Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Med Microbiol ; 306(6): 429-42, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27267179

RESUMEN

Clostridium perfringens is a Gram-positive, anaerobic, spore-forming bacterium that causes human gas gangrene (clostridial myonecrosis) and food poisoning. Early studies showed that virulence was regulated by the VirSR two-component signal transduction system. However, our identification of the RevR orphan response regulator indicated that more than one system was involved in controlling virulence. To further characterize this virulence-associated regulator, gel mobility shift experiments, coupled with DNase I footprinting, were used to identify the RevR DNA binding sequence. Bioinformatics analysis suggested that an orphan sensor histidine kinase, CPE1757 (renamed RevS), was the cognate sensor of RevR. Interaction between RevS and RevR was demonstrated by use of a bacterial two-hybrid system and validated by protein-protein interaction studies using biolayer interferometry. To assess the involvement of RevS in virulence regulation, the revS gene was inactivated by Targetron insertion. When isogenic wild-type, revS and complemented revS strains were tested in a mouse myonecrosis model, the revS mutant was found to be attenuated in virulence, which was similar to the attenuation observed previously with the revR mutant. However, transcriptional analysis of selected RevR-regulated genes in the revS mutant revealed a different pattern of expression to a revR mutant, suggesting that the RevSR system is more complex than originally thought. Taken together, the results have led to the identification and characterization of the two essential parts of a new regulatory network that is involved in the regulation of virulence in C. perfringens.


Asunto(s)
Clostridium perfringens/fisiología , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Clostridium perfringens/genética , Huella de ADN , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Histidina Quinasa/genética , Ratones Endogámicos BALB C , Mutagénesis Insercional , Unión Proteica , Mapeo de Interacción de Proteínas , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos , Virulencia
2.
Vet Microbiol ; 169(3-4): 163-70, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24467929

RESUMEN

Leptospira borgpetersenii serovar Hardjo subtype Hardjobovis (Hardjobovis) is the main causative agent of bovine leptospirosis in Australia, New Zealand, North America and elsewhere. Bovine leptospirosis can result in spontaneous abortion, stillbirth and reduced milk output. The organism is shed in the urine of infected animals and contact with contaminated materials can result in zoonotic infections in humans. Protective immunity in cattle against Hardjobovis involves stimulation of a Th1 cell mediated immune response, which can be characterized by the production of IFN-γ when blood from vaccinated animals is exposed to Hardjobovis antigens. However, the leptospiral components involved in stimulating this response have yet to be identified. In this study, 238 recombinant leptospiral proteins were evaluated for their ability to stimulate IFN-γ production in blood of cattle vaccinated with a commercial monovalent Hardjobovis vaccine. The conserved lipoprotein LipL32 is the major outer membrane protein of pathogenic Leptospira spp. A pool of soluble recombinant proteins which included LipL32, as well as LipL32 alone, stimulated significant IFN-γ production in blood of vaccinated cattle. A number of recombinant LipL32 fragments was generated, which identified the amino acids between 20 and 200 as containing the bovine T-cell reactive regions of LipL32. However, whether LipL32 plays a role in stimulating protective immunity in mammals has yet to be conclusively determined.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Enfermedades de los Bovinos/inmunología , Interferón gamma/metabolismo , Leptospira/inmunología , Leptospirosis/veterinaria , Lipoproteínas/inmunología , Proteínas Recombinantes/inmunología , Aminoácidos/inmunología , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Bovinos , Femenino , Inmunidad Celular/inmunología , Interferón gamma/sangre , Leptospirosis/inmunología , Lipoproteínas/genética , Proteínas Recombinantes/genética
3.
mBio ; 4(1): e00019-13, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23386432

RESUMEN

Clostridium perfringens is an anaerobic bacterium that causes numerous important human and animal diseases, primarily as a result of its ability to produce many different protein toxins. In chickens, C. perfringens causes necrotic enteritis, a disease of economic importance to the worldwide poultry industry. The secreted pore-forming toxin NetB is a key virulence factor in the pathogenesis of avian necrotic enteritis and is similar to alpha-hemolysin, a ß-barrel pore-forming toxin from Staphylococcus aureus. To address the molecular mechanisms underlying NetB-mediated tissue damage, we determined the crystal structure of the monomeric form of NetB to 1.8 Å. Structural comparisons with other members of the alpha-hemolysin family revealed significant differences in the conformation of the membrane binding domain. These data suggested that NetB may recognize different membrane receptors or use a different mechanism for membrane-protein interactions. Consistent with this idea, electrophysiological experiments with planar lipid bilayers revealed that NetB formed pores with much larger single-channel conductance than alpha-hemolysin. Channel conductance varied with phospholipid net charge. Furthermore, NetB differed in its ion selectivity, preferring cations over anions. Using hemolysis as a screen, we carried out a random-mutagenesis study that identified several residues that are critical for NetB-induced cell lysis. Mapping of these residues onto the crystal structure revealed that they were clustered in regions predicted to be required for oligomerization or membrane binding. Together these data provide an insight into the mechanism of NetB-mediated pore formation and will contribute to our understanding of the mode of action of this important toxin. IMPORTANCE Necrotic enteritis is an economically important disease of the worldwide poultry industry and is mediated by Clostridium perfringens strains that produce NetB, a ß-pore-forming toxin. We carried out structural and functional studies of NetB to provide a mechanistic insight into its mode of action and to assist in the development of a necrotic enteritis vaccine. We determined the structure of the monomeric form of NetB to 1.8 Å, used both site-directed and random mutagenesis to identify key residues that are required for its biological activity, and analyzed pore formation by NetB and its substitution-containing derivatives in planar lipid bilayers.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Clostridium perfringens/química , Clostridium perfringens/patogenicidad , Enterotoxinas/química , Enterotoxinas/metabolismo , Animales , Toxinas Bacterianas/genética , Transporte Biológico , Cationes/metabolismo , Pollos , Clostridium perfringens/genética , Cristalografía por Rayos X , Análisis Mutacional de ADN , Enterotoxinas/genética , Eritrocitos/efectos de los fármacos , Hemólisis , Modelos Moleculares , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína
4.
Vaccine ; 31(3): 495-9, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23176980

RESUMEN

Leptospirosis is a zoonotic disease affecting animals and humans worldwide. Leptospiral infection in cattle can cause reproductive failure and reduced weight gain, and importantly, infection represents a significant disease risk for farmers. Current bacterin vaccines offer protection that is short-lived and restricted at best to related serovars. The development of protective vaccines that stimulate immunity across multiple leptospiral serovars would therefore be advantageous. This study used a reverse vaccinology approach to evaluate a set of Leptospira borgpetersenii proteins in the hamster infection model. The L. borgpetersenii serovar Hardjo strain L550 genome sequence was analysed and genes encoding 262 predicted outer membrane or secreted proteins were selected. From this list, 238 proteins or protein fragments were successfully expressed and purified; 28 proteins (12%) were soluble, while the remaining 210 proteins (88%) were insoluble and purified under denaturing conditions. Proteins were mixed into 48 pools of up to five each and tested for protection against infection as assessed by renal colonisation in the hamster model of infection. None of the pools of antigens protected the hamsters against infection, despite a detectable antibody response being mounted against the majority of proteins (71%). This study is the first large scale evaluation of individual leptospiral proteins for ability to induce a protective immune response in the hamster infection model. It thus constitutes an important reference of protein immunogenicity and non-protective antigens that should be consulted before embarking on any future subunit vaccine experiments.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Enfermedades Renales/prevención & control , Leptospira/inmunología , Leptospirosis/prevención & control , Animales , Antígenos Bacterianos/administración & dosificación , Vacunas Bacterianas/administración & dosificación , Cricetinae , Modelos Animales de Enfermedad , Femenino , Enfermedades Renales/inmunología , Enfermedades Renales/microbiología , Leptospira/patogenicidad , Leptospirosis/inmunología , Masculino , Mesocricetus
5.
Plasmid ; 67(2): 139-47, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22244927

RESUMEN

Peptidoglycan hydrolases that are specifically associated with bacterial conjugation systems are postulated to facilitate the assembly of the transfer apparatus by creating a temporally and spatially controlled local opening in the peptidoglycan layer. To date little is known about the role of such enzymes in conjugation systems from Gram-positive bacteria. Conjugative plasmids from the Gram-positive pathogen Clostridium perfringens all encode two putative peptidoglycan hydrolases, TcpG and TcpI, within the conserved tcp transfer locus. Mutation and complementation analysis was used to demonstrate that a functional tcpG gene, but not the tcpI gene, was required for efficient conjugative transfer of pCW3. Furthermore, it was also shown that each of the two predicted catalytic domains of TcpG was functional in C. perfringens and that the predicted catalytic site residues, E-111, D-136, and C-238, present within these functional domains were required for optimal TcpG function. Escherichia coli cells producing TcpG demonstrated a distinctive autoagglutination phenotype and partially purified recombinant TcpG protein was shown to have peptidoglycan hydrolase-like activity on cognate peptidoglycan from C. perfringens. Based on these results it is suggested that TcpG is a functional peptidoglycan hydrolase that is required for efficient conjugative transfer of pCW3, presumably by facilitating the penetration of the pCW3 translocation complex through the cell wall.


Asunto(s)
Proteínas Bacterianas/genética , Clostridium perfringens/genética , Conjugación Genética , N-Acetil Muramoil-L-Alanina Amidasa/genética , Plásmidos/genética , Adhesión Bacteriana/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Catálisis , Dominio Catalítico , Clostridium perfringens/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Fenotipo , Plásmidos/metabolismo
6.
Biochem J ; 422(2): 295-303, 2009 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-19522701

RESUMEN

The complement system plays crucial roles in the immune system, but incorrect regulation causes inflammation and targeting of self-tissue, leading to diseases such as systemic lupus erythematosus, rheumatoid arthritis and age-related macular degeneration. In vivo, the initiating complexes of the classical complement and lectin pathways are controlled by SERPING1 [(C1 inhibitor) serpin peptidase inhibitor, clade G, member 1], which inactivates the components C1s and MASP-2 (mannan-binding lectin serine peptidase 2). GAGs (glycosaminoglycan) and DXS (dextran sulfate) are able to significantly accelerate SERPING1-mediated inactivation of C1s, the key effector enzyme of the classical C1 complex, although the mechanism is poorly understood. In the present study we have shown that C1s can bind to DXS and heparin and that these polyanions enhanced C1s proteolytic activity at low concentrations and inhibited it at higher concentrations. The recent determination of the crystal structure of SERPING1 has given rise to the hypothesis that both the serpin (serine protease inhibitor)-polyanion and protease-polyanion interactions might be required to accelerate the association rate of SERPING1 and C1s. To determine what proportion of the acceleration was due to protease-polyanion interactions, a chimaeric mutant of alpha1-antitrypsin containing the P4-P1 residues from the SERPING1 RCL (reactive-centre loop) was produced. Like SERPING1, this molecule is able to effectively inhibit C1s, but is unable to bind polyanions. DXS exerted a biphasic effect on the association rate of C1s which correlated strongly with the effect of DXS on C1s proteolytic activity. Thus, whereas polyanions are able to bind C1s and modulate its activity, polyanion interactions with SERPING1 must also play a vital role in the mechanism by which these cofactors accelerate the C1s-SERPING1 reaction.


Asunto(s)
Proteínas Inactivadoras del Complemento 1/metabolismo , Complemento C1s/metabolismo , Péptido Hidrolasas/metabolismo , Polímeros/metabolismo , Proteína Inhibidora del Complemento C1 , Activación Enzimática/fisiología , Humanos , Hidrólisis , Polielectrolitos , Unión Proteica/fisiología
7.
Int J Biochem Cell Biol ; 41(7): 1601-12, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19401154

RESUMEN

The newly excysted juvenile (NEJ) stage of the Fasciola hepatica lifecycle occurs just prior to invasion into the wall of the gut of the host, rendering it an important target for drug development. The cathepsin B enzymes from NEJ flukes have recently been demonstrated to be crucial to invasion and migration by the parasite. Here we characterize one of the cathepsin B enzymes (recombinant FhcatB1) from NEJ flukes. FhcatB1 has biochemical properties distinct from mammalian cathepsin B enzymes, with an atypical preference for Ile over Leu or Arg residues at the P(2) substrate position and an inability to act as an exopeptidase. FhcatB1 was active across a broad pH range (optimal activity at pH 5.5-7.0) and resistant to inhibition by cystatin family inhibitors from sheep and humans, suggesting that this enzyme would be able to function in extracellular environments in its mammalian hosts. It appears, however, that the FhcatB1 protease functions largely as a digestive enzyme in the gut of the parasite, due to the localization of a specific, fluorescently labeled inhibitor with an Ile at the P(2) position. Molecular modelling and dynamics were used to predict the basis for the unusual substrate specificity: a P(2) Ile residue positions the substrate optimally for interaction with catalytic residues of the enzyme, and the enzyme lacks an occluding loop His residue crucial for exopeptidase activity. The unique features of the enzyme, particularly with regard to its specificity and likely importance to a vital stage of the parasite's life cycle, make it an excellent target for therapeutic inhibitors or vaccination.


Asunto(s)
Catepsina B/metabolismo , Fasciola hepatica/enzimología , Fasciola hepatica/crecimiento & desarrollo , Tracto Gastrointestinal/enzimología , Estadios del Ciclo de Vida , Parásitos/enzimología , Parásitos/crecimiento & desarrollo , Animales , Dominio Catalítico , Catepsina B/antagonistas & inhibidores , Catepsina B/química , Catepsinas/antagonistas & inhibidores , Cistatinas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Activación Enzimática/efectos de los fármacos , Fasciola hepatica/efectos de los fármacos , Humanos , Cinética , Estadios del Ciclo de Vida/efectos de los fármacos , Sondas Moleculares/química , Parásitos/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Ovinos , Homología Estructural de Proteína , Especificidad por Sustrato/efectos de los fármacos
8.
J Neurochem ; 106(4): 1614-23, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18513370

RESUMEN

Tyrosine hydroxylase (TH), the rate-limiting enzyme in the biosynthesis of the catecholamines dopamine, noradrenaline and adrenaline, is regulated acutely by feedback inhibition by the catecholamines and relief of this inhibition by phosphorylation of serine 40 (Ser40). Phosphorylation of serine 40 abolishes the binding of dopamine to a high affinity (K(D) < 4 nM) site on TH, thereby increasing the activity of the enzyme. We have found that TH also contains a second low affinity (K(D) = 90 nM) dopamine-binding site, which is present in both the non-phosphorylated and the Ser40-phosphorylated forms of the enzyme. Binding of dopamine to the high-affinity site decreases V(max) and increases the K(m) for the cofactor tetrahydrobiopterin, while binding of dopamine to the low-affinity site regulates TH activity by increasing the K(m) for tetrahydrobiopterin. Kinetic analysis indicates that both sites are present in each of the four human TH isoforms. Dissociation of dopamine from the low-affinity site increases TH activity 12-fold for the non-phosphorylated enzyme and 9-fold for the Ser40-phosphorylated enzyme. The low-affinity dopamine-binding site has the potential to be the primary mechanism responsible for the regulation of catecholamine synthesis under most conditions.


Asunto(s)
Dopamina/fisiología , Tirosina 3-Monooxigenasa/metabolismo , Animales , Sitios de Unión/fisiología , Catecolaminas/biosíntesis , Dopamina/metabolismo , Activación Enzimática/fisiología , Humanos , Fosforilación , Ratas , Serina/metabolismo
9.
Biol Chem ; 387(8): 1053-61, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16895475

RESUMEN

The liver fluke, Fasciola hepatica, apparently uses a number of cysteine proteases during its life cycle, most likely for feeding, immune evasion and invasion of tissues. A cathepsin B-like enzyme (herein referred to as FhcatB1) appears to be a major enzyme secreted by the invasive, newly excysted juvenile flukes of this parasite. To examine the processing mechanisms for this enzyme, a recombinant form was expressed in Pichia pastoris and purified to yield a homogenous pool of the enzyme. The purified enzyme could be autoactivated at low pH via a bi-molecular mechanism, a process that was greatly accelerated by the presence of large, negatively charged molecules such as dextran sulfate. The enzyme could also apparently be processed to the correct size by an asparaginyl endopeptidase via cleavage in an unusual insertion N-terminal to the normal cleavage site used to yield the active form of the enzyme. Thus, there appear to be a number of ways in which this enzyme can be processed to its optimally active form prior to secretion by F. hepatica.


Asunto(s)
Catepsina B/metabolismo , Fasciola hepatica/enzimología , Fasciola hepatica/parasitología , Procesamiento Proteico-Postraduccional , Animales , Sitios de Unión , Catepsina B/efectos de los fármacos , Cristalografía por Rayos X , Sulfato de Dextran/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Heparina/farmacología , Concentración de Iones de Hidrógeno , Modelos Moleculares , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Factores de Tiempo
10.
Chem Biol Drug Des ; 68(1): 11-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16923021

RESUMEN

Inhibitors of procoagulant enzymes, such as factor Xa (fXa) and thrombin, are important for treating thrombosis. Thrombin has complex pro- and anti-coagulant roles and thus fXa is thought to represent an ideal target. Discrete kcat and Km values for cleavage of a library of fluorescence-quenched substrates by fXa were determined. The results highlighted the low selectivity of fXa at its prime sites, and its poor efficiency compared with thrombin, creating a challenge for the design of fXa-specific peptidic inhibitors. We hypothesized that Km rather than kcat/Km values may be better indicators of inhibitor potential for a peptidic sequence, leading us to design peptide sequences for both fXa and thrombin in three forms: fluorescence-quenched substrates, standard alpha-peptides and peptides containing a beta-homoarginine at the cleavage site. Kinetic and competitive inhibition assays with both fXa and thrombin showed the fluorescence-quenched substrates to be the best inhibitors, while the inhibitory effect of the beta-homoarginine peptides varied for the two proteases. Importantly, fXa was inhibited to a much greater extent by the beta-peptides than the corresponding alpha-peptides, resulting in an increased selectivity for fXa inhibition over thrombin for those peptides containing a beta-amino acid at the cleavage site.


Asunto(s)
Diseño de Fármacos , Inhibidores del Factor Xa , Péptidos/química , Inhibidores de Serina Proteinasa/química , Secuencia de Aminoácidos , Anticoagulantes/química , Dominio Catalítico , Factor Xa/química , Colorantes Fluorescentes/química , Humanos , Cinética , Ligandos , Datos de Secuencia Molecular , Especificidad por Sustrato , Trombina/antagonistas & inhibidores , Trombina/química
11.
J Biol Chem ; 280(47): 39510-4, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16169853

RESUMEN

The complement system is a central component of host defense but can also contribute to the inflammation seen in pathological conditions. The C1s protease of the first complement component, the C1 complex, initiates the pathway. In this study we have elucidated the full specificity of the enzyme for the first time using a randomized phage display library. It was found that, aside from the crucial P(1) position, the S(3) and S(2) subsites (in that order) played the greatest role in determining specificity. C1s prefers Leu or Val at P(3) and Gly or Ala residues at P(2). Apart from the S(2)' position, which showed specificity for Leu, prime subsites did not greatly affect specificity. It was evident, however, that together they significantly contributed to the efficiency of cleavage of a peptide. A peptide substrate based on the top sequence obtained in the phage display validated these results and produced the best kinetics of any C1s substrate to date. The results allow an understanding of the active site specificity of the C1s protease for the first time and provide a basis for the development of specific inhibitors aimed at controlling inflammation associated with complement activation in adverse pathological situations.


Asunto(s)
Complemento C1s/metabolismo , Vía Clásica del Complemento/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Dominio Catalítico/genética , Complemento C1s/química , ADN/genética , Humanos , Técnicas In Vitro , Modelos Moleculares , Oligopéptidos/química , Oligopéptidos/genética , Oligopéptidos/metabolismo , Biblioteca de Péptidos , Especificidad por Sustrato
12.
J Biol Chem ; 280(52): 43168-78, 2005 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-16141197

RESUMEN

Antichymotrypsin (SERPINA3) is a widely expressed member of the serpin superfamily, required for the regulation of leukocyte proteases released during an inflammatory response and with a permissive role in the development of amyloid encephalopathy. Despite its biological significance, there is at present no available structure of this serpin in its native, inhibitory state. We present here the first fully refined structure of a murine antichymotrypsin orthologue to 2.1 A, which we propose as a template for other antichymotrypsin-like serpins. A most unexpected feature of the structure of murine serpina3n is that it reveals the reactive center loop (RCL) to be partially inserted into the A beta-sheet, a structural motif associated with ligand-dependent activation in other serpins. The RCL is, in addition, stabilized by salt bridges, and its plane is oriented at 90 degrees to the RCL of antitrypsin. A biochemical and biophysical analysis of this serpin demonstrates that it is a fast and efficient inhibitor of human leukocyte elastase (ka: 4 +/- 0.9 x 10(6) m(-1) s(-)1) and cathepsin G (ka: 7.9 +/- 0.9 x 10(5) m(-1) s(-)1) giving a spectrum of activity intermediate between that of human antichymotrypsin and human antitrypsin. An evolutionary analysis reveals that residues subject to positive selection and that have contributed to the diversity of sequences in this sub-branch (A3) of the serpin superfamily are essentially restricted to the P4-P6' region of the RCL, the distal hinge, and the loop between strands 4B and 5B.


Asunto(s)
Serpinas/química , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Dicroismo Circular , Codón , Cristalografía por Rayos X , Evolución Molecular , Humanos , Inflamación , Cinética , Elastasa de Leucocito/metabolismo , Leucocitos/patología , Ligandos , Funciones de Verosimilitud , Ratones , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Filogenia , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Sales (Química)/farmacología , Homología de Secuencia de Aminoácido , Serpinas/fisiología , Temperatura , Treonina/química , Factores de Tiempo , Tripsina/química , Tripsina/farmacología
13.
Int J Biochem Cell Biol ; 36(3): 386-9, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14687916

RESUMEN

Antithrombin is a serine proteinase inhibitor (serpin) which controls the process of coagulation. It has a well defined structure, consisting of three beta-sheets, nine alpha-helices and a reactive centre loop (RCL). The RCL contains the reactive centre which harbours a bait sequence for target proteases; cleavage results in inhibition by a unique mechanism. The inhibitory activity of antithrombin is controlled by its interaction with the co-factor, heparin, which accelerates its interaction with target proteases. This ensures that heparin and its newer derivatives, such as heparin pentasaccharide, are the mainstay therapeutics for control of thrombosis or inappropriate clotting. The clinical importance of antithrombin is manifested by its clear association with thrombosis when deficiency states occur.


Asunto(s)
Antitrombina III/metabolismo , Coagulación Sanguínea , Inhibidores de Serina Proteinasa/metabolismo , Animales , Antitrombina III/química , Deficiencia de Antitrombina III/genética , Deficiencia de Antitrombina III/metabolismo , Heparina/metabolismo , Humanos , Conformación Proteica , Trombosis/etiología
14.
Biochemistry ; 42(50): 14939-45, 2003 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-14674770

RESUMEN

The classical complement pathway, which plays a vital role in preventing infection, is initiated by the action of the serine proteases C1r and C1s. We have examined the hydrolysis of substrates representing cleavage sequences in the physiological substrates for C1s, C2 and C4. These studies showed that the P(1)'-P(4)' substrate residues of C2 and C4 conferred greater affinity of substrate for enzyme and also induced a sigmoidal dependence of enzyme velocity on substrate concentration. This indicates that the substrate gave rise to homotropic positive cooperative behavior in the enzyme. When C1s was in complex with C1q and C1r, as would occur under physiological conditions, the same behavior was observed, indicating that this mechanism is relevant in the complement pathway in vivo. We further investigated the requirements of C1s for prime side amino acids by examining a substrate library in which each of the P(1)'-P(4)' positions had been substituted by different classes of amino acids. This revealed that the P(1)' position was a major determinant of the selectivity of the enzyme, while certain substitutions at the P(1)'-P(4)' positions abolished the allosteric behavior, indicating that contact residues at these positions in the C1s enzyme must mediate the cooperativity. The studies reported here highlight the importance of prime subsites in C1s for interaction with its cognate substrates in the complement pathway and therefore yield greater understanding of the mechanism of interaction between this vital protease and its physiological substrates.


Asunto(s)
Complemento C1s/química , Vía Clásica del Complemento , Serina Endopeptidasas/química , Sustitución de Aminoácidos , Sitios de Unión , Complemento C1/química , Complemento C1/metabolismo , Complemento C1s/metabolismo , Complemento C2/química , Complemento C2/metabolismo , Complemento C4/química , Complemento C4/metabolismo , Cumarinas/síntesis química , Cumarinas/metabolismo , Humanos , Hidrólisis , Oligopéptidos/síntesis química , Oligopéptidos/metabolismo , Biblioteca de Péptidos , Serina Endopeptidasas/metabolismo , Especificidad por Sustrato
15.
Biochemistry ; 42(34): 10169-73, 2003 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-12939144

RESUMEN

The shutter region of serpins consists of a number of highly conserved residues that are critical for both stability and function. Several variants of antithrombin with substitutions in this region are unstable and predispose the carrier to thrombosis. Although most mutations in the shutter region investigated to date are deleterious with respect to serpin stability and function, the substitution of Phe51 by Leu in alpha(1)-antitrypsin results in enhanced stability. Here, we have investigated the effects of introducing an analogous mutation into antithrombin (Phe 77 to Leu). The mutation did not affect the kinetics of interaction with proteases. Strikingly, however, the thermostability of the protein was markedly decreased, with the serpin displaying a 13 degrees C decrease in melting temperature as compared to wild-type recombinant antithrombin. Further studies revealed that in contrast to wild-type antithrombin, the mutant adopted the latent (inactive) conformation upon mild heating. Previous studies on shutter region mutations that destabilize antithrombin revealed that such variants possess enhanced affinity for both heparin pentasaccharide and full-length heparin. The N135A/F77L mutant had unchanged affinity for heparin pentasaccharide, but the affinity for full-length heparin was increased. We suggest that the Phe77Leu mutation causes conformational changes around the top of the D-helix in antithrombin, in particular, to the arginine 132 and 133 residues that may mediate additional antithrombin/heparin interactions. This paper also demonstrates that there are major differences between the shutter regions of antithrombin and alpha(1)-antitrypsin since a stabilizing mutation in antitrypsin has the converse effect in antithrombin.


Asunto(s)
Antitrombinas/metabolismo , Heparina/metabolismo , Sustitución de Aminoácidos , Antitrombinas/química , Antitrombinas/genética , Estabilidad de Medicamentos , Heparina/análogos & derivados , Calor , Humanos , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Concentración Osmolar , Desnaturalización Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , alfa 1-Antitripsina/química , alfa 1-Antitripsina/metabolismo
16.
J Biol Chem ; 277(18): 15971-8, 2002 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-11854268

RESUMEN

The control of coagulation enzymes by antithrombin is vital for maintenance of normal hemostasis. Antithrombin requires the co-factor, heparin, to efficiently inhibit target proteinases. A specific pentasaccharide sequence (H5) in high affinity heparin induces a conformational change in antithrombin that is particularly important for factor Xa (fXa) inhibition. Thus, synthetic H5 accelerates the interaction between antithrombin and fXa 100-fold as compared with only 2-fold versus thrombin. We built molecular models and identified residues unique to the active site of fXa that we predicted were important for interacting with the reactive center loop of H5-activated antithrombin. To test our predictions, we generated the mutants E37A, E37Q, E39A, E39Q, Q61A, S173A, and F174A in human fXa and examined the rate of association of these mutants with antithrombin in the presence and absence of H5. fXa(Q61A) interacts with antithrombin alone with a nearly normal k(ass); however, we observe only a 4-fold increase in k(ass) in the presence of H5. The x-ray crystal structure of fXa reveals that Gln(61) forms part of the S1' and S3' pocket, suggesting that the P' region of the reactive center loop of antithrombin is crucial for mediating the acceleration in the rate of inhibition of fXa by H5-activated antithrombin.


Asunto(s)
Antitrombinas/metabolismo , Proteínas de Escherichia coli , Factor Xa/metabolismo , Heparina/farmacología , Proteínas Periplasmáticas , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas Bacterianas/farmacología , Línea Celular , Factor Xa/química , Factor Xa/genética , Heparina/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oligosacáridos/química , Oligosacáridos/metabolismo , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...