Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Phys Biol ; 8(1): 015014, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21301066

RESUMEN

The routine observation of tumor emboli in the peripheral blood of patients with carcinomas raises questions about the clinical relevance of these circulating tumor cells. Thrombosis is a common clinical manifestation of cancer, and circulating tumor cells may play a pathogenetic role in this process. The presence of coagulation-associated molecules on cancer cells has been described, but the mechanisms by which circulating tumor cells augment or alter coagulation remains unclear. In this study we utilized suspensions of a metastatic adenocarcinoma cell line, MDA-MB-231, and a non-metastatic breast epithelial cell line, MCF-10A, as models of circulating tumor cells to determine the thrombogenic activity of these blood-foreign cells. In human plasma, both metastatic MDA-MB-231 cells and non-metastatic MCF-10A cells significantly enhanced clotting kinetics. The effect of MDA-MB-231 and MCF-10A cells on clotting times was cell number-dependent and inhibited by a neutralizing antibody to tissue factor (TF) as well as inhibitors of activated factor X and thrombin. Using fluorescence microscopy, we found that both MDA-MB-231 and MCF-10A cells supported the binding of fluorescently labeled thrombin. Furthermore, in a model of thrombus formation under pressure-driven flow, MDA-MB-231 and MCF-10A cells significantly decreased the time to occlusion. Our findings indicate that the presence of breast epithelial cells in blood can stimulate coagulation in a TF-dependent manner, suggesting that tumor cells that enter the circulation may promote the formation of occlusive thrombi under shear flow conditions.


Asunto(s)
Adenocarcinoma/complicaciones , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/secundario , Trombosis/etiología , Coagulación Sanguínea , Línea Celular , Línea Celular Tumoral , Femenino , Humanos
2.
J Thromb Haemost ; 5 Suppl 1: 81-94, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17635714

RESUMEN

The specificity of blood coagulation proteinases for substrate, inhibitor, and effector recognition is mediated by exosites on the surfaces of the catalytic domains, physically separated from the catalytic site. Some thrombin ligands bind specifically to either exosite I or II, while others engage both exosites. The involvement of different, overlapping constellations of exosite residues enables binding of structurally diverse ligands. The flexibility of the thrombin structure is central to the mechanism of complex formation and the specificity of exosite interactions. Encounter complex formation is driven by electrostatic ligand-exosite interactions, followed by conformational rearrangement to a stable complex. Exosites on some zymogens are in low affinity proexosite states and are expressed concomitant with catalytic site activation. The requirement for exosite expression controls the specificity of assembly of catalytic complexes on the coagulation pathway, such as the membrane-bound factor Xa*factor Va (prothrombinase) complex, and prevents premature assembly. Substrate recognition by prothrombinase involves a two-step mechanism with initial docking of prothrombin to exosites, followed by a conformational change to engage the FXa catalytic site. Prothrombin and its activation intermediates bind prothrombinase in two alternative conformations determined by the zymogen to proteinase transition that are hypothesized to involve prothrombin (pro)exosite I interactions with FVa, which underpin the sequential activation pathway. The role of exosites as the major source of substrate specificity has stimulated development of exosite-targeted anticoagulants for treatment of thrombosis.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Coagulación Sanguínea , Factores de Coagulación Sanguínea/química , Humanos , Modelos Moleculares , Especificidad por Sustrato
3.
J Thromb Haemost ; 13 Suppl 1: S106-14, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26149011

RESUMEN

Group A streptococci (GAS) express soluble and surface-bound virulence factors. Secreted streptokinase (SK) allelic variants exhibit varying abilities to activate host plasminogen (Pg), and GAS pathogenicity is associated with Pg activation and localization of the resulting plasmin (Pm) on the bacterial surface to promote dissemination. The various mechanisms by which GAS usurp the host proteolytic system are discussed, including the molecular sexuality mechanism of conformational activation of the Pg zymogen (Pg*) and subsequent proteolytic activation of substrate Pg by the S•KPg* and SK•Pm catalytic complexes. Substantial progress has been made to delineate both processes in a unified mechanism. Pm coats the bacteria by direct and indirect binding pathways involving plasminogen-binding group A streptococcal M-like (PAM) protein and host fibrin(ogen). Transgenic mouse models using human Pg are being optimized to mimic infections by SK variants in humans and to define in vivo combined mechanisms of these variants and PAM.


Asunto(s)
Fibrinolisina/metabolismo , Fibrinólisis , Plasminógeno/metabolismo , Infecciones Estreptocócicas/sangre , Streptococcus pyogenes/enzimología , Estreptoquinasa/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Fibrina/metabolismo , Interacciones Huésped-Patógeno , Humanos , Modelos Moleculares , Unión Proteica , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/patogenicidad , Virulencia
4.
Protein Sci ; 10(9): 1897-904, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11514680

RESUMEN

Bothrojaracin (BJC) is a 27-kD snake venom protein from Bothrops jararaca that has been characterized as a potent thrombin inhibitor. BJC binds to exosites I and II, with a dissociation constant of 0.7 nM, and influences but does not block the proteinase catalytic site. BJC also binds prothrombin through an interaction that has not been characterized. In the present work we characterize the interaction of BJC with prothrombin quantitatively for the first time, and identify the BJC binding site on human prothrombin. Gel filtration chromatography demonstrated calcium-independent, 1:1 complex formation between fluorescein-labeled BJC ([5F]BJC) and prothrombin, whereas no interactions were observed with activation fragments 1 or 2 of prothrombin. Isothermal titration calorimetry showed that binding of BJC to prothrombin is endothermic, with a dissociation constant of 76 +/- 32 nM. The exosite I-specific ligand, hirudin(54-65) (Hir(54-65) (SO(3)(-)), displaced competitively [5F]BJC from prothrombin. Titration of the fluorescent hirudin(54-65) derivative, [5F]Hir(54-65)(SO(3)(-)), with human prothrombin showed a dissociation constant of 7.0 +/- 0.2 microM, indicating a approximately 100-fold lower binding affinity than that exhibited by BJC. Both ligands, however, displayed a similar, approximately 100-fold increase in affinity for exosite I when prothrombin was activated to thrombin. BJC efficiently displaced [5F]Hir(54-65)(SO(3)(-)) from complexes formed with thrombin or prothrombin with dissociation constants of 0.7 +/- 0.9 nM and 11 +/- 80 nM, respectively, indicating that BJC and Hir(54-65)(SO(3)(-)) compete for the same exosite on these molecules. The results indicate that BJC is a potent and specific probe of the partially exposed anion-binding exosite (proexosite I) of human prothrombin.


Asunto(s)
Bothrops , Venenos de Crotálidos/química , Venenos de Crotálidos/metabolismo , Protrombina/química , Protrombina/metabolismo , Animales , Unión Competitiva , Calorimetría , Cromatografía en Gel , Polarización de Fluorescencia , Humanos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Termodinámica , Trombina/antagonistas & inhibidores , Trombina/metabolismo
5.
FEBS Lett ; 494(1-2): 129-32, 2001 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-11297749

RESUMEN

The activation of the matrix metalloproteinase progelatinase A (MMP-2) has been of keen interest because an increase in MMP-2 activity has been implicated in disease states such as cancer and atherosclerosis. Activation of MMP-2 occurs on the surface of specific cell types in two steps. In the first step, primary cleavage of MMP-2 by a membrane-type matrix metalloproteinase generates an intermediate. A secondary cleavage and activation of the intermediate is thought to occur autocatalytically. Previous studies have shown that thrombin can also activate progelatinase A in the presence of endothelial cells. We show that this cell-dependent mechanism of MMP-2 activation also occurs with THP-1 cells and involves binding of thrombin to thrombomodulin present on the cell surface and generation of the anti-coagulant protein, activated protein C. We demonstrate that activated protein C is directly responsible for activation and cleavage of the gelatinase A intermediate. This work contributes new mechanistic insights into the activation of MMP-2 and provides a novel link between matrix metalloproteinase activation and anti-coagulation.


Asunto(s)
Precursores Enzimáticos/metabolismo , Gelatinasas/metabolismo , Metaloendopeptidasas/metabolismo , Proteína C/metabolismo , Trombina/metabolismo , Trombomodulina/metabolismo , Línea Celular , Células Cultivadas , Endotelio Vascular/citología , Activación Enzimática , Humanos
9.
Biochemistry ; 27(17): 6633-9, 1988 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-3219359

RESUMEN

The feasibility of a new approach to incorporation of spectroscopic probes into the active sites of certain serine proteases has been demonstrated. The method is based on inactivation of a serine protease with a thioester derivative of a peptide chloromethyl ketone. The thiol group generated by reaction of the covalent enzyme-inhibitor complex with NH2OH provides a unique site for subsequent labeling with thiol-reactive probes. To evaluate the method, N alpha-[(acetylthio)acetyl]-D-Phe-Pro-Arg-CH2Cl was synthesized by reaction of the thrombin-specific tripeptide chloromethyl ketone with succinimidyl (acetylthio)acetate and purified by sulfopropyl-Sephadex and Sephadex G-10 chromatography. Reverse-phase high-performance liquid chromatography indicated that the product was 90 +/- 3% pure. The compound was quantitated by using 5,5'-dithiobis(2-nitrobenzoic acid) to measure the concentration of thiol produced in the presence of NH2OH. On this basis, titrations of the irreversible loss of human alpha-thrombin activity had end points of 1.1 +/- 0.1 mol of inhibitor/mol of active sites, indicating a 1:1 stoichiometry for inactivation. Incubation of N alpha-[(acetylthio)acetyl]-D-Phe-Pro-Arg-thrombin with 5-(iodoacetamido)fluorescein in the presence of NH2OH resulted in incorporation of 0.96 mol of the fluorescence probe/mol of active sites and the appearance of fluorescein fluorescence associated with the active site containing B-chain on sodium dodecyl sulfate-polyacrylamide gels. Fluorescence labeling of thrombin required reaction of the inhibitor at the active site as well as subsequent generation of the thiol group with NH2OH. It is concluded that active site selective labeling can be achieved by using this approach, which is likely to be applicable to other proteases, peptide chloromethyl ketones, and a wide variety of probes.


Asunto(s)
Clorometilcetonas de Aminoácidos/metabolismo , Serina Endopeptidasas/metabolismo , Trombina/metabolismo , Clorometilcetonas de Aminoácidos/síntesis química , Sitios de Unión , Cromatografía Líquida de Alta Presión , Humanos , Indicadores y Reactivos , Cinética , Espectroscopía de Resonancia Magnética , Unión Proteica
10.
J Biol Chem ; 267(21): 14963-73, 1992 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-1634535

RESUMEN

In a new strategy for labeling the active sites of serine proteinases with fluorescence probes (Bock, P. E. (1988) Biochemistry 27, 6633-6639), a thioester peptide chloromethyl ketone inhibitor is incorporated into the enzyme active center and used to produce a unique thiol group which provides a site for selective chemical modification with any one of many thiol-reactive fluorescence probes. This approach was developed to increase the opportunities for identifying fluorescent proteinase derivatives that act as reporters of binding interactions by allowing a large number of derivatives, representing a broad range of probe spectral properties, to be readily prepared. In the studies described here, the specificity of the labeling approach was evaluated quantitatively for the labeling of human alpha and beta/gamma-thrombin with the thioester peptide chloromethyl ketones, N alpha-[(acetylthio)acetyl]-D-Phe-Pro-Arg-CH2Cl and N alpha-[(acetylthio)acetyl]-D-Phe-Phe-Arg-CH2Cl, and the thiol-reactive fluorescence probe, 5-(iodoacetamido)fluorescein. Irreversible inactivation of thrombin by the inhibitors was accompanied by incorporation of 0.98 +/- 0.06 mol/mol of the thioester group into the active site, independent of a 470-fold difference between the thioester peptide chloromethyl ketones in the bimolecular rate constants of alpha-thrombin affinity labeling. Subsequent mild treatment of the covalent thrombin-inhibitor complexes with NH2OH in the presence of 5-(iodoacetamido)fluorescein resulted in generation of the thiol group together with its selective modification and incorporation of 0.96 +/- 0.07 mol of probe/mol of active sites. The incorporated label was localized to a 9000 molecular weight region of alpha and beta/gamma-thrombin containing the catalytic-site histidine residue. Evaluation of competing, side reactions showed that they did not significantly compromise the active site specificity of labeling. These results demonstrated equivalent, active-site-selective fluorescence probe labeling of alpha and beta/gamma-thrombin by use of either of the thioester peptide chloromethyl ketones, with a site specificity of greater than or equal to 94%.


Asunto(s)
Clorometilcetonas de Aminoácidos/metabolismo , Precursores Enzimáticos/metabolismo , Protrombina/metabolismo , Trombina/metabolismo , Acetilación , Alquilación , Sitios de Unión , Electroforesis en Gel de Poliacrilamida , Colorantes Fluorescentes , Humanos , Hidrólisis , Cinética , Especificidad por Sustrato , Trombina/antagonistas & inhibidores
11.
J Biol Chem ; 267(21): 14974-81, 1992 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-1634536

RESUMEN

The behavior of an array of fluorescent human alpha-thrombin derivatives in reporting binding of the fragment 2 domain of prothrombin was characterized as a representative application of the active-site-selective labeling approach to studies of blood coagulation proteinase regulatory interactions. An array of 16 thrombin derivatives was prepared by affinity labeling of the proteinase active site with the thioester peptide chloromethyl ketones, N alpha-[(acetylthio)acetyl]-D-Phe-Pro-Arg-CH2Cl or N alpha-[(acetylthio)acetyl]-D-Phe-Phe-Arg-CH2Cl, followed by selective modification of the NH2OH-generated thiol group on the covalently incorporated inhibitors with each of eight thiol-reactive fluorescence probes. The changes in probe fluorescence intensity of the derivatives, signaling changes in the environment of the catalytic site associated with fragment 2 binding, appeared to be a unique and unpredictable function of the structure of the probe and the connecting peptide. These results demonstrated the utility of the labeling approach for overcoming the problem of not being able to predict which fluorescent label will provide the most useful proteinase derivative for investigating an interaction by enabling a greater variety of them to be prepared and screened for those with the most desirable properties. To determine whether the approach could be extended to other proteinases, the specificity of labeling with the fluorescence probe iodoacetamide, 5-(iodoacetamido)fluorescein, by use of the two thioester inhibitors was evaluated for several other blood coagulation proteinases and related trypsin-like enzymes. All of the proteinases were labeled in an active-site-selective manner. The combined results of quantitating the labeling reactions for the proteinase and inhibitor combinations studied thus far showed active-site-specific incorporation of 0.98 +/- 0.10 mol of inhibitor/mol of active sites and 0.92 +/- 0.11 mol of probe/mol of active sites, representing an overall greater than or equal to 93% site-specificity of labeling. These results demonstrated the broad applicability of the labeling approach for fluorescence studies of proteinases that differ greatly in their catalytic specificities.


Asunto(s)
Clorometilcetonas de Aminoácidos/metabolismo , Factores de Coagulación Sanguínea/metabolismo , Precursores Enzimáticos/metabolismo , Protrombina/metabolismo , Trombina/metabolismo , Sitios de Unión , Catálisis , Electroforesis en Gel de Poliacrilamida , Colorantes Fluorescentes , Humanos , Hidrólisis , Fragmentos de Péptidos/metabolismo , Especificidad por Sustrato
12.
Biochemistry ; 37(38): 13143-52, 1998 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-9748321

RESUMEN

In an essential step of blood coagulation, factor V is proteolytically processed by thrombin to generate the activated protein cofactor, factor Va, and to release the activation fragments E and C1. For the identification and characterization of sites of thrombin binding to factor V and its activation products, a new method was developed for immobilizing thrombin and other serine proteinases specifically (>/=92%) through their active sites and used in affinity chromatography studies of the interactions. Interactions of factor V with exosite I of thrombin were shown to regulate the factor V activation pathway from the 93% +/- 12% inhibition of the rate of activation correlated with specific binding of hirudin54-65 to this exosite. Chromatography of factor V on active-site-immobilized thrombin showed only a weak interaction, while the factor Va heterodimer bound specifically and with apparently higher affinity, in an interaction that was prevented by hirudin54-65. The heavy chain of subunit-dissociated factor Va bound to immobilized thrombin, while the light-chain subunit and fragment E had no detectable affinity. These results demonstrate a previously undescribed, exosite I-dependent interaction of thrombin with factor Va that occurs through the factor Va heavy chain. They support the further conclusion that similar exosite I-dependent binding of thrombin to the heavy-chain region of factor V contributes to recognition of factor V as a specific thrombin substrate and thereby regulates proteolytic activation of the protein cofactor.


Asunto(s)
Enzimas Inmovilizadas/metabolismo , Factor V/metabolismo , Factor Va/metabolismo , Serina Endopeptidasas/metabolismo , Trombina/metabolismo , Sitios de Unión/efectos de los fármacos , Cromatografía de Afinidad/métodos , Factor Va/química , Factor Va/efectos de los fármacos , Hirudinas/farmacología , Humanos , Hidrólisis , Fragmentos de Péptidos/farmacología , Sefarosa/metabolismo , Especificidad por Sustrato
13.
Anal Biochem ; 296(2): 254-61, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11554721

RESUMEN

Biotin derivatives of peptide chloromethyl ketones have ideal properties for specific labeling of the catalytic sites of serine proteinases but have not been widely used as probes because of the difficulty of synthesis and their instability. To make the reagents more accessible, a simple, economical method was developed for preparation of three biotin derivatives of the thrombin-specific inhibitor D-Phe-Pro-Arg-CH2Cl containing increasing lengths of the spacer connecting biotin. Reaction of the peptide with biotin-succinimidyl esters and purification by conventional chromatography yielded the compounds in 91-96% purity. The biotin-labeled inhibitors bound avidin with stoichiometries of 0.88-1.02 mol biotin compound/mol avidin subunits and irreversibly inactivated human thrombin with stoichiometries of 0.89-1.10 mol inhibitor/mol thrombin. Comparison of the three inhibitors by Western blotting indicated that a > or = 7- to 14-atom spacer was needed for sensitive (approximately 10 ng) detection of thrombin, with the derivative lacking a spacer only weakly detected because of its greatly reduced affinity for avidin. Application of the compounds to identify catalytically active products of factor Xa-catalyzed human prethrombin 1 activation in the absence of the protein cofactor, factor Va, allowed the direct observation of transient, low levels of the active intermediate, meizothrombin des-fragment 1, in addition to thrombin. Formation of this intermediate is concluded to reflect an intrinsic property of factor Xa activation of prethrombin 1 that is modulated by factor Va. The methods developed for preparation and characterization of the biotin-labeled inhibitors may be applicable to other tripeptide chloromethyl ketones, and the reagents can be employed for labeling of serine proteinases of diverse substrate specificity.


Asunto(s)
Biotina/química , Oligopéptidos/metabolismo , Serina Endopeptidasas/metabolismo , Trombina/metabolismo , Marcadores de Afinidad/química , Marcadores de Afinidad/metabolismo , Sitios de Unión , Precursores Enzimáticos/metabolismo , Oligopéptidos/química , Protrombina/metabolismo
14.
Anal Biochem ; 135(1): 172-9, 1983 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-6670737

RESUMEN

High-molecular-weight kininogen, a nonenzymatic glycoprotein of the intrinsic blood coagulation system, is proteolytically cleaved by kallikrein as an early event in the activation of this system. The light chain of cleaved kininogen retains the ability to form specific noncovalent complexes with prekallikrein and factor XI, other members of this system. We have determined the molecular weight of human kininogen light chain by equilibrium sedimentation in buffers of differing density, using an air-driven benchtop ultracentrifuge. The resulting molecular weight (30,500 +/- 800 g/mol) and partial specific volume (0.660 +/- 0.008 ml/g) are consistent with the idea that a sizeable fraction of the carbohydrate of high-molecular-weight kininogen is associated with the light chain. This level of precision is relatively easy to attain. The procedures are detailed, along with expressions for error propagation, to permit ready application of the technique.


Asunto(s)
Quininógenos , Aire , Humanos , Matemática , Peso Molecular , Espectrometría de Fluorescencia , Ultracentrifugación/métodos
15.
J Biol Chem ; 258(24): 15079-86, 1983 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-6558074

RESUMEN

Limited proteolysis of high molecular weight kininogen by kallikrein resulted in the generation of an inactive heavy chain of Mr = 64,000 and active light chains of Mr = 64,000 and 51,000 when analyzed by sodium dodecyl sulfate (SDS)-gel electrophoresis under reducing conditions. Starting with kininogen from outdated plasma, a light chain with an apparent molecular weight of 51,000 on 7.5% SDS gels was purified and characterized. Molecular weights of 28,900 +/- 1,100 and 30,500 +/- 1,600 were obtained by gel filtration of the reduced and alkylated protein in 6 M guanidine HCl and equilibrium sedimentation under nondenaturing conditions in the air-driven ultracentrifuge, respectively. The light chain stained positively with periodic acid-Schiff reagent on SDS gels indicating that covalently attached carbohydrate may be responsible for the anomalously high molecular weight estimated by SDS-gel electrophoresis. A single light chain thiol group reacted with 5,5'-dithiobis-(2-nitrobenzoic acid) (DTNB) in the presence and absence of 6 M guanidine HCl. Specific fluorescent labeling of the thiol group with 5-(iodoacetamido)fluorescein (IAF) occurred without loss of clotting activity. Addition of purified human plasma prekallikrein to the IAF-light chain resulted in a maximum increase in fluorescence anisotropy of 0.041 +/- 0.001 and no change in the fluorescence intensity. Fluorescence anisotropy measurements of the equilibrium binding of prekallikrein to the IAF-light chain yielded an average Kd of 17.3 +/- 2.5 nM and stoichiometry of 1.07 +/- 0.07 mol of prekallikrein/mol of IAF-light chain. Measurements of the interaction of prekallikrein with iodoacetamide-alkylated light chain using the IAF-light chain as a probe gave an average Kd of 16 +/- 4 nM and stoichiometry of 1.0 +/- 0.2 indicating indistinguishable affinities for prekallikrein.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Calicreínas/metabolismo , Quininógenos/metabolismo , Precalicreína/metabolismo , Ácido Ditionitrobenzoico/farmacología , Fluoresceínas/metabolismo , Polarización de Fluorescencia , Humanos , Matemática , Peso Molecular
16.
J Biol Chem ; 251(18): 5630-6, 1976 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-9393

RESUMEN

The kinetics of inactivation and reactivation of rabbit skeletal muscle phosphofructokinase have been studied as a function of pH and enzyme concentration at constant temperature in phosphate buffer. From the enzyme concentration dependence, we conclude that the minimal mechanism for inactivation involves a protonation step followed by isomerization to an inactive form and then dissociation to a species of one-half the molecular weight. Other data indicate a subsequent isomerization of the dissociated form. The pH and temperature dependence of the inactivation process shows that it is controlled by ionizable groups, and that the apparent pK for these groups is temperature-dependent in such a way as to make the enzyme show the characteristic of cold lability below pH 7. Reactivation of the inactive enzyme occurs by a kinetically different pathway involving deprotonation of an inactive, dissociated form to a form which may either isomerize to another inactive form, or dimerize to the active enzyme. A general mechanism is postulated in which the inactivation and reactivation processes are different aspects of the same mechanism. This mechanism assumes four species (two containing four subunits and two containing two subunits) each of which can exist in a protonated and unprotonated form. Inactivation or reactivation induced by changes in pH or temperature reflect the kinetic establishment of a new steady state between these forms. How the apparent pK values which control the distribution of the enzyme between protonated and unprotonated forms describe the pH-dependent characteristics of the enzyme is discussed in terms of the proposed mechanism.


Asunto(s)
Músculos/enzimología , Fosfofructoquinasa-1/metabolismo , Animales , Activación Enzimática , Concentración de Iones de Hidrógeno , Cinética , Conejos , Temperatura
17.
J Biol Chem ; 251(18): 5637-43, 1976 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-9394

RESUMEN

The effect of ligands, including substrates and allosteric effectors, on the pH-dependent inactivation and reactivation of rabbit muscle phosphofructokinase has been examined in terms of the mechanism proposed previously (Bock, P.E. and Fireden, C. (1976) J. Biol. Chem. 251, 5630-5636). It is concluded thatt many ligands exert their effect by binding preferentially to either protonated or unprotonated forms of the enzyme and thus shifting an apparent pK for the inactivation or reactivation process. ATP and fructose 6-phosphate influence the apparent pK to different extents and in different directions, with ATP binding preferentially to the protonated forms and fructose 6-phosphate to the unprotonated forms. Enzyme inactivated by ATP can be reactivated by the addition of fructose 6-phosphate. The experiments indicate that inactivation and reactivation in the presence of these ligands can occur by kinetically different pathways as has been found for these processes in the absence of ligands. The results are discussed in relation to what might be expected for ligand binding properties of the enzyme as a function of pH, temperature, and enzyme concentration. The effect of ATP and MgATP is complex, perhaps representing more than one site of binding. Citrate appears to bind preferentially to protonated forms of the enzyme while fructose 1,6-bisphosphate and AMP bind preferentially to the unprotonated forms. ADP, K+, and NH4+ appear to have little or no preference in binding to different enzyme forms.


Asunto(s)
Músculos/enzimología , Fosfofructoquinasa-1/metabolismo , Adenosina Trifosfato/farmacología , Animales , Sitios de Unión , Activación Enzimática , Concentración de Iones de Hidrógeno , Nucleótidos de Inosina/farmacología , Cinética , Ligandos , Magnesio/farmacología , Unión Proteica , Conformación Proteica , Conejos
18.
Biochemistry ; 39(45): 13974-81, 2000 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-11076540

RESUMEN

Binding of streptokinase (SK) to plasminogen (Pg) activates the zymogen conformationally and initiates its conversion into the fibrinolytic proteinase, plasmin (Pm). Equilibrium binding studies of SK interactions with a homologous series of catalytic site-labeled fluorescent Pg and Pm analogues were performed to resolve the contributions of lysine binding site interactions, associated changes between extended and compact conformations of Pg, and activation of the proteinase domain to the affinity for SK. SK bound to fluorescein-labeled [Glu]Pg(1) and [Lys]Pg(1) with dissociation constants of 624 +/- 112 and 38 +/- 5 nM, respectively, whereas labeled [Lys]Pm(1) bound with a 57000-fold tighter dissociation constant of 11 +/- 2 pM. Saturation of lysine binding sites with 6-aminohexanoic acid had no effect on SK binding to labeled [Glu]Pg(1), but weakened binding to labeled [Lys]Pg(1) and [Lys]Pm(1) 31- and 20-fold, respectively. At low Cl(-) concentrations, where [Glu]Pg assumes the extended conformation without occupation of lysine binding sites, a 23-fold increase in the affinity of SK for labeled [Glu]Pg(1) was observed, which was quantitatively accounted for by expression of new lysine binding site interactions. The results support the conclusion that the SK affinity for the fluorescent Pg and Pm analogues is enhanced 13-16-fold by conversion of labeled [Glu]Pg to the extended conformation of the [Lys]Pg derivative as a result of lysine binding site interactions, and is enhanced 3100-3500-fold further by the increased affinity of SK for the activated proteinase domain. The results imply that binding of SK to [Glu]Pg results in transition of [Glu]Pg to an extended conformation in an early event in the SK activation mechanism.


Asunto(s)
Dominio Catalítico , Lisina/metabolismo , Plasminógeno/química , Plasminógeno/metabolismo , Estreptoquinasa/química , Estreptoquinasa/metabolismo , Clorometilcetonas de Aminoácidos/metabolismo , Ácido Aminocaproico/química , Sitios de Unión , Cloruros/química , Fibrinolisina/metabolismo , Ácido Glutámico/metabolismo , Humanos , Conformación Proteica , Inhibidores de Serina Proteinasa/metabolismo
19.
Arch Biochem Biophys ; 286(2): 533-45, 1991 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-1897976

RESUMEN

A variation of the quantitative affinity chromatography (QAC) method of Winzor, Chaiken, and co-workers for the analysis of protein-ligand interactions has been developed and used to characterize sequence-specific and nonspecific protein-heparin interactions relevant to blood coagulation. The method allows quantitation of the binding of two components, A and B, from the competitive effect of one component, B, on the partitioning of the other component, A, between an immobilized acceptor phase and solution phase at equilibrium. Under the conditions employed, the differences in total A concentrations yielding an equivalent degree of saturation of the immobilized acceptor in the absence and presence of B defines the concentration of A bound to B in solution, thereby enabling conventional Scatchard or nonlinear least-squares analysis of the A-B equilibrium interaction. Like the QAC method, quantitation of the competitor interaction does not depend on the nature of the affinity matrix interaction, which need only be described empirically. The additional advantage of the difference method is that only the total rather than the free competitor ligand concentration need be known. The method requires that the partitioning component A be univalent, but allows for multivalency in the competitor, B, and can in principle be used to study binding interactions involving nonidentical, interacting, or nonspecific overlapping sites. Both the binding constant and the stoichiometry for the specific antithrombin-heparin interaction as well as the apparent binding constant for the nonspecific thrombin-heparin interaction at low thrombin binding densities obtained using this technique were in excellent agreement with values determined using spectroscopic probes.


Asunto(s)
Cromatografía de Afinidad/métodos , Heparina/metabolismo , Proteínas/metabolismo , Trombina/metabolismo , Antitrombina III/metabolismo , Unión Competitiva , Cinética , Matemática , Modelos Teóricos , Unión Proteica , Soluciones
20.
J Biol Chem ; 275(19): 14579-89, 2000 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-10799544

RESUMEN

Binding of streptokinase (SK) to plasminogen (Pg) conformationally activates the zymogen and converts both Pg and plasmin (Pm) into specific Pg activators. The interaction of SK with Pm and its relationship to the mechanism of Pg activation were evaluated in equilibrium binding studies with active site-labeled fluorescent Pm derivatives and in kinetic studies of SK-induced changes in the catalytic specificity of Pm. SK bound to fluorescein-labeled and native Pm with dissociation constants of 11 +/- 2 pm and 12 +/- 4 pm, which represented a 1,000-10,000-fold higher affinity than determined for Pg. Stoichiometric binding of SK to native Pm was followed by generation of a two-fragment form of SK cleaved at Lys(59) (SK'), which exhibited an indistinguishable affinity for labeled Pm, while a truncated, SK(55-414) species had a 120-360-fold reduced affinity. Binding of SK to native Pm was accompanied by a >50-fold enhancement in specificity for activation of Pg, which was paralleled by a surprising 2.6-10-fold loss of specificity of Pm for 8 of 11 tripeptide-pNA substrates. Further studies with Pm labeled at the active site with 2-anilinonaphthalene-6-sulfonic acid demonstrated directly that binding of SK to Pm resulted in expression of a new substrate binding exosite for Pg on the SK.Pm complex. It is concluded that SK activates Pg in part by preferential binding to the active zymogen conformation. High affinity binding of SK to Pm enhances Pg substrate specificity principally through emergence of a substrate recognition exosite.


Asunto(s)
Fibrinolisina/metabolismo , Plasminógeno/metabolismo , Estreptoquinasa/metabolismo , Animales , Sitios de Unión , Bovinos , Colorantes Fluorescentes , Humanos , Hidrólisis , Cinética , Proteínas Recombinantes/metabolismo , Espectrometría de Fluorescencia , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda