Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NPJ Vaccines ; 8(1): 176, 2023 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-37952003

RESUMEN

Seasonal influenza vaccines must be updated annually and suboptimally protect against strains mismatched to the selected vaccine strains. We previously developed a subunit vaccine antigen consisting of a stabilized trimeric influenza A group 1 hemagglutinin (H1) stem protein that elicits broadly neutralizing antibodies. Here, we further optimized the stability and manufacturability of the H1 stem antigen (H1 stem v2, also known as INFLUENZA G1 mHA) and characterized its formulation and potency with different adjuvants in vitro and in animal models. The recombinant H1 stem antigen (50 µg) was administered to influenza-naïve non-human primates either with aluminum hydroxide [Al(OH)3] + NaCl, AS01B, or SLA-LSQ formulations at week 0, 8 and 34. These SLA-LSQ formulations comprised of varying ratios of the synthetic TLR4 agonist 'second generation synthetic lipid adjuvant' (SLA) with liposomal QS-21 (LSQ). A vaccine formulation with aluminum hydroxide or SLA-LSQ (starting at a 10:25 µg ratio) induced HA-specific antibodies and breadth of neutralization against a panel of influenza A group 1 pseudoviruses, comparable with vaccine formulated with AS01B, four weeks after the second immunization. A formulation with SLA-LSQ in a 5:2 µg ratio contained larger fused or aggregated liposomes and induced significantly lower humoral responses. Broadly HA stem-binding antibodies were detectable for the entire period after the second vaccine dose up to week 34, after which they were boosted by a third vaccine dose. These findings inform about potential adjuvant formulations in clinical trials with an H1 stem-based vaccine candidate.

2.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35131851

RESUMEN

For an efficacious vaccine immunogen, influenza hemagglutinin (HA) needs to maintain a stable quaternary structure, which is contrary to the inherently dynamic and metastable nature of class I fusion proteins. In this study, we stabilized HA with three substitutions within its pH-sensitive regions where the refolding starts. An X-ray structure reveals how these substitutions stabilize the intersubunit ß-sheet in the base and form an interprotomeric aliphatic layer across the stem while the native prefusion HA fold is retained. The identification of the stabilizing substitutions increases our understanding of how the pH sensitivity is structurally accomplished in HA and possibly other pH-sensitive class I fusion proteins. Our stabilization approach in combination with the occasional back mutation of rare amino acids to consensus results in well-expressing stable trimeric HAs. This repair and stabilization approach, which proves broadly applicable to all tested influenza A HAs of group 1 and 2, will improve the developability of influenza vaccines based on different types of platforms and formats and can potentially improve efficacy.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Hemaglutininas/genética , Aminoácidos/genética , Línea Celular , Humanos , Concentración de Iones de Hidrógeno , Vacunas contra la Influenza/genética , Gripe Humana/virología , Mutación/genética , Conformación Proteica en Lámina beta/genética
3.
Protein Sci ; 27(2): 509-522, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29114958

RESUMEN

Neutrophils contain high levels of chymotrypsin-like serine proteases (NSPs) within their azurophilic granules that have a multitude of functions within the immune system. In response, the pathogen Staphylococcus aureus has evolved three potent inhibitors (Eap, EapH1, and EapH2) that protect the bacterium as well as several of its secreted virulence factors from the degradative action of NSPs. We previously showed that these so-called EAP domain proteins represent a novel class of NSP inhibitors characterized by a non-covalent inhibitory mechanism and a distinct target specificity profile. Based upon high levels of structural homology amongst the EAP proteins and the NSPs, as well as supporting biochemical data, we predicted that the inhibited complex would be similar for all EAP/NSP pairs. However, we present here evidence that EapH1 and EapH2 bind the canonical NSP, Neutrophil Elastase (NE), in distinct orientations. We discovered that alteration of EapH1 residues at the EapH1/NE interface caused a dramatic loss of affinity and inhibition of NE, while mutation of equivalent positions in EapH2 had no effect on NE binding or inhibition. Surprisingly, mutation of residues in an altogether different region of EapH2 severely impacted both the NE binding and inhibitory properties of EapH2. Even though EapH1 and EapH2 bind and inhibit NE and a second NSP, Cathepsin G, equally well, neither of these proteins interacts with the structurally related, but non-proteolytic granule protein, azurocidin. These studies expand our understanding of EAP/NSP interactions and suggest that members of this immune evasion protein family are capable of diverse target recognition modes.


Asunto(s)
Elastasa de Leucocito/metabolismo , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/metabolismo , Staphylococcus aureus/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Línea Celular , Humanos , Evasión Inmune , Modelos Moleculares , Mutación , Unión Proteica , Dominios Proteicos , Inhibidores de Serina Proteinasa/genética , Staphylococcus aureus/enzimología
4.
Science ; 349(6254): 1301-6, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26303961

RESUMEN

The identification of human broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin (HA) stem revitalized hopes of developing a universal influenza vaccine. Using a rational design and library approach, we engineered stable HA stem antigens ("mini-HAs") based on an H1 subtype sequence. Our most advanced candidate exhibits structural and bnAb binding properties comparable to those of full-length HA, completely protects mice in lethal heterologous and heterosubtypic challenge models, and reduces fever after sublethal challenge in cynomolgus monkeys. Antibodies elicited by this mini-HA in mice and nonhuman primates bound a wide range of HAs, competed with human bnAbs for HA stem binding, neutralized H5N1 viruses, and mediated antibody-dependent effector activity. These results represent a proof of concept for the design of HA stem mimics that elicit bnAbs against influenza A group 1 viruses.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Humanos , Ratones , Multimerización de Proteína , Estructura Secundaria de Proteína
5.
Appl Microbiol Biotechnol ; 99(21): 9037-48, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26160391

RESUMEN

Recent studies have shown that the Gram-positive bacterium Lactococcus lactis can be exploited for the expression of heterologous proteins; however, a versatile set of vectors suitable for inducible extracellular protein production and subsequent purification of the expressed proteins by immobilized metal affinity chromatography was so far lacking. Here we describe three novel vectors that, respectively, facilitate the nisin-inducible production of N- or C-terminally hexa-histidine (His6)-tagged proteins in L. lactis. One of these vectors also encodes a tobacco etch virus (TEV) protease cleavage site allowing removal of the N-terminal His6-tag from expressed proteins. Successful application of the developed vectors for protein expression, purification and/or functional studies is exemplified with six different cell wall-bound or secreted proteins from Staphylococcus aureus. The results show that secretory production of S. aureus proteins is affected by the position, N- or C-terminal, of the His6-tag. This seems to be due to an influence of the His6-tag on protein stability. Intriguingly, the S. aureus IsdB protein, which is phosphorylated in S. aureus, was also found to be phosphorylated when heterologously produced in L. lactis, albeit not on the same Tyr residue. This implies that this particular post-translational protein modification is to some extent conserved in S. aureus and L. lactis. Altogether, we are confident that the present vector set combined with the L. lactis expression host has the potential to become a very useful tool in optimization of the expression, purification and functional analysis of extracytoplasmic bacterial proteins.


Asunto(s)
Vectores Genéticos , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Ingeniería de Proteínas/métodos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Cromatografía de Afinidad , Nisina/metabolismo , Péptido Hidrolasas/metabolismo , Proteolisis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Staphylococcus aureus/genética , Activación Transcripcional/efectos de los fármacos
6.
Proc Natl Acad Sci U S A ; 111(36): 13187-92, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25161283

RESUMEN

Neutrophils are indispensable for clearing infections with the prominent human pathogen Staphylococcus aureus. Here, we report that S. aureus secretes a family of proteins that potently inhibits the activity of neutrophil serine proteases (NSPs): neutrophil elastase (NE), proteinase 3, and cathepsin G. The NSPs, but not related serine proteases, are specifically blocked by the extracellular adherence protein (Eap) and the functionally orphan Eap homologs EapH1 and EapH2, with inhibitory-constant values in the low-nanomolar range. Eap proteins are together essential for NSP inhibition by S. aureus in vitro and promote staphylococcal infection in vivo. The crystal structure of the EapH1/NE complex showed that Eap molecules constitute a unique class of noncovalent protease inhibitors that occlude the catalytic cleft of NSPs. These findings increase our insights into the complex pathogenesis of S. aureus infections and create opportunities to design novel treatment strategies for inflammatory conditions related to excessive NSP activity.


Asunto(s)
Neutrófilos/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , Staphylococcus aureus/metabolismo , Animales , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Biocatálisis , Espacio Extracelular/metabolismo , Femenino , Humanos , Elastasa de Leucocito/antagonistas & inhibidores , Elastasa de Leucocito/metabolismo , Ratones Endogámicos C57BL , Modelos Moleculares , Infecciones Estafilocócicas/patología
7.
Immunol Lett ; 146(1-2): 31-9, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22575527

RESUMEN

Microbial resistance to antimicrobial drugs is promoting a search for new antimicrobial agents that target highly conservative structures of pathogens. Human CD14 - a known pattern recognition receptor (PRR) which recognizes multiple ligands from different microbes might be a worthy candidate. The aim of our work was to create a CD14/Fc dimer protein and evaluate its whole bacteria binding and opsonizing capabilities. Fusion of CD14 with the fragment crystallisable (Fc) part of human IgG1 could not only lead to an artificial opsonin but the dimerization through the Fc part might also increase its affinity to different ligands. Human CD14 and the Fc part of human IgG1 was fused and expressed in HEK293 cells. A histidine tagged CD14 (CD14/His) was also expressed as control. Using flow cytometry we could prove that CD14/Fc bound to whole Gram-negative bacteria, especially to short lipopolysaccharide (Ra and Re) mutants, and weak interaction was observed between the fusion protein and Listeria monocytogenes. Other Gram-positive bacteria and fungi did not show any association with CD14/Fc. CD14/His showed about 50-times less potent binding to Gram-negative bacteria. CD14/Fc acted as an opsonin and enhanced phagocytosis of these bacteria by neutrophil granulocytes, monocyte-derived macrophages and dendritic cells. Internalization of bacteria was confirmed by trypan blue quenching and confocal microscopy. On neutrophils the Fc part of the fusion protein was recognized by Fc receptors (CD16, CD32), as determined by blocking experiments. CD14/Fc enhanced the killing of bacteria in an ex vivo whole blood assay. Our experiments confirm that PRR/Fc fusion proteins can give a boost to FcR dependent phagocytosis and killing provided the antimicrobial part binds efficiently to microbes.


Asunto(s)
Bacterias Gramnegativas/inmunología , Fragmentos Fc de Inmunoglobulinas/genética , Receptores de Lipopolisacáridos/genética , Neutrófilos/inmunología , Proteínas Opsoninas/inmunología , Receptores Fc/inmunología , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Citometría de Flujo , Expresión Génica/inmunología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/crecimiento & desarrollo , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/crecimiento & desarrollo , Bacterias Grampositivas/inmunología , Células HEK293 , Humanos , Inmunidad Innata , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Neutrófilos/efectos de los fármacos , Neutrófilos/microbiología , Proteínas Opsoninas/biosíntesis , Proteínas Opsoninas/genética , Fagocitosis/inmunología , Unión Proteica , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Transfección
8.
J Mol Biol ; 415(3): 573-83, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22154939

RESUMEN

Pseudomonas aeruginosa secretes the virulence factor alkaline protease (AprA) to enhance its survival. AprA cleaves one of the key microbial recognition molecules, monomeric flagellin, and thereby diminishes Toll-like receptor 5 activation. In addition, AprA degrades host proteins such as complement proteins and cytokines. P. aeruginosa encodes a highly potent inhibitor of alkaline protease (AprI) that is solely located in the periplasm where it is presumed to protect periplasmic proteins against secreted AprA. We set out to study the enzyme-inhibitor interactions in more detail in order to provide a basis for future drug development. Structural and mutational studies reveal that the conserved N-terminal residues of AprI occupy the protease active site and are essential for inhibitory activity. We constructed peptides mimicking the N-terminus of AprI; however, these were incapable of inhibiting AprA-mediated flagellin cleavage. Furthermore, we expressed and purified AprI of P. aeruginosa and the homologous (37% sequence identity) AprI of Pseudomonas syringae, which remarkably show species specificity for their cognate protease. Exchange of the first five N-terminal residues between AprI of P. syringae and P. aeruginosa did not affect the observed specificity, whereas exchange of only six residues located at the AprI surface that contacts the protease did abolish specificity. These findings are elementary steps toward the design of molecules derived from the natural inhibitor of the virulence factor AprA and their use in therapeutic applications in Pseudomonas and other Gram-negative infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endopeptidasas/metabolismo , Inhibidores de Proteasas/metabolismo , Pseudomonas aeruginosa/enzimología , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Dominio Catalítico , Dicroismo Circular , Análisis Mutacional de ADN , Endopeptidasas/genética , Flagelina/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Proteolisis , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Pseudomonas syringae/enzimología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Factores de Virulencia/genética
9.
J Immunol ; 188(1): 386-93, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22131330

RESUMEN

The complement system rapidly detects and kills Gram-negative bacteria and supports bacterial killing by phagocytes. However, bacterial pathogens exploit several strategies to evade detection by the complement system. The alkaline protease (AprA) of Pseudomonas aeruginosa has been associated with bacterial virulence and is known to interfere with complement-mediated lysis of erythrocytes, but its exact role in bacterial complement escape is unknown. In this study, we analyzed how AprA interferes with complement activation and whether it could block complement-dependent neutrophil functions. We found that AprA potently blocked phagocytosis and killing of Pseudomonas by human neutrophils. Furthermore, AprA inhibited opsonization of bacteria with C3b and the formation of the chemotactic agent C5a. AprA specifically blocked C3b deposition via the classical and lectin pathways, whereas the alternative pathway was not affected. Serum degradation assays revealed that AprA degrades both human C1s and C2. However, repletion assays demonstrated that the mechanism of action for complement inhibition is cleavage of C2. In summary, we showed that P. aeruginosa AprA interferes with classical and lectin pathway-mediated complement activation via cleavage of C2.


Asunto(s)
Proteínas Bacterianas/inmunología , Exopeptidasas/inmunología , Neutrófilos/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/patogenicidad , Factores de Virulencia/inmunología , Proteínas Bacterianas/metabolismo , Complemento C2/inmunología , Complemento C2/metabolismo , Complemento C3b/inmunología , Complemento C3b/metabolismo , Complemento C5a/inmunología , Complemento C5a/metabolismo , Lectina de Unión a Manosa de la Vía del Complemento , Exopeptidasas/metabolismo , Humanos , Evasión Inmune , Neutrófilos/metabolismo , Fagocitosis/inmunología , Infecciones por Pseudomonas/enzimología , Pseudomonas aeruginosa/enzimología , Factores de Virulencia/metabolismo
10.
J Mol Med (Berl) ; 88(2): 115-20, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20062962

RESUMEN

The plasma proteins of the complement system are essential in the innate immune response against bacteria. Complement labels bacteria with opsonins to support phagocytosis and generates chemoattractants to attract phagocytes to the site of infection. In turn, bacterial human pathogens have evolved different strategies to specifically impair the complement response. Here, we review the large arsenal of complement inhibitors produced by the gram-positive pathogens Staphylococcus aureus and Group A Streptococcus. We discuss how these bacterial molecules provide us with new tools to treat both infectious and inflammatory disease conditions in humans.


Asunto(s)
Proteínas Bacterianas/metabolismo , Activación de Complemento/inmunología , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes/patogenicidad , Proteínas Bacterianas/genética , Bacterias Grampositivas/genética , Bacterias Grampositivas/metabolismo , Bacterias Grampositivas/patogenicidad , Humanos , Evasión Inmune , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo
11.
Nat Rev Immunol ; 8(1): 48-58, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18064050

RESUMEN

Complement in mammalian plasma recognizes pathogenic, immunogenic and apoptotic cell surfaces, promotes inflammatory responses and marks particles for cell lysis, phagocytosis and B-cell stimulation. At the heart of the complement system are two large proteins, complement component C3 and protease factor B. These two proteins are pivotal for amplification of the complement response and for labelling of the target particles, steps that are required for effective clearance of the target. Here we review the molecular mechanisms of complement activation, in which proteolysis and complex formation result in large conformational changes that underlie the key offensive step of complement executed by C3 and factor B. Insights into the mechanisms of complement amplification are crucial for understanding host defence and pathogen immune evasion, and for the development of complement-immune therapies.


Asunto(s)
Activación de Complemento/inmunología , Complemento C3/inmunología , Factor B del Complemento/inmunología , Animales , Humanos
12.
J Immunol ; 179(5): 2989-98, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17709514

RESUMEN

The pathogenic bacterium Staphylococcus aureus counteracts the host immune defense by excretion of the 85 residue staphylococcal complement inhibitor (SCIN). SCIN inhibits the central complement convertases; thereby, it reduces phagocytosis following opsonization and efficiently blocks all downstream effector functions. In this study, we present the crystal structure of SCIN at 1.8 A resolution and the identification of its active site. Functional characterization of structure based chimeric proteins, consisting of SCIN and the structurally but nonfunctional homologue open reading frame-D, indicate an 18-residue segment (Leu-31-Gly-48) crucial for SCIN activity. In all complement activation pathways, chimeras lacking these SCIN residues completely fail to inhibit production of the potent mediator of inflammation C5a. Inhibition of alternative pathway-mediated opsonization (C3b deposition) and formation of the lytic membrane attack complex (C5b-9 deposition) are strongly reduced for these chimeras as well. For inhibition of the classical/lectin pathway-mediated C3b and C5b-9 deposition, the same residues are critical although additional sites are involved. These chimeras also display reduced capacity to stabilize the C3 convertases of both the alternative and the classical/lectin pathway indicating the stabilizing effect is pivotal for the complement inhibitory activity of SCIN. Because SCIN specifically and efficiently inhibits complement, it has a high potential in anti-inflammatory therapy. Our data are a first step toward the development of a second generation molecule suitable for such therapeutic complement intervention.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Inactivadoras de Complemento/química , Staphylococcus aureus/patogenicidad , Secuencia de Aminoácidos , Sitios de Unión , Complemento C5a/antagonistas & inhibidores , Cristalografía por Rayos X , Humanos , Datos de Secuencia Molecular , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Staphylococcus aureus/inmunología
13.
Nat Struct Mol Biol ; 14(3): 224-8, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17310251

RESUMEN

Factor B is the central protease of the complement system of immune defense. Here, we present the crystal structure of human factor B at 2.3-A resolution, which reveals how the five-domain proenzyme is kept securely inactive. The canonical activation helix of the Von Willebrand factor A (VWA) domain is displaced by a helix from the preceding domain linker. The two helices conformationally link the scissile-activation peptide and the metal ion-dependent adhesion site required for binding of the ligand C3b. The data suggest that C3b binding displaces the three N-terminal control domains and reshuffles the two central helices. Reshuffling of the helices releases the scissile bond for final proteolytic activation and generates a new interface between the VWA domain and the serine protease domain. This allosteric mechanism is crucial for tight regulation of the complement-amplification step in the immune response.


Asunto(s)
Factor B del Complemento/química , Factor B del Complemento/metabolismo , Proteínas del Sistema Complemento/inmunología , Dominio Catalítico , Convertasas de Complemento C3-C5/química , Cristalografía por Rayos X , Activación Enzimática , Humanos , Modelos Moleculares , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Secuencias Reguladoras de Ácidos Nucleicos/genética , Relación Estructura-Actividad , Factor de von Willebrand/química
14.
Structure ; 14(10): 1587-97, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17027507

RESUMEN

C2a provides the catalytic center to the convertase complexes of the classical and lectin-binding pathways of complement activation. We determined two crystal structures of full-length C2a, with and without a pseudo ligand bound. Both structures reveal a near-active conformation of the catalytic center of the serine protease domains, while the von Willebrand factor A-type domains display an intermediate activation state of helix alpha7 with an open, activated metal-ion-dependent adhesion site. The open adhesion site likely serves to enhance the affinity for the ligand C4b, similar to "inside-out" signaling in integrins. Surprisingly, the N-terminal residues of C2a are buried in a crevice near helix alpha7, indicative of a structural switch between C2 and C2a. Extended loops on the protease domain possibly envelop the protruding anaphylatoxin domain of the substrate C3. Together with a putative substrate-induced completion of the oxyanion hole, this may contribute to the high substrate specificity of the convertases.


Asunto(s)
Complemento C2a/química , Modelos Moleculares , Aminoácidos/química , Aminoácidos/genética , Dominio Catalítico , Activación de Complemento , Complemento C2a/genética , Humanos , Ligandos , Mutación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Especificidad por Sustrato
15.
Biochim Biophys Acta ; 1619(2): 139-43, 2003 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-12527109

RESUMEN

The NifL PAS domain from Azotobacter vinelandii is a flavoprotein with FAD as the prosthetic group. Here we describe a novel immobilization procedure for the large-scale preparation of apo NifL PAS domain and its efficient reconstitution with either 2,4a-13C-FAD or 2,4a-13C-FMN. In this procedure, the His-tagged holoprotein is bound to an immobilized metal affinity column and the flavin is released by washing the column with buffer containing 2 M KBr and 2 M urea. The apoprotein is reconstituted on-column with the (artificial) flavin cofactor, and then eluted with buffer containing 250 mM imidazole. Alternatively, the immobilized apoprotein can be released from the column matrix before reconstitution. The His-tag based immobilization method of preparing reconstituted (or apo) NifL PAS domain protein has the advantage that it combines a protein affinity chromatography technique with limited protein loss, resulting in a high protein yield with extremely efficient flavin reconstitution. This on-column reconstitution method can also be used in cases where the apoprotein is unstable. Therefore, it may develop as a universal method for replacement of flavin or other cofactors.


Asunto(s)
Proteínas Bacterianas/química , Cromatografía de Afinidad/métodos , Flavoproteínas/química , Apoproteínas/química , Azotobacter vinelandii/genética , Proteínas Bacterianas/biosíntesis , Dicroismo Circular , Escherichia coli/metabolismo , Mononucleótido de Flavina/química , Flavina-Adenina Dinucleótido/química , Flavoproteínas/biosíntesis , Vectores Genéticos , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína , Proteínas Recombinantes/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...