Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
BMC Evol Biol ; 19(1): 27, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30654737

RESUMO

BACKGROUND: The plasminogen (PLG) activation system is composed by a series of serine proteases, inhibitors and several binding proteins, which together control the temporal and spatial generation of the active serine protease plasmin. As this proteolytic system plays a central role in human physiology and pathophysiology it has been extensively studied in mammals. The serine proteases of this system are believed to originate from an ancestral gene by gene duplications followed by domain gains and deletions. However, the identification of ancestral forms in primitive chordates supporting these theories remains elusive. In addition, evolutionary studies of the non-proteolytic members of this system are scarce. RESULTS: Our phylogenetic analyses place lamprey PLG at the root of the vertebrate PLG-group, while lamprey PLG-related growth factors represent the ancestral forms of the jawed-vertebrate orthologues. Furthermore, we find that the earliest putative orthologue of the PLG activator group is the hyaluronan binding protein 2 (HABP2) gene found in lampreys. The prime plasminogen activators (tissue- and urokinase-type plasminogen activator, tPA and uPA) first occur in cartilaginous fish and phylogenetic analyses confirm that all orthologues identified compose monophyletic groups to their mammalian counterparts. Cartilaginous fishes exhibit the most ancient vitronectin of all vertebrates, while plasminogen activator inhibitor 1 (PAI-1) appears for the first time in cartilaginous fishes and is conserved in the rest of jawed vertebrate clades. PAI-2 appears for the first time in the common ancestor of reptiles and mammals, and represents the latest appearing plasminogen activator inhibitor. Finally, we noted that the urokinase-type plasminogen activator receptor (uPAR)-and three-LU domain containing genes in general-occurred later in evolution and was first detectable after coelacanths. CONCLUSIONS: This study identifies several primitive orthologues of the mammalian plasminogen activation system. These ancestral forms provide clues to the origin and diversification of this enzyme system. Further, the discovery of several members-hitherto unknown in mammals-provide new perspectives on the evolution of this important enzyme system.


Assuntos
Cordados/genética , Variação Genética , Filogenia , Plasminogênio/genética , Sequência de Aminoácidos , Animais , Bases de Dados de Proteínas , Humanos , Funções Verossimilhança , Inibidor 1 de Ativador de Plasminogênio/química , Domínios Proteicos , Análise de Sequência de RNA , Transcriptoma/genética , Ativador de Plasminogênio Tipo Uroquinase/química , Vitronectina/química
2.
Biochim Biophys Acta Gen Subj ; 1862(9): 2017-2023, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29959058

RESUMO

Some peptide sequences can behave as either substrates or inhibitors of serine proteases. Working with a cyclic peptidic inhibitor of the serine protease urokinase-type plasminogen activator (uPA), we have now demonstrated a new mechanism for an inhibitor-to-substrate switch. The peptide, CSWRGLENHAAC (upain-2), is a competitive inhibitor of human uPA, but is also slowly converted to a substrate in which the bond between Arg4 and Gly5 (the P1-P1' bond) is cleaved. Substituting the P2 residue Trp3 to an Ala or substituting the P1 Arg4 residue with 4-guanidino-phenylalanine strongly increased the substrate cleavage rate. We studied the structural basis for the inhibitor-to-substrate switch by determining the crystal structures of the various peptide variants in complex with the catalytic domain of uPA. While the slowly cleaved peptides bound clearly in inhibitory mode, with the oxyanion hole blocked by the side chain of the P3' residue Glu7, peptides behaving essentially as substrates with a much accelerated rate of cleavage was observed to be bound to the enzyme in substrate mode. Our analysis reveals that the inhibitor-to-substrate switch was associated with a 7 Štranslocation of the P2 residue, and we conclude that the inhibitor-to-substrate switch of upain-2 is a result of a major conformational change in the enzyme-bound state of the peptide. This conclusion is in contrast to findings with so-called standard mechanism inhibitors in which the inhibitor-to-substrate switch is linked to minor conformational changes in the backbone of the inhibitory peptide stretch.


Assuntos
Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sítios de Ligação , Domínio Catalítico , Cristalização , Cristalografia por Raios X , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Especificidade por Substrato
3.
EMBO Rep ; 17(7): 982-98, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27189837

RESUMO

Components of the plasminogen activation system including urokinase (uPA), its inhibitor (PAI-1) and its cell surface receptor (uPAR) have been implicated in a wide variety of biological processes related to tissue homoeostasis. Firstly, the binding of uPA to uPAR favours extracellular proteolysis by enhancing cell surface plasminogen activation. Secondly, it promotes cell adhesion and signalling through binding of the provisional matrix protein vitronectin. We now report that uPA and plasmin induces a potent negative feedback on cell adhesion through specific cleavage of the RGD motif in vitronectin. Cleavage of vitronectin by uPA displays a remarkable receptor dependence and requires concomitant binding of both uPA and vitronectin to uPAR Moreover, we show that PAI-1 counteracts the negative feedback and behaves as a proteolysis-triggered stabilizer of uPAR-mediated cell adhesion to vitronectin. These findings identify a novel and highly specific function for the plasminogen activation system in the regulation of cell adhesion to vitronectin. The cleavage of vitronectin by uPA and plasmin results in the release of N-terminal vitronectin fragments that can be detected in vivo, underscoring the potential physiological relevance of the process.


Assuntos
Motivos de Aminoácidos , Adesão Celular , Plasminogênio/metabolismo , Domínios e Motivos de Interação entre Proteínas , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Vitronectina/química , Vitronectina/metabolismo , Linhagem Celular Tumoral , Retroalimentação Fisiológica , Fibrinolisina/metabolismo , Fibronectinas/metabolismo , Expressão Gênica , Humanos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Proteólise , Ativador de Plasminogênio Tipo Uroquinase/genética
4.
J Biol Chem ; 291(27): 14340-14355, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27189939

RESUMO

Hepatocyte growth factor activator inhibitor-1 (HAI-1) is a type I transmembrane protein and inhibitor of several serine proteases, including hepatocyte growth factor activator and matriptase. The protein is essential for development as knock-out mice die in utero due to placental defects caused by misregulated extracellular proteolysis. HAI-1 contains two Kunitz-type inhibitor domains (Kunitz), which are generally thought of as a functionally self-contained protease inhibitor unit. This is not the case for HAI-1, where our results reveal how interdomain interactions have evolved to stimulate the inhibitory activity of an integrated Kunitz. Here we present an x-ray crystal structure of an HAI-1 fragment covering the internal domain and Kunitz-1. The structure reveals not only that the previously uncharacterized internal domain is a member of the polycystic kidney disease domain family but also how the two domains engage in interdomain interactions. Supported by solution small angle x-ray scattering and a combination of site-directed mutagenesis and functional assays, we show that interdomain interactions not only stabilize the fold of the internal domain but also stimulate the inhibitory activity of Kunitz-1. By completing our structural characterization of the previously unknown N-terminal region of HAI-1, we provide new insight into the interplay between tertiary structure and the inhibitory activity of a multidomain protease inhibitor. We propose a previously unseen mechanism by which the association of an auxiliary domain stimulates the inhibitory activity of a Kunitz-type inhibitor (i.e. the first structure of an intramolecular interaction between a Kunitz and another domain).


Assuntos
Glicoproteínas de Membrana/metabolismo , Doenças Renais Policísticas/metabolismo , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Glicoproteínas de Membrana/química , Camundongos , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Proteínas Secretadas Inibidoras de Proteinases , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espalhamento a Baixo Ângulo , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
5.
J Biol Chem ; 291(29): 15156-68, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226628

RESUMO

A peptide segment that binds the active site of a serine protease in a substrate-like manner may behave like an inhibitor or a substrate. However, there is sparse information on which factors determine the behavior a particular peptide segment will exhibit. Here, we describe the first x-ray crystal structure of a nanobody in complex with a serine protease. The nanobody displays a new type of interaction between an antibody and a serine protease as it inserts its complementary determining region-H3 loop into the active site of the protease in a substrate-like manner. The unique binding mechanism causes the nanobody to behave as a strong inhibitor as well as a poor substrate. Intriguingly, its substrate behavior is incomplete, as 30-40% of the nanobody remained intact and inhibitory after prolonged incubation with the protease. Biochemical analysis reveals that an intra-loop interaction network within the complementary determining region-H3 of the nanobody balances its inhibitor versus substrate behavior. Collectively, our results unveil molecular factors, which may be a general mechanism to determine the substrate versus inhibitor behavior of other protease inhibitors.


Assuntos
Camelídeos Americanos/imunologia , Serina Proteases/imunologia , Anticorpos de Domínio Único/química , Sequência de Aminoácidos , Animais , Domínio Catalítico , Regiões Determinantes de Complementaridade , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Conformação Proteica , Serina Proteases/química , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/metabolismo , Anticorpos de Domínio Único/metabolismo , Especificidade por Substrato , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/imunologia
6.
Biochim Biophys Acta ; 1860(3): 599-606, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26691138

RESUMO

BACKGROUND: Serine proteases are one of the most studied group of enzymes. Despite the extensive mechanistic studies, some crucial details remain controversial, for example, how the cleaved product is released in the catalysis reaction. A cyclic peptidyl inhibitor (CSWRGLENHRMC, upain-1) of a serine protease, urokinase-type plasminogen activator (uPA), was found to become a slow substrate and cleaved slowly upon the replacement of single residue (W3A). METHODS: By taking advantage of the unique property of this peptide, we report the high-resolution structures of uPA in complex with upain-1-W3A peptide at four different pH values by X-ray crystallography. RESULTS: In the structures obtained at low pH (pH4.6 and 5.5), the cyclic peptide upain-1-W3A was found to be intact and remained in the active site of uPA. At 7.4, the scissile bond of the peptide was found cleaved, showing that the peptide became a uPA substrate. At pH9.0, the C-terminal part of the substrate was no longer visible, and only the P1 residue occupying the S1 pocket was identified. CONCLUSIONS: The analysis of these structures provides explanations why the upain-1-W3A is a slow substrate. In addition, we clearly identified the cleaved fragments of the peptide at both sides of the scissile bond in the active site of the enzyme, showing a slow release of the cleaved peptide. GENERAL SIGNIFICANCE: This work indicates that the quick release of the cleaved P' fragment after the first step of hydrolysis may not always be needed for the second hydrolysis.


Assuntos
Peptídeos Cíclicos/química , Serina Proteases/química , Inibidores de Serina Proteinase/química , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/química , Concentração de Íons de Hidrogênio
7.
Nucleic Acids Res ; 43(21): e139, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26163061

RESUMO

Nucleic acid aptamer selection by systematic evolution of ligands by exponential enrichment (SELEX) has shown great promise for use in the development of research tools, therapeutics and diagnostics. Typically, aptamers are identified from libraries containing up to 10(16) different RNA or DNA sequences by 5-10 rounds of affinity selection towards a target of interest. Such library screenings can result in complex pools of many target-binding aptamers. New high-throughput sequencing techniques may potentially revolutionise aptamer selection by allowing quantitative assessment of the dynamic changes in the pool composition during the SELEX process and by facilitating large-scale post-SELEX characterisation. In the present study, we demonstrate how high-throughput sequencing of SELEX pools, before and after a single round of branched selection for binding to different target variants, can provide detailed information about aptamer binding sites, preferences for specific target conformations, and functional effects of the aptamers. The procedure was applied on a diverse pool of 2'-fluoropyrimidine-modified RNA enriched for aptamers specific for the serpin plasminogen activator inhibitor-1 (PAI-1) through five rounds of standard selection. The results demonstrate that it is possible to perform large-scale detailed characterisation of aptamer sequences directly in the complex pools obtained from library selection methods, thus without the need to produce individual aptamers.


Assuntos
Aptâmeros de Nucleotídeos/química , Sequenciamento de Nucleotídeos em Larga Escala , Técnica de Seleção de Aptâmeros , Sítios de Ligação , Ligantes , Mutação , Conformação de Ácido Nucleico , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , RNA/química , Alinhamento de Sequência , Análise de Sequência de RNA
8.
J Biol Chem ; 290(43): 25795-804, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26324706

RESUMO

Thrombosis is a leading cause of death worldwide. Recombinant tissue-type plasminogen activator (tPA) is the Food and Drug Administration-approved thrombolytic drug. tPA is rapidly inactivated by endogenous plasminogen activator inhibitor-1 (PAI-1). Engineering on tPA to reduce its inhibition by PAI-1 without compromising its thrombolytic effect is a continuous effort. Precise details, with atomic resolution, of the molecular interactions between tPA and PAI-1 remain unknown despite previous extensive studies. Here, we report the crystal structure of the tPA·PAI-1 Michaelis complex, which shows significant differences from the structure of its urokinase-type plasminogen activator analogue, the uPA·PAI-1 Michaelis complex. The PAI-1 reactive center loop adopts a unique kinked conformation. The structure provides detailed interactions between tPA 37- and 60-loops with PAI-1. On the tPA side, the S2 and S1ß pockets open up to accommodate PAI-1. This study provides structural basis to understand the specificity of PAI-1 and to design newer generation of thrombolytic agents with reduced PAI-1 inactivation.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/química , Ativador de Plasminogênio Tecidual/química , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Estrutura Molecular , Proteínas Recombinantes/química
9.
Bioconjug Chem ; 27(4): 918-26, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-26926041

RESUMO

In drug development, molecular intervention strategies are usually based on interference with a single protein function, such as enzyme activity or receptor binding. However, in many cases, protein drug targets are multifunctional, with several molecular functions contributing to their pathophysiological actions. Aptamers and peptides are interesting synthetic building blocks for the design of multivalent molecules capable of modulating multiple functions of a target protein. Here, we report a molecular trap with the ability to interfere with the activation, catalytic activity, receptor binding, etc. of the serine protease urokinase-type plasminogen activator (uPA) by a rational combination of two RNA aptamers and a peptide with different inhibitory properties. The assembly of these artificial inhibitors into one molecule enhanced the inhibitory activity between 10- and 10,000-fold toward several functions of uPA. The study highlights the potential of multivalent designs and illustrates how they can easily be constructed from aptamers and peptides using nucleic acid engineering, chemical synthesis, and bioconjugation chemistry. By aptamer to aptamer and aptamer to peptide conjugation, we created, to the best of our knowledge, the first trivalent molecule which combines three artificial inhibitors binding to three different sites in a protein target. We hypothesize that by simultaneously preventing all of the functional interactions and activities of the target protein, this approach may represent an alternative to siRNA technology for a functional knockout.


Assuntos
Aptâmeros de Nucleotídeos/química , Peptídeos/química , Serina Proteases/química , Sequência de Aminoácidos
10.
Biochem J ; 466(2): 299-309, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25510835

RESUMO

A decade ago, motif at N-terminus with eight-cysteines (MANEC) was defined as a new protein domain family. This domain is found exclusively at the N-terminus of >400 multi-domain type-1 transmembrane proteins from animals. Despite the large number of MANEC-containing proteins, only one has been characterized at the protein level: hepatocyte growth factor activator inhibitor-1 (HAI-1). HAI-1 is an essential protein, as knockout mice die in utero due to placental defects. HAI-1 is an inhibitor of matriptase, hepsin and hepatocyte growth factor (HGF) activator, all serine proteases with important roles in epithelial development, cell growth and homoeostasis. Dysregulation of these proteases has been causatively implicated in pathological conditions such as skin diseases and cancer. Detailed functional understanding of HAI-1 and other MANEC-containing proteins is hampered by the lack of structural information on MANEC. Although many MANEC sequences exist, sequence-based database searches fail to predict structural homology. In the present paper, we present the NMR solution structure of the MANEC domain from HAI-1, the first three-dimensional (3D) structure from the MANEC domain family. Unexpectedly, MANEC is a new subclass of the PAN/apple domain family, with its own unifying features, such as two additional disulfide bonds, two extended loop regions and additional α-helical elements. As shown for other PAN/apple domain-containing proteins, we propose a similar active role of the MANEC domain in intramolecular and intermolecular interactions. The structure provides a tool for the further elucidation of HAI-1 function as well as a reference for the study of other MANEC-containing proteins.


Assuntos
Modelos Moleculares , Proteínas Secretadas Inibidoras de Proteinases/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Humanos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/classificação , Proteínas Mutantes/metabolismo , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/química , Sinais Direcionadores de Proteínas , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Secretadas Inibidoras de Proteinases/classificação , Proteínas Secretadas Inibidoras de Proteinases/genética , Proteínas Secretadas Inibidoras de Proteinases/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/classificação , Proteínas Recombinantes/metabolismo , Espalhamento a Baixo Ângulo , Solubilidade , Difração de Raios X
11.
Biochem J ; 449(1): 161-6, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23016918

RESUMO

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.


Assuntos
Anticorpos Monoclonais/farmacologia , Precursores Enzimáticos/antagonistas & inibidores , Precursores Enzimáticos/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Anticorpos Monoclonais/química , Linhagem Celular Tumoral , Cristalografia por Raios X , Indução Enzimática/fisiologia , Precursores Enzimáticos/química , Estabilidade Enzimática/fisiologia , Humanos , Proteólise , Ativador de Plasminogênio Tipo Uroquinase/química
12.
Angew Chem Int Ed Engl ; 53(37): 9751-4, 2014 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-25049220

RESUMO

The metastability of the native fold makes serpin (serine protease inhibitor) proteins prone to pathological conformational change, often by insertion of an extra ß-strand into the central ß-sheet A. How this insertion is made possible is a hitherto unresolved question. By the use of advanced hydrogen/deuterium-exchange mass spectrometry (HDX-MS) it is shown that the serpin plasminogen activator inhibitor 1 (PAI-1) transiently unfolds under native condition, on a second-to-minute time scale. The unfolding regions comprise ß-strand 5A as well as the underlying hydrophobic core, including ß-strand 6B and parts of helices A, B, and C. Based thereon, a mechanism is proposed by which PAI-1 makes transitions through progressively more unfolded states along the reaction coordinate to the inactive, so-called latent form. Our results highlight the profound utility of HDX-MS in detecting sparsely populated, transiently unfolded protein states.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/metabolismo , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/genética , Conformação Proteica , Serpinas
13.
Biochemistry ; 52(40): 7114-26, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24079451

RESUMO

Serine protease catalytic activity is in many cases regulated by conformational changes initiated by binding of physiological modulators to exosites located distantly from the active site. Inhibitory monoclonal antibodies binding to such exosites are potential therapeutics and offer opportunities for elucidating fundamental allosteric mechanisms. The monoclonal antibody mU1 has previously been shown to be able to inhibit the function of murine urokinase-type plasminogen activator in vivo. We have now mapped the epitope of mU1 to the catalytic domain's 37- and 70-loops, situated about 20 Å from the S1 specificity pocket of the active site. Our data suggest that binding of mU1 destabilizes the catalytic domain and results in conformational transition into a state, in which the N-terminal amino group of Ile16 is less efficiently stabilizing the oxyanion hole and in which the active site has a reduced affinity for substrates and inhibitors. Furthermore, we found evidence for functional interactions between residues in uPA's C-terminal catalytic domain and its N-terminal A-chain, as deletion of the A-chain facilitates the mU1-induced conformational distortion. The inactive, distorted state is by several criteria similar to the E* conformation described for other serine proteases. Hence, agents targeting serine protease conformation through binding to exosites in the 37- and 70-loops represent a new class of potential therapeutics.


Assuntos
Serina Endopeptidases/imunologia , Inibidores de Serina Proteinase/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/imunologia , Regulação Alostérica , Animais , Anticorpos Monoclonais , Domínio Catalítico , Ativação Enzimática/efeitos dos fármacos , Epitopos/metabolismo , Fibrinolisina/metabolismo , Cinética , Camundongos , Modelos Moleculares , Conformação Proteica/efeitos dos fármacos , Ressonância de Plasmônio de Superfície , Ativador de Plasminogênio Tipo Uroquinase/efeitos dos fármacos
14.
J Biol Chem ; 287(12): 8994-9001, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22275370

RESUMO

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.


Assuntos
Anticorpos Monoclonais/metabolismo , Fator VIIa/química , Fator VIIa/metabolismo , Regulação Alostérica , Animais , Anticorpos Monoclonais/química , Sítios de Ligação , Fator VIIa/genética , Humanos , Cinética , Camundongos , Ligação Proteica , Tromboplastina/genética , Tromboplastina/metabolismo
15.
J Biol Chem ; 287(33): 27526-36, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22733817

RESUMO

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.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/metabolismo , Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Sequência de Bases , Clonagem Molecular , Dados de Sequência Molecular , Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Estrutura Terciária de Proteína , Ativador de Plasminogênio Tipo Uroquinase/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
16.
Chembiochem ; 14(16): 2179-88, 2013 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-24115455

RESUMO

The development of protease inhibitors for pharmacological intervention has taken a new turn with the use of peptide-based inhibitors. Here, we report the rational design of bicyclic peptide inhibitors of the serine protease urokinase-type plasminogen activator (uPA), based on the established monocyclic peptide, upain-2. It was successfully converted to a bicyclic peptide, without loss of inhibitory properties. The aim was to produce a peptide cyclised by an amide bond with an additional stabilising across-the-ring covalent bond. We expected this bicyclic peptide to exhibit a lower entropic burden upon binding. Two bicyclic peptides were synthesised with affinities similar to that of upain-2, and their binding energetics were evaluated by isothermal titration calorimetry. Indeed, compared to upain-2, the bicyclic peptides showed reduced loss of entropy upon binding to uPA. We also investigated the solution structures of the bicyclic peptide by NMR spectroscopy to map possible conformations. An X-ray structure of the bicyclic-peptide-uPA complex confirmed an interaction similar to that for the previous upain-1/upain-2-uPA complexes. These physical studies of the peptide-protease interactions will aid future designs of bicyclic peptide protease inhibitors.


Assuntos
Peptídeos Cíclicos/metabolismo , Inibidores de Proteases/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Cinética , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores
17.
Biochemistry ; 51(39): 7804-11, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22950516

RESUMO

The catalytic activity of serine proteases depends on a salt-bridge between the amino group of residue 16 and the side chain of Asp194. The salt-bridge stabilizes the oxyanion hole and the S1 specificity pocket of the protease. Some serine proteases exist in only partially active forms, in which the amino group of residue 16 is exposed to the solvent. Such a partially active state is assumed by a truncated form of the murine urokinase-type plasminogen activator (muPA), consisting of residues 16-243. Here we investigated the allosteric interconversion between partially active states and the fully active state. Both a monoclonal antibody (mU3) and a peptidic inhibitor (mupain-1--16) stabilize the active state. The epitope of mU3 is located in the 37- and 70-loops at a site homologous to exosite I of thrombin. The N-terminus((Ile16)) of muPA((16--243)) was less exposed upon binding of mU3 or mupain-1--16. In contrast, introduction of the mutations F40Y or E137A into muPA((16--243)) increased exposure of the N-terminus((Ile16)) and resulted in large changes in the thermodynamic parameters for mupain-1--16 binding. We conclude that the distorted state of muPA((16--243)) is conformationally ordered upon binding of ligands to the active site and upon binding of mU3 to the 37- and 70-loops. Our study establishes the 37- and 70-loops as a unique site for binding to compounds stabilizing the active state of serine proteases.


Assuntos
Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Regulação Alostérica , Animais , Domínio Catalítico , Ativação Enzimática , Células HEK293 , Humanos , Camundongos , Modelos Moleculares , Peptídeos Cíclicos/metabolismo , Mutação Puntual , Conformação Proteica , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/genética
18.
Biochemistry ; 51(41): 8256-66, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-22957734

RESUMO

The native fold of plasminogen activator inhibitor 1 (PAI-1) represents an active metastable conformation that spontaneously converts to an inactive latent form. Binding of the somatomedin B domain (SMB) of the endogenous cofactor vitronectin to PAI-1 delays the transition to the latent state and increases the thermal stability of the protein dramatically. We have used hydrogen/deuterium exchange mass spectrometry to assess the inherent structural flexibility of PAI-1 and to monitor the changes induced by SMB binding. Our data show that the PAI-1 core consisting of ß-sheet B is rather protected against exchange with the solvent, while the remainder of the molecule is more dynamic. SMB binding causes a pronounced and widespread stabilization of PAI-1 that is not confined to the binding interface with SMB. We further explored the local structural flexibility in a mutationally stabilized PAI-1 variant (14-1B) as well as the effect of stabilizing antibody Mab-1 on wild-type PAI-1. The three modes of stabilizing PAI-1 (SMB, Mab-1, and the mutations in 14-1B) all cause a delayed latency transition, and this effect was accompanied by unique signatures on the flexibility of PAI-1. Reduced flexibility in the region around helices B, C, and I was seen in all three cases, which suggests an involvement of this region in mediating structural flexibility necessary for the latency transition. These data therefore add considerable depth to our current understanding of the local structural flexibility in PAI-1 and provide novel indications of regions that may affect the functional stability of PAI-1.


Assuntos
Espectrometria de Massas/métodos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Somatomedinas/metabolismo , Vitronectina/metabolismo , Deutério , Hidrogênio , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/química , Somatomedinas/química , Vitronectina/química
19.
J Biol Chem ; 286(9): 7027-32, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21199867

RESUMO

Plasminogen activator inhibitor-1 (PAI-1), together with its physiological target urokinase-type plasminogen activator (uPA), plays a pivotal role in fibrinolysis, cell migration, and tissue remodeling and is currently recognized as being among the most extensively validated biological prognostic factors in several cancer types. PAI-1 specifically and rapidly inhibits uPA and tissue-type PA (tPA). Despite extensive structural/functional studies on these two reactions, the underlying structural mechanism has remained unknown due to the technical difficulties of obtaining the relevant structures. Here, we report a strategy to generate a PAI-1·uPA(S195A) Michaelis complex and present its crystal structure at 2.3-Å resolution. In this structure, the PAI-1 reactive center loop serves as a bait to attract uPA onto the top of the PAI-1 molecule. The P4-P3' residues of the reactive center loop interact extensively with the uPA catalytic site, accounting for about two-thirds of the total contact area. Besides the active site, almost all uPA exosite loops, including the 37-, 60-, 97-, 147-, and 217-loops, are involved in the interaction with PAI-1. The uPA 37-loop makes an extensive interaction with PAI-1 ß-sheet B, and the 147-loop directly contacts PAI-1 ß-sheet C. Both loops are important for initial Michaelis complex formation. This study lays down a foundation for understanding the specificity of PAI-1 for uPA and tPA and provides a structural basis for further functional studies.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/química , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Domínio Catalítico , Cristalografia , Ativação Enzimática/fisiologia , Humanos , Mutação , Pichia/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Ativador de Plasminogênio Tipo Uroquinase/genética
20.
J Biol Chem ; 286(34): 29709-17, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21697084

RESUMO

The serpin plasminogen activator inhibitor-1 (PAI-1) is a crucial regulator in fibrinolysis and tissue remodeling. PAI-1 has been associated with several pathological conditions and is a validated prognostic marker in human cancers. However, structural information about the native inhibitory form of PAI-1 has been elusive because of its inherent conformational instability and rapid conversion to a latent, inactive structure. Here we report the crystal structure of PAI-1 W175F at 2.3 Å resolution as the first model of the metastable native molecule. Structural comparison with a quadruple mutant (14-1B) previously used as representative of the active state uncovered key differences. The most striking differences occur near the region that houses three of the four mutations in the 14-1B PAI-1 structure. Prominent changes are localized within a loop connecting ß-strand 3A with the F helix, in which a previously observed 3(10)-helix is absent in the new structure. Notably these structural changes are found near the binding site for the cofactor vitronectin. Because vitronectin is the only known physiological regulator of PAI-1 that slows down the latency conversion, the structure of this region is important. Furthermore, the previously identified chloride-binding site close to the F-helix is absent from the present structure and likely to be artifactual, because of its dependence on the 14-1B mutations. Instead we found a different chlorine-binding site that is likely to be present in wild type PAI-1 and that more satisfactorily accounts for the chlorine stabilizing effect on PAI-1.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/química , Cristalografia por Raios X , Humanos , Mutação de Sentido Incorreto , Inibidor 1 de Ativador de Plasminogênio/genética , Estabilidade Proteica , Estrutura Secundária de Proteína , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA