Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 138(14): 1258-1268, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34077951

ABSTRACT

Hemophilia A is a bleeding disorder resulting from deficient factor VIII (FVIII), which normally functions as a cofactor to activated factor IX (FIXa) that facilitates activation of factor X (FX). To mimic this property in a bispecific antibody format, a screening was conducted to identify functional pairs of anti-FIXa and anti-FX antibodies, followed by optimization of functional and biophysical properties. The resulting bispecific antibody (Mim8) assembled efficiently with FIXa and FX on membranes, and supported activation with an apparent equilibrium dissociation constant of 16 nM. Binding affinity with FIXa and FX in solution was much lower, with equilibrium dissociation constant values for FIXa and FX of 2.3 and 1.5 µM, respectively. In addition, the activity of Mim8 was dependent on stimulatory activity contributed by the anti-FIXa arm, which enhanced the proteolytic activity of FIXa by 4 orders of magnitude. In hemophilia A plasma and whole blood, Mim8 normalized thrombin generation and clot formation, with potencies 13 and 18 times higher than a sequence-identical analogue of emicizumab. A similar potency difference was observed in a tail vein transection model in hemophilia A mice, whereas reduction of bleeding in a severe tail-clip model was observed only for Mim8. Furthermore, the pharmacokinetic parameters of Mim8 were investigated and a half-life of 14 days shown in cynomolgus monkeys. In conclusion, Mim8 is an activated FVIII mimetic with a potent and efficacious hemostatic effect based on preclinical data.


Subject(s)
Antibodies, Bispecific/therapeutic use , Hemophilia A/drug therapy , Hemorrhage/drug therapy , Animals , Factor IXa/antagonists & inhibitors , Factor VIIIa/therapeutic use , Factor X/antagonists & inhibitors , Female , Humans , Male , Mice, Inbred C57BL
2.
Cell Chem Biol ; 23(6): 700-8, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27265748

ABSTRACT

Most serpins are fast and specific inhibitors of extracellular serine proteases controlling biological processes such as blood coagulation, fibrinolysis, tissue remodeling, and inflammation. The inhibitory activity of serpins is based on a conserved metastable structure and their conversion to a more stable state during reaction with the target protease. However, the metastable state also makes serpins vulnerable to mutations, resulting in disease caused by inactive and misfolded monomeric or polymeric forms ("serpinopathy"). Misfolding can occur either intracellularly (type-I serpinopathies) or extracellularly (type-II serpinopathies). We have isolated a 2'-fluoropyrimidine-modified RNA aptamer, which inhibits a mutation-induced inactivating misfolding of the serpin α1-antichymotrypsin. It is the first agent able to stabilize a type-II mutation of a serpin without interfering with the inhibitory mechanism, thereby presenting a solution for the long-standing challenge of preventing pathogenic misfolding without compromising the inhibitory function.


Subject(s)
Aptamers, Nucleotide/pharmacology , Mutation , Protein Folding/drug effects , Serpins/genetics , Serpins/metabolism , Aptamers, Nucleotide/chemistry , Deuterium Exchange Measurement , Humans , Mass Spectrometry , Models, Molecular , Serpins/chemistry , Surface Plasmon Resonance
3.
Thromb Haemost ; 111(1): 29-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24085288

ABSTRACT

Protease nexin-1 (PN-1) belongs to the serpin family and is an inhibitor of thrombin, plasmin, urokinase-type plasminogen activator, and matriptase. Recent studies have suggested PN-1 to play important roles in vascular-, neuro-, and tumour-biology. The serpin inhibitory mechanism consists of the serpin presenting its so-called reactive centre loop as a substrate to its target protease, resulting in a covalent complex with the inactivated enzyme. Previously, three mechanisms have been proposed for the inactivation of serpins by monoclonal antibodies: steric blockage of protease recognition, conversion to an inactive conformation or induction of serpin substrate behaviour. Until now, no inhibitory antibodies against PN-1 have been thoroughly characterised. Here we report the development of three monoclonal antibodies binding specifically and with high affinity to human PN-1. The antibodies all abolish the protease inhibitory activity of PN-1. In the presence of the antibodies, PN-1 does not form a complex with its target proteases, but is recovered in a reactive centre cleaved form. Using site-directed mutagenesis, we mapped the three overlapping epitopes to an area spanning the gap between the loop connecting α-helix F with ß-strand 3A and the loop connecting α-helix A with ß-strand 1B. We conclude that antibody binding causes a direct blockage of the final critical step of protease translocation, resulting in abortive inhibition and premature release of reactive centre cleaved PN-1. These new antibodies will provide a powerful tool to study the in vivo role of PN-1's protease inhibitory activity.


Subject(s)
Antibodies, Monoclonal/chemistry , Serpin E2/antagonists & inhibitors , Serpin E2/chemistry , Antibody Affinity , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Epitope Mapping , Escherichia coli/metabolism , Glycosylation , Heparin/chemistry , Humans , Kinetics , Mutagenesis, Site-Directed , Neoplasms/metabolism , Peptide Hydrolases/chemistry , Protein Conformation , Protein Transport , Serpin E2/genetics , Surface Plasmon Resonance , Time Factors
4.
Biochem J ; 449(1): 161-6, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23016918

ABSTRACT

An important regulatory mechanism of serine proteases is the proteolytic conversion of the inactive pro-enzyme, or zymogen, into the active enzyme. This activation process is generally considered an irreversible process. In the present study, we demonstrate that an active enzyme can be converted back into its zymogen form. We determined the crystal structure of uPA (urokinase-type plasminogen activator) in complex with an inhibitory antibody, revealing that the antibody 'rezymogenizes' already activated uPA. The present study demonstrates a new regulatory mechanism of protease activity, which is also an extreme case of protein allostery. Mechanistically, the antibody binds a single surface-exposed loop, named the autolysis loop, thereby preventing the stabilization of uPA in its active conformation. We argue that this autolysis loop is a key structural element for rezymogenation of other proteases, and will be a new target site for pharmacological intervention with serine protease activity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Enzyme Precursors/antagonists & inhibitors , Enzyme Precursors/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Enzyme Induction/physiology , Enzyme Precursors/chemistry , Enzyme Stability/physiology , Humans , Proteolysis , Urokinase-Type Plasminogen Activator/chemistry
5.
J Biol Chem ; 287(33): 27526-36, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22733817

ABSTRACT

Plasminogen activation catalyzed by urokinase-type plasminogen activator (uPA) plays an important role in normal and pathological tissue remodeling processes. Since its discovery in the mid-1980s, the cell membrane-anchored urokinase-type plasminogen activator receptor (uPAR) has been believed to be central to the functions of uPA, as uPA-catalyzed plasminogen activation activity appeared to be confined to cell surfaces through the binding of uPA to uPAR. However, a functional uPAR has so far only been identified in mammals. We have now cloned, recombinantly produced, and characterized two zebrafish proteases, zfuPA-a and zfuPA-b, which by several criteria are the fish orthologs of mammalian uPA. Thus, both proteases catalyze the activation of fish plasminogen efficiently and both proteases are inhibited rapidly by plasminogen activator inhibitor-1 (PAI-1). But zfuPA-a differs from mammalian uPA by lacking the exon encoding the uPAR-binding epidermal growth factor-like domain; zfuPA-b differs from mammalian uPA by lacking two cysteines of the epidermal growth factor-like domain and a uPAR-binding sequence comparable with that found in mammalian uPA. Accordingly, no zfuPA-b binding activity could be found in fish white blood cells or fish cell lines. We therefore propose that the current consensus of uPA-catalyzed plasminogen activation taking place on cell surfaces, derived from observations with mammals, is too narrow. Fish uPAs appear incapable of receptor binding in the manner known from mammals and uPA-catalyzed plasminogen activation in fish may occur mainly in solution. Studies with nonmammalian vertebrate species are needed to obtain a comprehensive understanding of the mechanism of plasminogen activation.


Subject(s)
Plasminogen Activator Inhibitor 1/metabolism , Plasminogen/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Plasminogen/genetics , Plasminogen Activator Inhibitor 1/genetics , Protein Structure, Tertiary , Urokinase-Type Plasminogen Activator/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
J Biol Chem ; 287(12): 8994-9001, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22275370

ABSTRACT

In the absence of its cofactor tissue factor (TF), coagulation factor VIIa (FVIIa) predominantly exists in a zymogen-like, catalytically incompetent state. Here we demonstrate that conformation-specific monoclonal antibodies (mAbs) can be used to characterize structural features determining the activity of FVIIa. We isolated two classes of mAbs, which both increased the catalytic efficiency of FVIIa more than 150-fold. The effects of the antibodies were retained with a FVIIa variant, which has been shown to be inert to allosteric activation by the natural activator TF, suggesting that the antibodies and TF employ distinct mechanisms of activation. The antibodies could be classified into two groups based on their patterns of affinities for different conformations of FVIIa. Whereas one class of antibodies affected both the K(m) and k(cat), the other class mainly affected the K(m). The antibody-induced activity enhancement could be traced to maturation of the S1 substrate binding pocket and the oxyanion hole, evident by an increased affinity for p-aminobenzamidine, an increased rate of antithrombin inhibition, an increased rate of incorporation of diisopropylfluorophosphate, and an enhanced fraction of molecules with a buried N terminus of the catalytic domain in the presence of antibodies. As demonstrated by site-directed mutagenesis, the two groups of antibodies appear to have overlapping, although clearly different, epitopes in the 170-loop. Our findings suggest that binding of ligands to specific residues in the 170-loop or its spatial vicinity may stabilize the S1 pocket and the oxyanion hole, and they may have general implications for the molecular understanding of FVIIa regulatory mechanisms.


Subject(s)
Antibodies, Monoclonal/metabolism , Factor VIIa/chemistry , Factor VIIa/metabolism , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Factor VIIa/genetics , Humans , Kinetics , Mice , Protein Binding , Thromboplastin/genetics , Thromboplastin/metabolism
7.
Mol Pharmacol ; 80(4): 585-97, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21719463

ABSTRACT

There is increasing interest in developing peptides for pharmacological intervention with pathophysiological functions of serine proteases. From phage-displayed peptide libraries, we previously isolated peptidylic inhibitors of urokinase-type plasminogen activator, a potential target for intervention with cancer invasion. The two peptides, upain-1 (CSWRGLENHRMC) and mupain-1 (CPAYSRYLDC), are competitive inhibitors of human and murine urokinase-type plasminogen activator, respectively. Both have an Arg as the P1 residue, inserting into the S1 pocket in the active site of the enzymes, but their specificity depends to a large extent on interactions outside the enzymes' active sites, so-called exosite interactions. Here we describe upain-2 (CSWRGLENHAAC) and the synthesis of a number of upain-2 and mupain-1 variants in which the P1 Arg was substituted with novel non-natural Arg analogs and achieved considerable improvement in the affinity of the peptides to their targets. Using chimeras of human and murine urokinase-type plasminogen activator as well as X-ray crystallography, we delineated the relative contribution of the P1 residue and exosite interactions to the affinity and specificity of the inhibitors for their target enzyme. The effect of inserting a particular non-natural amino acid into the P1 position is determined by the fact that changes in interactions of the P1 residue in the S1 pocket lead to changed exosite interactions and vice versa. These findings are of general interest when the affinities and specificities of serine protease inhibitors to be used for pharmacological intervention are considered and could pave the way for potential drug candidates for the treatment of cancer.


Subject(s)
Arginine/metabolism , Peptides, Cyclic/metabolism , Serine Proteinase Inhibitors/metabolism , Amino Acid Sequence , Animals , Arginine/chemistry , Arginine/genetics , Catalytic Domain/physiology , Crystallography, X-Ray , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/genetics , Protein Binding/physiology , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/genetics
8.
Biochemistry ; 48(40): 9606-17, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19705874

ABSTRACT

Serine proteases are initially synthesized as single-chain proenzymes with activities that are many orders of magnitude lower than those of the mature enzyme. Proteolytic cleavage of an exposed loop liberates a new amino terminus that inserts into a hydrophobic pocket and forms a stabilizing salt bridge with a ubiquitously conserved aspartate residue, resulting in a conformational change organizing the mature oxyanion hole. In a decisive 1976 work, Huber and Bode [Bode, W., and Huber, R. (1976) FEBS Lett. 68, 231-236] demonstrated that peptides sequentially similar to the new amino terminus in combination with a catalytic site inhibitor could specifically induce a trypsin-like conformation in trypsinogen. We now demonstrate that an Ile-Ile or Ile-Val dipeptide can induce limited enzyme activity in the single-chain zymogen form of urokinase-type plasminogen activator (uPA) or its K158A variant, which cannot be activated proteolytically. Furthermore, the slow formation of a covalent serpin-protease complex between single-chain uPA and PAI-1 is significantly accelerated in the presence of specific dipeptide sequences. The technique of using a dipeptide mimic as a surrogate for the liberated amino terminus further provides a novel means by which to covalently label the immature active site of single-chain uPA with a fluorescent probe, permitting fluorescence approaches for direct observations of conformational changes within the protease domain during zymogen activation. These data demonstrate the structural plasticity of the protease domain, reinforce the notion of "molecular sexuality", and provide a novel way of studying conformational changes of zymogens during proteolytic activation.


Subject(s)
Dipeptides/physiology , Peptide Hydrolases , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/chemistry , Animals , Catalysis , Cattle , Chromogenic Compounds/chemistry , Chromogenic Compounds/metabolism , Dipeptides/chemistry , Dipeptides/metabolism , Enzyme Induction , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Humans , Peptide Hydrolases/chemistry , Peptide Hydrolases/physiology , Substrate Specificity
9.
Biochem J ; 412(3): 447-57, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18318660

ABSTRACT

uPA (urokinase-type plasminogen activator) is a potential therapeutic target in a variety of pathological conditions, including cancer. In order to find new principles for inhibiting uPA in murine cancer models, we screened a phage-displayed peptide library with murine uPA as bait. We thereby isolated several murine uPA-binding peptide sequences, the predominant of which was the disulfide-bridged constrained sequence CPAYSRYLDC, which we will refer to as mupain-1. A chemically synthesized peptide corresponding to this sequence was found to be a competitive inhibitor of murine uPA, inhibiting its activity towards a low-molecular-mass chromogenic substrate as well as towards its natural substrate plasminogen. The K(i) value for inhibition as well as the K(D) value for binding were approx. 400 nM. Among a variety of other murine and human serine proteases, including trypsin, mupain-1 was found to be highly selective for murine uPA and did not even measurably inhibit human uPA. The cyclic structure of mupain-1 was indispensable for binding. Alanine scanning mutagenesis identified Arg(6) of mupain-1 as the P1 residue and indicated an extended binding interaction including the P5, P3, P2, P1 and P1' residues of mupain-1 and the specificity pocket, the catalytic triad and amino acids 41, 99 and 192 located in and around the active site of murine uPA. Exchanging His(99) of human uPA by a tyrosine residue, the corresponding residue in murine uPA, conferred mupain-1 susceptibility on to the latter. Peptide-derived inhibitors, such as mupain-1, may provide novel mechanistic information about enzyme-inhibitor interactions, provide alternative methodologies for designing effective protease inhibitors, and be used for target validation in murine model systems.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Amino Acid Substitution , Animals , Humans , Mice , Species Specificity , Urokinase-Type Plasminogen Activator/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL