its various substrates has been carried out using a collection of thrombin mutants generated by systematic alanine scanning mutagenesis. A thrombin mutant, E229K, has been identified that has essentially lost all of its procoagulant properties while retaining its ability to activate protein C, thus functioning as an anticoagulant in vitro and in vivo. It is also found that specific and distinct domains are involved in thrombin's interaction with thrombomodulin (TM) and the subsequent activation by the thrombin/TM complex of protein C and the thrombin-activatable fibrinolysis inhibitor (TAFI).


Asunto(s)
Mutagénesis Sitio-Dirigida/fisiología , Trombina/metabolismo , Animales , Sitios de Unión/fisiología , Carboxipeptidasa B2 , Carboxipeptidasas/metabolismo , Interacciones Farmacológicas/fisiología , Activación Enzimática/fisiología , Humanos , Proteína C/metabolismo , Especificidad por Sustrato/fisiología , Trombomodulina/metabolismo
12.
J Biol Chem ; 274(36): 25510-6, 1999 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-10464282

RESUMEN

A collection of 56 purified thrombin mutants, in which 76 charged or polar surface residues on thrombin were mutated to alanine, was used to identify key residues mediating the interactions of thrombin with thrombomodulin (TM), protein C, and thrombin-activatable fibrinolysis inhibitor (TAFI). Comparison of protein C activation in the presence and absence of TM identified 11 residues mediating the thrombin-TM interaction (Lys(21), Gln(24), Arg(62), Lys(65), His(66), Arg(68), Thr(69), Tyr(71), Arg(73), Lys(77), Lys(106)). Three mutants (E25A, D51A, R89A/R93A/E94A) were found to have decreased ability to activate TAFI yet retained normal protein C activation, whereas three other mutants (R178A/R180A/D183A, E229A, R233A) had decreased ability to activate protein C but maintained normal TAFI activation. One mutant (W50A) displayed decreased activation of both substrates. Mapping of these functional residues on thrombin revealed that the 11 residues mediating the thrombin-TM interaction are all located in exosite I. Residues important in TAFI activation are located above the active-site cleft, whereas residues involved in protein C are located below the active-site cleft. In contrast to the extensive overlap of residues mediating TM binding and fibrinogen clotting, these data show that distinct domains in thrombin mediate its interactions with TM, protein C, and TAFI. These studies demonstrate that selective enzymatic properties of thrombin can be dissociated by site-directed mutagenesis.


Asunto(s)
Carboxipeptidasas/metabolismo , Fibrinólisis , Proteína C/metabolismo , Trombina/metabolismo , Trombomodulina/metabolismo , Animales , Sitios de Unión , Carboxipeptidasa B2 , Carboxipeptidasas/genética , Línea Celular , Humanos , Mutación Puntual , Unión Proteica , Proteína C/genética , Trombina/genética , Trombomodulina/genética
13.
Br J Haematol ; 102(4): 957-64, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9734646

RESUMEN

This study investigated the interaction of apoptotic polymorphonuclear neutrophils (PMN) with thrombospondin (TSP), an important event mediating the clearance of apoptotic neutrophils by macrophages. We developed an in vitro assay to examine this interaction. Based on this assay, we found that apoptotic but not fresh PMN bound specifically to surface-immobilized TSP (33 +/- 0.03 x 10(3) cells/well) compared to fibrinogen, fibronectin or laminin (8.0 +/- 0.3 x 10(3) cells/well). Moreover, the binding was specific for surface bound but not soluble TSP and appeared to be divalent cation dependent, was not significantly inhibited by heparin and was sensitive to cycloheximide (CHX) treatment of senescent PMN (>90%) inhibition at 10 microM CHX). In contrast to the binding studies, phagocytosis of senescent PMN by macrophages was not affected by EDTA or cycloheximide. Phosphatidyl-L-serine liposomes, phospho-L-serine, glucosamine, galactosamine, and the acetylated sugars had no effect on phagocytosis. We conclude that: (i) there was specific binding of senescent human PMN to immobilized TSP, which is divalent cation dependent and requires new protein synthesis in the PMN during senescence; (ii) in addition to the recently defined TSP-dependent pathway, there is a TSP-independent pathway mediating phagocytosis of senescent PMN by macrophages. The identity of this pathway remains to be defined.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Macrófagos/fisiología , Neutrófilos/metabolismo , Fagocitosis/fisiología , Trombospondinas/metabolismo , Carbohidratos/farmacología , Técnicas de Cultivo de Célula , Senescencia Celular , Cicloheximida/farmacología , Ácido Edético/farmacología , Heparina/farmacología , Humanos , Fagocitosis/efectos de los fármacos , Fosfatidilserinas/farmacología
14.
Cell Mol Life Sci ; 53(9): 731-6, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9368669

RESUMEN

Thrombin is a serine proteinase that can interact with a large number of diverse macromolecular substrates, which results in either a procoagulant or anticoagulant effect. These divergent properties are physiologically regulated by the endogenous protein thrombomodulin. This review summarizes recent work on a variety of methods used to exploit the allosteric nature of the enzyme. The procoagulant and anticoagulant functions of thrombin can be modulated by sodium binding, site-directed mutagenesis, and a small synthetic molecule. Modulation of thrombin's intrinsic properties represents a novel approach to the development of unique antithrombotic agents.


Asunto(s)
Anticoagulantes/química , Antitrombinas/química , Fibrinolíticos/química , Trombina/química , Regulación Alostérica , Animales , Antitrombina III/farmacología , Humanos , Conformación Proteica , Ingeniería de Proteínas , Relación Estructura-Actividad , Trombomodulina/fisiología
15.
Thromb Haemost ; 78(1): 577-80, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9198219

RESUMEN

The procoagulant and anticoagulant functions of thrombin are controlled physiologically by allosteric changes induced by Na+ and vascular cell-surface TM. Key residues that mediate Na+ interaction with thrombin have been identified. Based on a site-directed mutagenesis approach, E229K thrombin is found to be the most optimal and potent PC activator with a marked shift in substrate specificity for PC over fibrinogen. E229K thrombin demonstrates significant anticoagulant and antithrombotic efficacy in animal models in vivo. Alternatively, a synthetic organic molecule (LY254603) has been discovered which interacts with thrombin and effectively modulates its functions in vitro. This new class of antithrombotic agents exploits the powerful natural PC anticoagulant pathway and may have a superior therapeutic profile than direct thrombin inhibitors.


Asunto(s)
Anticoagulantes/metabolismo , Coagulación Sanguínea/fisiología , Trombina/metabolismo , Regulación Alostérica , Humanos , Proteína C/metabolismo , Ingeniería de Proteínas
17.
Biochemistry ; 35(51): 16449-57, 1996 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-8987977

RESUMEN

Previous alanine scanning mutagenesis of thrombin revealed that substitution of residues W50, K52, E229, and R233 (W60d, K60f, E217, and R221 in chymotrypsinogen numbering) with alanine altered the substrate specificity of thrombin to favor the anticoagulant substrate protein C. Saturation mutagenesis, in which residues W50, K52, E229, and R233 were each substituted with all 19 naturally occurring amino acids, resulted in the identification of a single mutation, E229K, that shifted the substrate specificity of thrombin by 130-fold to favor the activation of the anticoagulant substrate protein C over the procoagulant substrate fibrinogen. E229K thrombin was also less effective in activating platelets (18-fold), was resistant to inhibition by antithrombin III (33-fold and 22-fold in the presence and absence of heparin), and displayed a prolonged half-life in plasma in vitro (26-fold). Thus E229K thrombin displayed an optimal phenotype to function as a potent and specific activator of endogenous protein C and as an anticoagulant in vivo. Upon infusion in Cynomolgus monkeys E229K thrombin caused an anticoagulant effect through the activation of endogenous protein C without coincidentally stimulating fibrinogen clotting and platelet activation as observed with wild-type thrombin. In addition, E229K thrombin displayed enhanced potency in vivo relative to the prototype protein C activator E229A thrombin. This enhanced potency may be attributable to decreased clearance by antithrombin III, the principal physiological inhibitor of thrombin.


Asunto(s)
Anticoagulantes/farmacología , Ingeniería de Proteínas , Trombina/genética , Trombina/farmacología , Animales , Coagulación Sanguínea/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Fibrinógeno/metabolismo , Semivida , Humanos , Cinética , Macaca fascicularis , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Activación Plaquetaria/efectos de los fármacos , Proteína C/metabolismo , Conformación Proteica , Especificidad por Sustrato , Trombina/metabolismo
18.
J Biol Chem ; 271(26): 15381-5, 1996 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-8663130

RESUMEN

Clearance of apoptotic neutrophils by macrophages is a crucial event following the resolution of acute inflammation. CD36, together with alphavbeta3, has been identified as one of the adhesion molecules on the surface of macrophages implicated in the clearance of polymorphonuclear leukocytes. The domain on CD36 implicated in the phagocytosis of aged neutrophils remains to be elucidated. In this study, COS cells transfected with human CD36 cDNA had a significantly higher capacity to phagocytose human apoptotic neutrophils compared with murine CD36 cDNA. Moreover, monoclonal antibodies 10/5 or OKM5 (epitopes identified on amino acids 155-183) but not monoclonal antibody 13/10 (epitope identified on amino acids 30-76) inhibited phagocytosis of apoptotic neutrophils by COS cells transfected by human CD36. Swapping the human CD36 155-183 domain from human to murine CD36 (human-murine CD36 chimera) imparted to murine CD36-transfected COS cells an increased capacity to phagocytose apoptotic neutrophils. Conversely, when the murine domain 155-183 was inserted in human CD36, a decreased phagocytic capacity was observed. In addition, a synthetic peptide(155-169) but not its scrambled form significantly inhibited phagocytosis. These results identify for the first time a functional domain encompassing amino acids 155-183 on human CD36 implicated in the recognition and phagocytosis of apoptotic neutrophils.


Asunto(s)
Apoptosis , Antígenos CD36/química , Neutrófilos/fisiología , Fagocitosis , Animales , Chlorocebus aethiops , Humanos , Proteínas Recombinantes de Fusión/inmunología , Relación Estructura-Actividad , Transfección
19.
Biochemistry ; 34(46): 15328-33, 1995 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-7578149

RESUMEN

Tissue factor (TF) is a transmembrane protein that functions in the initiation of blood coagulation in vivo. At sites of vascular injury, TF serves as a cell-surface receptor for the serine protease factor VIIa (FVIIa), forming an enzyme--cofactor complex and enhancing the catalytic activity of FVIIa. Tissue factor, along with the receptors for alpha- and gamma-interferons, is a member of the class 2 cytokine receptor superfamily. Crystallographic analysis demonstrated that the extracellular domain of TF consists of two immunoglobulin-like domains joined by a linker region. Each domain is comprised of two antiparallel beta-sheets containing seven conserved beta-strands separated by more variable loop regions. Extensive mutagenesis has been performed in order to map the FVIIa binding site on TF. Results indicated that the discontinuous binding site for FVIIa lies at the domain--domain interface and includes residues from extended loops and beta-strands within both the N- and C-terminal domains. Our previous study provided evidence that three consecutive residues (D44, W45, K46) within the TF loop region between beta-strands C and C' of the N-terminal domain were important for interactions with FVIIa. We have presently extended our alanine-scanning mutagenesis to include the residues within the flanking beta-strands. Thirteen sTF mutants were screened for their ability to enhance FVIIa activity. Three residues within strand C (Y34, Q37, I38) and two residues within C' (K48, Y51) were shown to be important for TF cofactor function.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Factor VIIa/antagonistas & inhibidores , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/farmacología , Tromboplastina/antagonistas & inhibidores , Tromboplastina/química , Alanina , Secuencia de Aminoácidos , Sitios de Unión , Ciclización , Escherichia coli/genética , Factor VIIa/metabolismo , Expresión Génica , Datos de Secuencia Molecular , Mutagénesis , Conformación Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión , Solubilidad
20.
J Biol Chem ; 270(33): 19370-6, 1995 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-7642616

RESUMEN

The thrombin aptamer is a single-stranded DNA of 15 nucleotides that was identified by the selection of thrombin-binding molecules from a large combinatorial library of oligonucleotides. This prototype aptamer of thrombin has a unique double G-tetrad structure capable of inhibiting thrombin at nanomolar concentrations through binding to a specific region within thrombin exosite I. Substitution of arginine 70 in thrombin exosite I with glutamic acid effectively eliminated binding of the prototype thrombin aptamer. In contrast, aptamers selected against R70E thrombin were able to bind and inhibit both wild-type and R70E thrombins, and displayed potassium-independent inhibition. Aptamers selected against R70-E thrombin bound to sites identical or overlapping with that of the prototype thrombin aptamer. These aptamers retained the potential to form double G-tetrad structures; however, these structures would be destabilized by a T-->A substitution, disrupting the T4-T13 base pairing found in the prototype. This destabilization appeared to be partially compensated by newly recruited structural elements. Thus, selection against R70E thrombin did not lead to aptamers that bound to alternative sites, but instead to ssDNA structures with a suppressor mutation that accommodated the mutation in thrombin within a double G-tetrad context. These results provide insight into the aptamer-thrombin interaction and suggest that the binding site for the prototype is the dominant aptamorigenic site on thrombin.


Asunto(s)
Oligonucleótidos , Polinucleótidos/metabolismo , Supresión Genética , Trombina/metabolismo , Secuencia de Aminoácidos , Animales , Aptámeros de Nucleótidos , Secuencia de Bases , Sitios de Unión , Línea Celular , Cricetinae , Datos de Secuencia Molecular , Potasio/metabolismo , Trombina/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...