Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Biol Chem ; 293(2): 651-661, 2018 01 12.
Article de Anglais | MEDLINE | ID: mdl-29150443

RÉSUMÉ

Bispecific antibodies (bsAbs) combine the antigen specificities of two distinct Abs and demonstrate therapeutic promise based on novel mechanisms of action. Among the many platforms for creating bsAbs, controlled Fab-arm exchange (cFAE) has proven useful based on minimal changes to native Ab structure and the simplicity with which bsAbs can be formed from two parental Abs. Despite a published protocol for cFAE and its widespread use in the pharmaceutical industry, the reaction mechanism has not been determined. Knowledge of the mechanism could lead to improved yields of bsAb at faster rates as well as foster adoption of process control. In this work, a combination of Förster resonance energy transfer (FRET), nonreducing SDS-PAGE, and strategic mutation of the Ab hinge region was employed to identify and characterize the individual steps of cFAE. Fluorescence correlation spectroscopy (FCS) was used to determine the affinity of parental (homodimer) and bispecific (heterodimer) interactions within the CH3 domain, further clarifying the thermodynamic basis for bsAb formation. The result is a clear sequence of events with rate constants that vary with experimental conditions, where dissociation of the K409R parental Ab into half-Ab controls the rate of the reaction.


Sujet(s)
Anticorps bispécifiques/métabolisme , Fragments Fab d'immunoglobuline/métabolisme , Animaux , Humains , Cinétique , Spectrométrie de fluorescence
2.
J Biol Chem ; 288(39): 27789-800, 2013 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-23940050

RÉSUMÉ

Long-standing dogma proposes a profound contribution of membrane binding by prothrombin in determining the rate at which it is converted to thrombin by prothrombinase. We have examined the action of prothrombinase on full-length prothrombin variants lacking γ-carboxyglutamate modifications (desGla) with impaired membrane binding. We show an unexpectedly modest decrease in the rate of thrombin formation for desGla prothrombin but with a major effect on the pathway for substrate cleavage. Using desGla prothrombin variants in which the individual cleavage sites have been singly rendered uncleavable, we find that loss of membrane binding and other Gla-dependent functions in the substrate leads to a decrease in the rate of cleavage at Arg(320) and a surprising increase in the rate of cleavage at Arg(271). These compensating effects arise from a loss in the membrane component of exosite-dependent tethering of substrate to prothrombinase and a relaxation in the constrained presentation of the individual cleavage sites for active site docking and catalysis. Loss of constraint is evident as a switch in the pathway for prothrombin cleavage and the intermediate produced but without the expected profound decrease in rate. Extension of these findings to the action of prothrombinase assembled on platelets and endothelial cells on fully carboxylated prothrombin reveals new mechanistic insights into function on physiological membranes. Cell-dependent enzyme function is probably governed by a differential ability to support prothrombin binding and the variable accumulation of intermediates from the two possible pathways of prothrombin activation.


Sujet(s)
Membrane cellulaire/métabolisme , Prothrombine/composition chimique , Thromboplastine/composition chimique , Coagulation sanguine , Plaquettes/métabolisme , Proaccélérine/composition chimique , Facteur Va/composition chimique , Facteur Xa/composition chimique , Cellules endothéliales de la veine ombilicale humaine , Humains , Liaison aux protéines , Spécificité du substrat
3.
Biochim Biophys Acta ; 1823(11): 2079-86, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-22705352

RÉSUMÉ

As the importance of ubiquitylation in certain disease states becomes increasingly apparent, the enzymes responsible for removal of ubiquitin (Ub) from target proteins, deubiquitylases (DUBs), are becoming attractive targets for drug discovery. For rapid identification of compounds that alter DUB function, in vitro assays must be able to provide statistically robust data over a wide dynamic range of both substrate and enzyme concentrations during high throughput screening (HTS). The most established reagents for HTS are Ubs with a quenched fluorophore conjugated to the C-terminus; however, a luciferase-based strategy for detecting DUB activity (DUB-Glo™, Promega) provides a wider dynamic range than traditional fluorogenic reagents. Unfortunately, this assay requires high enzyme concentrations and lacks specificity for DUBs over other isopeptidases (e.g. desumoylases), as it is based on an aminoluciferin (AML) derivative of a peptide derived from the C-terminus of Ub (Z-RLRGG-). Conjugation of aminoluciferin to a full-length Ub (Ub-AML) yields a substrate that has a wide dynamic range, yet displays detection limits for DUBs 100- to 1000-fold lower than observed with DUB-Glo™. Ub-AML was even a sensitive substrate for DUBs (e.g. JosD1 and USP14) that do not show appreciable activity with DUB-Glo™. Aminoluciferin derivatives of hSUMO2 and NEDD8 were also shown to be sensitive substrates for desumoylases and deneddylases, respectively. Ub/Ubl-AML substrates are amenable to HTS (Z'=0.67) yielding robust signal, and providing an alternative drug discovery platform for Ub/Ubl isopeptidases. This article is part of a Special Issue entitled: Ubiquitin Drug Discovery and Diagnostics.


Sujet(s)
Endopeptidases/métabolisme , Tests de criblage à haut débit , Mesures de luminescence , Proteasome endopeptidase complex/métabolisme , Ubiquitine/métabolisme , Humains , Protéine NEDD8 , Sensibilité et spécificité , Petites protéines modificatrices apparentées à l'ubiquitine/métabolisme , Ubiquitines/métabolisme
4.
Methods Mol Biol ; 705: 15-30, 2011.
Article de Anglais | MEDLINE | ID: mdl-21125378

RÉSUMÉ

The preparation of sufficient amounts of high-quality protein samples is the major bottleneck for structural proteomics. The use of recombinant proteins has increased significantly during the past decades. The most commonly used host, Escherichia coli, presents many challenges including protein misfolding, protein degradation, and low solubility. A novel SUMO fusion technology appears to enhance protein expression and solubility ( http://www.lifesensors.com ). Efficient removal of the SUMO tag by SUMO protease in vitro facilitates the generation of target protein with a native N-terminus. In addition to its physiological relevance in eukaryotes, SUMO can be used as a powerful biotechnology tool for enhanced functional protein expression in prokaryotes and eukaryotes.


Sujet(s)
Escherichia coli/génétique , Pliage des protéines , Protéines de fusion recombinantes/biosynthèse , Protéine SUMO-1/biosynthèse , Animaux , Cellules eucaryotes/métabolisme , Exopeptidases/génétique , Exopeptidases/métabolisme , Humains , Protéines de fusion recombinantes/génétique , Protéine SUMO-1/génétique
5.
Mol Pharmacol ; 72(4): 897-906, 2007 Oct.
Article de Anglais | MEDLINE | ID: mdl-17622579

RÉSUMÉ

PAI-749 is a potent and selective synthetic antagonist of plasminogen activator inhibitor 1 (PAI-1) that preserved tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) activities in the presence of PAI-1 (IC(50) values, 157 and 87 nM, respectively). The fluorescence (Fl) of fluorophore-tagged PAI-1 (PAI-NBD119) was quenched by PAI-749; the apparent K(d) (254 nM) was similar to the IC(50) (140 nM) for PAI-NBD119 inactivation. PAI-749 analogs displayed the same potency rank order for neutralizing PAI-1 activity and perturbing PAI-NBD119 Fl; hence, binding of PAI-749 to PAI-1 and inactivation of PAI-1 activity are tightly linked. Exposure of PAI-1 to PAI-749 for 5 min (sufficient for full inactivation) followed by PAI-749 sequestration with Tween 80 micelles yielded active PAI-1; thus, PAI-749 did not irreversibly inactivate PAI-1, a known metastable protein. Treatment of PAI-1 with a PAI-749 homolog (producing less assay interference) blocked the ability of PAI-1 to displace p-aminobenzamidine from the uPA active site. Consistent with this observation, PAI-749 abolished formation of the SDS-stable tPA/PAI-1 complex. PAI-749-mediated neutralization of PAI-1 was associated with induction of PAI-1 polymerization as assessed by native gel electrophoresis. PAI-749 did not turn PAI-1 into a substrate for tPA; however, PAI-749 promoted plasmin-mediated degradation of PAI-1. In conclusion, PAI-1 inactivation by PAI-749 using purified components can result from a dual mechanism of action. First, PAI-749 binds directly to PAI-1, blocks PAI-1 from accessing the active site of tPA, and abrogates formation of the SDS-stable tPA/PAI-1 complex. Second, binding of PAI-749 to PAI-1 renders PAI-1 vulnerable to plasmin-mediated proteolytic degradation.


Sujet(s)
Indoles/pharmacologie , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Tétrazoles/pharmacologie , Biopolymères/métabolisme , Électrophorèse sur gel de polyacrylamide , Fluorescence , Humains , Hydrolyse , Micelles , Vitronectine/métabolisme , Vitronectine/pharmacologie
6.
Proc Natl Acad Sci U S A ; 102(29): 10099-104, 2005 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-16006504

RÉSUMÉ

Prothrombinase catalyzes thrombin formation by the ordered cleavage of two peptide bonds in prothrombin. Although these bonds are likely approximately 36 A apart, sequential cleavage of prothrombin at Arg-320 to produce meizothrombin, followed by its cleavage at Arg-271, are both accomplished by equivalent exosite interactions that tether each substrate to the enzyme and facilitate presentation of the scissile bond to the active site of the catalyst. We show that impairing the conformational transition from zymogen to active proteinase that accompanies the formation of meizothrombin has no effect on initial cleavage at Arg-320 but inhibits subsequent cleavage at Arg-271. Full thermodynamic rescue of this defective mutant was achieved by stabilizing the proteinase-like conformation of the intermediate with a reversible, active site-specific inhibitor. Irreversible stabilization of intact prothrombin in a proteinase-like state, even without prior cleavage at Arg-320, also enhanced cleavage at Arg-271. Our results indicate that the sequential presentation and cleavage of the two scissile bonds in prothrombin activation is accomplished by substrate bound either in the zymogen or proteinase conformations. The ordered cleavage of prothrombin by prothrombinase is driven by ratcheting of the substrate from the zymogen to the proteinase-like states.


Sujet(s)
Proenzymes/métabolisme , Modèles moléculaires , Conformation des protéines , Prothrombine/métabolisme , Thrombine/métabolisme , Thromboplastine/métabolisme , Sites de fixation , Activation enzymatique/physiologie , Proenzymes/génétique , Fluorescence , Humains , Cinétique , Mutation/génétique , Nitracrine/analogues et dérivés , Relation structure-activité , Spécificité du substrat , Thrombine/génétique , Thromboplastine/génétique
7.
J Biol Chem ; 279(52): 54927-36, 2004 Dec 24.
Article de Anglais | MEDLINE | ID: mdl-15494418

RÉSUMÉ

Thrombin formation results from cleavage of prothrombin following Arg(271) and Arg(320). Both bonds are accessible for cleavage, yet the sequential action of prothrombinase on Arg(320) followed by Arg(271) is implied by the intermediate observed during prothrombin activation. We have studied the individual cleavage reactions catalyzed by prothrombinase by using a series of recombinant derivatives: wild type prothrombin (II(WT)) contained both cleavage sites; II(Q271) contained a single cleavable site at Arg(320); II(Q320) and II(A320) contained a single cleavable site at Arg(271); and II(QQ) was resistant to cleavage. Cleavage at Arg(320) in II(Q271) could account for the initial cleavage reaction leading to the consumption of either plasma prothrombin or II(WT), whereas cleavage at Arg(271) in either II(Q320) or II(A320) was found to be approximately 30-fold slower. Equivalent kinetic constants were obtained for three of the four possible half-reactions. Slow cleavage at Arg(271) in intact prothrombin resulted from an approximately 30-fold reduction in V(max). Thus, the observed pathway of bond cleavage by prothrombinase can be explained by the kinetic constants for the four possible individual cleavage reactions. II(Q320) was a competitive inhibitor of II(Q271) cleavage, and II(QQ) was a competitive inhibitor for each reaction with K(i) approximately K(m). The data are inconsistent with previous proposals and suggest a model in which substrates for each of the four possible half-reactions bind in a mutually exclusive manner and with equal affinity to prothrombinase in a cleavage site-independent way. Despite equivalent exosite binding interactions between all four possible substrates and the enzyme, we propose that ordered bond cleavage results from the constraints associated with the binding of substrates in one of two conformations to a single form of prothrombinase.


Sujet(s)
Conformation des protéines , Prothrombine/composition chimique , Prothrombine/métabolisme , Thromboplastine/composition chimique , Thromboplastine/métabolisme , Séquence d'acides aminés , Arginine/métabolisme , Sites de fixation , Lignée cellulaire , Clonage moléculaire , Électrophorèse sur gel de polyacrylamide , Humains , Cinétique , Mutagenèse par insertion , Fragments peptidiques/composition chimique , Liaison aux protéines , Prothrombine/génétique , Protéines recombinantes/métabolisme , Relation structure-activité , Spécificité du substrat , Transfection
8.
J Biol Chem ; 277(48): 46191-6, 2002 Nov 29.
Article de Anglais | MEDLINE | ID: mdl-12370181

RÉSUMÉ

The specific action of serine proteinases on protein substrates is a hallmark of blood coagulation and numerous other physiological processes. Enzymic recognition of substrate sequences preceding the scissile bond is considered to contribute dominantly to specificity and function. We have investigated the contribution of active site docking by unique substrate residues preceding the scissile bond to the function of prothrombinase. Mutagenesis of the authentic P(1)-P(3) sequence in prethrombin 2/fragment 1.2 yielded substrate variants that could be converted to thrombin by prothrombinase. Proteolytic activation was also observed with a substrate variant containing the P(1)-P(3) sequence found in a coagulation zymogen not known to be activated by prothrombinase. Lower rates of activation of the variants derived from a decrease in maximum catalytic rate but not in substrate affinity. Replacement of the P(1) residue with Gln yielded an uncleavable derivative that retained the affinity of the wild type substrate for prothrombinase but did not engage the active site of the enzyme. Thus, active site docking of the substrate contributes to catalytic efficiency, but it is does not determine substrate affinity nor does it fully explain the specificity of prothrombinase. Therefore, extended interactions between prothrombinase and substrate regions removed from the cleavage site drive substrate affinity and enforce the substrate specificity of this enzyme complex.


Sujet(s)
Thromboplastine/métabolisme , Milieux de culture conditionnés , Électrophorèse sur gel de polyacrylamide , Humains , Cinétique , Mutagenèse , Prothrombine/métabolisme , Spécificité du substrat , Thromboplastine/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE