Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Protein Sci ; 31(10): e4432, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36173177

RESUMO

Structure determination of macromolecular complexes is challenging if subunits can dissociate during crystallization or preparation of electron microscopy grids. We present an approach where a labile complex is stabilized by linking subunits though introduction of a peptide tag in one subunit that is recognized by a nanobody tethered to a second subunit. This allowed crystal structure determination at 3.9 Å resolution of the highly non-globular 320 kDa proconvertase formed by complement components C3b, factor B, and properdin. Whereas the binding mode of properdin to C3b is preserved, an internal rearrangement occurs in the zymogen factor B von Willebrand domain type A domain compared to the proconvertase not bound to properdin. The structure emphasizes the role of two noncanonical loops in thrombospondin repeats 5 and 6 of properdin in augmenting the activity of the C3 convertase. We suggest that linking of subunits through peptide specific tethered nanobodies represents a simple alternative to approaches like affinity maturation and chemical cross-linking for the stabilization of large macromolecular complexes. Besides applications for structural biology, nanobody bridging may become a new tool for biochemical analysis of unstable macromolecular complexes and in vitro selection of highly specific binders for such complexes.


Assuntos
Properdina , Anticorpos de Domínio Único , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/metabolismo , Fator B do Complemento/química , Fator B do Complemento/metabolismo , Precursores Enzimáticos , Substâncias Macromoleculares , Properdina/química , Properdina/metabolismo , Trombospondinas
2.
Biomolecules ; 11(2)2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33671302

RESUMO

The complement system is part of the innate immune response, where it provides immediate protection from infectious agents and plays a fundamental role in homeostasis. Complement dysregulation occurs in several diseases, where the tightly regulated proteolytic cascade turns offensive. Prominent examples are atypical hemolytic uremic syndrome, paroxysmal nocturnal hemoglobinuria and Alzheimer's disease. Therapeutic intervention targeting complement activation may allow treatment of such debilitating diseases. In this review, we describe a panel of complement targeting nanobodies that allow modulation at different steps of the proteolytic cascade, from the activation of the C1 complex in the classical pathway to formation of the C5 convertase in the terminal pathway. Thorough structural and functional characterization has provided a deep mechanistic understanding of the mode of inhibition for each of the nanobodies. These complement specific nanobodies are novel powerful probes for basic research and offer new opportunities for in vivo complement modulation.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento , Nanomedicina/métodos , Anticorpos de Domínio Único/química , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C1/química , Convertases de Complemento C3-C5/química , Epitopos/química , Hemoglobinúria Paroxística/tratamento farmacológico , Hemoglobinúria Paroxística/imunologia , Humanos , Imunidade Inata , Imunoglobulina G/imunologia , Inflamação , Conformação Molecular , Ligação Proteica , Proteólise
3.
Toxicon ; 184: 68-77, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32526239

RESUMO

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. CVF, like C3b, forms a convertase with factor B. This bimolecular complex CVF, Bb is an enzyme that cleaves C3 and C5. However, CVF, Bb exhibits significantly different functional properties from C3b,Bb. Whereas both, CVF, Bb and C3b, Bb exhibit spontaneous decay-dissociation into the respective subunits, thereby eliminating the enzymatic activity, the CVF, Bb convertase is physico-chemically far more stable, decaying with a half-life that is more than two orders of magnitude slower than that of C3b,Bb. In addition, CVF, Bb is completely resistant to inactivation by Factors H and I. These two properties of CVF, Bb allow continuous activation of C3 and C5, and complement depletion in serum. In order to understand the structural basis for the physico-chemical stability of CVF,Bb, we have created recombinant hybrid proteins of CVF and human C3, based on structural differences between CVF and human C3b in the C-terminal C345C domain. Here we describe three human C3/CVF hybrid proteins which differ in only one, two, or five amino acid residues from earlier described hybrid proteins. In all three cases, the hybrid proteins containing CVF residues form more stable convertases, and exhibit stronger complement-depletion activity than hybrid proteins with human C3 residues. Three bonds between CVF residues and Factor Bb residues could be identified by crystallographic modeling that contribute to the greater stability of the convertases.


Assuntos
Convertases de Complemento C3-C5/química , Fator B do Complemento/química , Venenos Elapídicos/química , Animais , Complemento C3 , Fator H do Complemento , Humanos , Proteínas Recombinantes de Fusão
4.
Molecules ; 24(18)2019 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-31505853

RESUMO

Complement (C) activation can underlie the infusion reactions to liposomes and other nanoparticle-based medicines, a hypersensitivity syndrome that can be partially reproduced in animal models. However, the sensitivities and manifestations substantially differ in different species, and C activation may not be the only cause of pathophysiological changes. In order to map the species variation of C-dependent and -independent pseudoallergy (CARPA/CIPA), here we used known C activators and C activator liposomes to compare their acute hemodynamic, hematological, and biochemical effects in rats. These C activators were cobra venom factor (CVF), zymosan, AmBisome (at 2 doses), its amphotericin B-free vehicle (AmBisombo), and a PEGylated cholesterol-containing liposome (PEG-2000-chol), all having different powers to activate C in rat blood. The pathophysiological endpoints measured were blood pressure, leukocyte and platelet counts, and plasma thromboxane B2, while C activation was assessed by C3 consumption using the Pan-Specific C3 assay. The results showed strong linear correlation between C activation and systemic hypotension, pointing to a causal role of C activation in the hemodynamic changes. The observed thrombocytopenia and leukopenia followed by leukocytosis also correlated with C3 conversion in case of C activators, but not necessarily with C activation by liposomes. These findings are consistent with the double hit hypothesis of hypersensitivity reactions (HSRs), inasmuch as strong C activation can fully account for all symptoms of HSRs, but in case of no-, or weak C activators, the pathophysiological response, if any, is likely to involve other activation pathways.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Síndrome de Hipersensibilidade a Medicamentos/tratamento farmacológico , Leucocitose/sangue , Lipossomos/farmacologia , Anfotericina B/química , Anfotericina B/farmacologia , Animais , Colesterol/química , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/farmacologia , Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/metabolismo , Síndrome de Hipersensibilidade a Medicamentos/etiologia , Síndrome de Hipersensibilidade a Medicamentos/patologia , Venenos Elapídicos/química , Venenos Elapídicos/farmacologia , Humanos , Hipotensão/sangue , Hipotensão/induzido quimicamente , Leucocitose/induzido quimicamente , Leucopenia/sangue , Leucopenia/induzido quimicamente , Lipossomos/química , Nanopartículas/química , Polietilenoglicóis/química , Ratos , Trombocitopenia/sangue , Trombocitopenia/induzido quimicamente , Zimosan/química , Zimosan/farmacologia
5.
Front Immunol ; 10: 2007, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507604

RESUMO

Properdin (FP) is a positive regulator of the immune system stimulating the activity of the proteolytically active C3 convertase C3bBb in the alternative pathway of the complement system. Here we present two crystal structures of FP and two structures of convertase bound FP. A structural core formed by three thrombospondin repeats (TSRs) and a TB domain harbors the convertase binding site in FP that mainly interacts with C3b. Stabilization of the interaction between the C3b C-terminus and the MIDAS bound Mg2+ in the Bb protease by FP TSR5 is proposed to underlie FP convertase stabilization. Intermolecular contacts between FP and the convertase subunits suggested by the structure were confirmed by binding experiments. FP is shown to inhibit C3b degradation by FI due to a direct competition for a common binding site on C3b. FP oligomers are held together by two sets of intermolecular contacts, where the first is formed by the TB domain from one FP molecule and TSR4 from another. The second and largest interface is formed by TSR1 and TSR6 from the same two FP molecules. Flexibility at four hinges between thrombospondin repeats is suggested to enable the oligomeric, polydisperse, and extended architecture of FP. Our structures rationalize the effects of mutations associated with FP deficiencies and provide a structural basis for the analysis of FP function in convertases and its possible role in pattern recognition.


Assuntos
Convertases de Complemento C3-C5/química , Proteínas do Sistema Complemento/química , Properdina/química , Multimerização Proteica , Sítios de Ligação , Convertases de Complemento C3-C5/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutação , Properdina/metabolismo , Ligação Proteica , Conformação Proteica , Processamento de Proteína Pós-Traducional , Proteólise , Relação Estrutura-Atividade
6.
Semin Immunopathol ; 40(1): 15-35, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29167939

RESUMO

This review is not intended to cover in detail all aspects of the discovery and evolution of our understanding of the "alternative pathway" of complement activation, there are many excellent reviews that do this (see Fearon (CRC Crit Rev Immunol 1:1-32, 1979), Pangburn and Müller-Eberhard (Springer Semin Immunopathol 7:163-192, 1984)), but instead to give sufficient background for current concepts to be put in context. The prevailing textbook view, of components having a primary role as an alternative "pathway" for C3 activation, is challenged, with an argument developed for the primary role of the system being that of providing a surface-dependent amplification loop for both C3 and C5 activation. Whatever the mechanism by which the initial C3b molecule is generated, deposition onto a surface has the potential to target that surface for elimination. Elimination or escape from initial targeting is determined by a sophisticated and highly regulated amplification loop for C3 activation. This viewpoint of the system is then briefly developed to provide a context for therapeutic treatment of disease caused, at least in part, by dysregulated amplification of C3 activation, and to highlight some of the challenges that such therapies will face and need to address.


Assuntos
Via Alternativa do Complemento , Properdina/metabolismo , Transdução de Sinais , Animais , Proteínas de Transporte/metabolismo , Membrana Celular/imunologia , Membrana Celular/metabolismo , Ativação do Complemento/imunologia , Fator Nefrítico do Complemento 3/imunologia , Fator Nefrítico do Complemento 3/metabolismo , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/imunologia , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento C3b/imunologia , Proteínas Inativadoras do Complemento C3b/metabolismo , Venenos Elapídicos/imunologia , Venenos Elapídicos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Properdina/imunologia , Ligação Proteica
7.
Semin Immunopathol ; 40(1): 3-14, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28808775

RESUMO

During the last decade, the complement field has experienced outstanding advancements in the mechanistic understanding of how complement activators are recognized, what C3 activation means, how protein complexes like the C3 convertases and the membrane attack complex are assembled, and how positive and negative complement regulators perform their function. All of this has been made possible mostly because of the contributions of structural biology to the study of the complement components. The wealth of novel structural data has frequently provided support to previously held knowledge, but often has added alternative and unexpected insights into complement function. Here, we will review some of these findings focusing in the alternative and terminal complement pathways.


Assuntos
Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/fisiologia , Animais , Proteínas de Transporte , Ativação do Complemento/imunologia , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/imunologia , Convertases de Complemento C3-C5/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Ligação Proteica , Conformação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Especificidade por Substrato
8.
EMBO J ; 36(8): 1084-1099, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28264884

RESUMO

Properdin (FP) is an essential positive regulator of the complement alternative pathway (AP) providing stabilization of the C3 and C5 convertases, but its oligomeric nature challenges structural analysis. We describe here a novel FP deficiency (E244K) caused by a single point mutation which results in a very low level of AP activity. Recombinant FP E244K is monomeric, fails to support bacteriolysis, and binds weakly to C3 products. We compare this to a monomeric unit excised from oligomeric FP, which is also dysfunctional in bacteriolysis but binds the AP proconvertase, C3 convertase, C3 products and partially stabilizes the convertase. The crystal structure of such a FP-convertase complex suggests that the major contact between FP and the AP convertase is mediated by a single FP thrombospondin repeat and a small region in C3b. Small angle X-ray scattering indicates that FP E244K is trapped in a compact conformation preventing its oligomerization. Our studies demonstrate an essential role of FP oligomerization in vivo while our monomers enable detailed structural insight paving the way for novel modulators of complement.


Assuntos
Convertases de Complemento C3-C5/química , Via Alternativa do Complemento , Properdina/química , Multimerização Proteica , Substituição de Aminoácidos , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Humanos , Mutação de Sentido Incorreto , Properdina/deficiência , Properdina/genética , Properdina/metabolismo , Domínios Proteicos
9.
J Biol Chem ; 291(32): 16494-507, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27252379

RESUMO

The lectin (LP) and classical (CP) pathways are two of the three main activation cascades of the complement system. These pathways start with recognition of different pathogen- or danger-associated molecular patterns and include identical steps of proteolytic activation of complement component C4, formation of the C3 proconvertase C4b2, followed by cleavage of complement component C2 within C4b2 resulting in the C3 convertase C4b2a. Here, we describe the solution structures of the two central complexes of the pathways, C3 proconvertase and C3 convertase, as well as the unbound zymogen C2 obtained by small angle x-ray scattering analysis. We analyzed both native and enzymatically deglycosylated C4b2 and C2 and showed that the resulting structural models were independent of the glycans. The small angle x-ray scattering-derived models suggest a different activation mode for the CP/LP C3 proconvertase as compared with that established for the alternative pathway proconvertase C3bB. This is likely due to the rather different structural and functional properties of the proteases activating the proconvertases. The solution structure of a stabilized form of the active CP/LP C3 convertase C4b2a is strikingly similar to the crystal structure of the alternative pathway C3 convertase C3bBb, which is in accordance with their identical functions in cleaving the complement proteins C3 and C5.


Assuntos
Complemento C2/química , Convertases de Complemento C3-C5/química , Complemento C4/química , Humanos , Difração de Raios X
10.
Nucleic Acid Ther ; 24(5): 326-35, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25093529

RESUMO

The species sensitivity and mechanism of complement pathway activation by a phosphorothioate oligonucleotide were investigated in monkey and human serum. Increasing concentrations of a phosphorothioate oligonucleotide, ISIS 2302, were incubated in either monkey or human serum. Complement activation in monkey serum was selective for the alternative pathway and occurred at concentrations ≥ 50 µg/mL ISIS 2302. By comparison, complement activation in human serum was absent. A similar difference in sensitivity for activation was also observed for a representative 2'-methoxyethyl (MOE)-modified oligonucleotide. The absence of oligonucleotide-induced complement activation was also observed in dogs. Protein binding with ISIS 2302 and enzyme competition studies suggested that factor H was important in oligonucleotide-mediated complement activation process, and addition of factor H to serum effectively prevented the activation in monkey serum. Furthermore, based on the immunoassay for factor H, there was an apparent decrease in factor H concentration as the ISIS 2302 concentration increased. This result suggests that ISIS 2302 binds to factor H and interferes with the factor H antibody from the immunoassay. Factor H is a regulatory protein that limits alternative pathway activation. Disruption of factor H interaction with C3 convertase by oligonucleotide could promote activation in this pathway.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Fator H do Complemento/metabolismo , Fármacos Gastrointestinais/sangue , Imunossupressores/sangue , Oligodesoxirribonucleotídeos Antissenso/sangue , Oligonucleotídeos Fosforotioatos/sangue , Animais , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/metabolismo , Fator H do Complemento/química , Via Alternativa do Complemento/efeitos dos fármacos , Cães , Fármacos Gastrointestinais/imunologia , Fármacos Gastrointestinais/farmacocinética , Humanos , Imunossupressores/imunologia , Imunossupressores/farmacocinética , Injeções Intravenosas , Macaca fascicularis , Macaca mulatta , Masculino , Oligodesoxirribonucleotídeos Antissenso/imunologia , Oligodesoxirribonucleotídeos Antissenso/farmacocinética , Oligonucleotídeos Fosforotioatos/imunologia , Oligonucleotídeos Fosforotioatos/farmacocinética , Ligação Proteica , Especificidade da Espécie
11.
PLoS One ; 8(11): e78617, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260121

RESUMO

Mutations and deletions within the human CFHR gene cluster on chromosome 1 are associated with diseases, such as dense deposit disease, CFHR nephropathy or age-related macular degeneration. Resulting mutant CFHR proteins can affect complement regulation. Here we identify human CFHR2 as a novel alternative pathway complement regulator that inhibits the C3 alternative pathway convertase and terminal pathway assembly. CFHR2 is composed of four short consensus repeat domains (SCRs). Two CFHR2 molecules form a dimer through their N-terminal SCRs, and each of the two C-terminal ends can bind C3b. C3b bound CFHR2 still allows C3 convertase formation but the CFHR2 bound convertases do not cleave the substrate C3. Interestingly CFHR2 hardly competes off factor H from C3b. Thus CFHR2 likely acts in concert with factor H, as CFHR2 inhibits convertases while simultaneously allowing factor H assisted degradation by factor I.


Assuntos
Proteínas Inativadoras do Complemento C3b/metabolismo , Via Alternativa do Complemento/fisiologia , Proteólise , Complemento C3/química , Complemento C3/genética , Complemento C3/metabolismo , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento C3b/química , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Fator I do Complemento/química , Fator I do Complemento/genética , Fator I do Complemento/metabolismo , Humanos , Multimerização Proteica/fisiologia , Estrutura Terciária de Proteína , Sequências Repetitivas de Aminoácidos
12.
Curr Mol Med ; 12(8): 1083-97, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22812419

RESUMO

As part of the innate immune system, the complement system recognises a wide range of non-self structures present on pathogens or altered self cells. Its activation elicits proteolytic cascades which eventually results in the cleavage of the C5 protein into two fragments, C5a and C5b. The small anaphylatoxin C5a induces a variety of biological responses upon binding to the 7TM receptors C5aR and the C5L2, while the large C5b fragment nucleates formation of the membrane attack complex capable of killing susceptible pathogens by the formation of a pore structure in association with complement components C6, C7, C8, and C9. A number of regulatory molecules help to control C5 mediated immune responses towards host cells, but in several major inflammatory conditions including sepsis and arthritis, C5a is believed to contribute significantly to disease etiology. Inhibition of membrane attack complex assembly is already approved for treatment of paroxysmal nocturnal haemoglobinuria and atypical hemolytic uremic syndrome. A number of recent crystal structures have provided a comprehensive insight into the architecture and properties of intact C5 and its fragments, and how pathogens interfere with their function. Here we review the functional and structural aspects of C5 and its fragments, the pathological conditions associated with them, and strategies employed by pathogens to interfere with the biological function of C5. Structural insight and elucidation of evasion strategies employed by pathogens present a unique opportunity for promoting the development of novel selective C5 inhibitors with therapeutic applications.


Assuntos
Complemento C5/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/fisiologia , Complemento C5/genética , Complemento C5/metabolismo , Complemento C5/fisiologia , Humanos , Inflamação/genética , Cinética , Modelos Moleculares , Mutação , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/fisiologia , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
13.
Curr Opin Struct Biol ; 22(3): 333-41, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22560446

RESUMO

Modular serine proteases are central to the complement cascade of the mammalian humoral immune system. These proteases form protein complexes through multi-domain interactions to achieve their proteolytic activity. We review the structural insights into complement initiation by auto-activation of the hetero-tetrameric proteases of the large danger-recognition protein complexes, amplification and labelling of particles by the formation and activity of C3 convertases, and regulation by convertase dissociation and degradation to prevent 'bystander' damage to healthy host cells and tissues. The data reveal that complex formation and large domain-domain rearrangements underlie the proteolytic reactions of the complement cascade, which enables the host to recognize and clear invading microbes and host debris from its blood and fluids surrounding tissues.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Serina Proteases/metabolismo , Animais , Ativação do Complemento/fisiologia , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/metabolismo , Proteínas do Sistema Complemento/química , Ativação Enzimática , Humanos , Modelos Biológicos , Modelos Moleculares , Conformação Proteica , Serina Proteases/química
14.
J Biol Chem ; 287(11): 8092-100, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22267731

RESUMO

PRELP is a 58-kDa proteoglycan found in a variety of extracellular matrices, including cartilage and at several basement membranes. In rheumatoid arthritis (RA), the cartilage tissue is destroyed and fragmented molecules, including PRELP, are released into the synovial fluid where they may interact with components of the complement system. In a previous study, PRELP was found to interact with the complement inhibitor C4b-binding protein, which was suggested to locally down-regulate complement activation in joints during RA. Here we show that PRELP directly inhibits all pathways of complement by binding C9 and thereby prevents the formation of the membrane attack complex (MAC). PRELP does not interfere with the interaction between C9 and already formed C5b-8, but inhibits C9 polymerization thereby preventing formation of the lytic pore. The alternative pathway is moreover inhibited already at the level of C3-convertase formation due to an interaction between PRELP and C3. This suggests that PRELP may down-regulate complement attack at basement membranes and on damaged cartilage and therefore limit pathological complement activation in inflammatory disease such as RA. The net outcome of PRELP-mediated complement inhibition will highly depend on the local concentration of other complement modulating molecules as well as on the local concentration of available complement proteins.


Assuntos
Artrite Reumatoide/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Membrana Basal/química , Membrana Basal/metabolismo , Membrana Basal/patologia , Ativação do Complemento/genética , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteína de Ligação ao Complemento C4b/química , Proteína de Ligação ao Complemento C4b/genética , Proteína de Ligação ao Complemento C4b/metabolismo , Complemento C9/química , Complemento C9/genética , Complemento C9/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/química , Complexo de Ataque à Membrana do Sistema Complemento/genética , Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Regulação para Baixo/genética , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Feminino , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Masculino
15.
EMBO J ; 30(3): 606-16, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21217642

RESUMO

Complement acts as a danger-sensing system in the innate immune system, and its activation initiates a strong inflammatory response and cleavage of the proteins C3 and C5 by proteolytic enzymes, the convertases. These contain a non-catalytic substrate contacting subunit (C3b or C4b) in complex with a protease subunit (Bb or C2a). We determined the crystal structures of the C3b homologue cobra venom factor (CVF) in complex with C5, and in complex with C5 and the inhibitor SSL7 at 4.3 Å resolution. The structures reveal a parallel two-point attachment between C5 and CVF, where the presence of SSL7 only slightly affects the C5-CVF interface, explaining the IgA dependence for SSL7-mediated inhibition of C5 cleavage. CVF functions as a relatively rigid binding scaffold inducing a conformational change in C5, which positions its cleavage site in proximity to the serine protease Bb. A general model for substrate recognition by the convertases is presented based on the C5-CVF and C3b-Bb-SCIN structures. Prior knowledge concerning interactions between the endogenous convertases and their substrates is rationalized by this model.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Complemento C5/metabolismo , Venenos Elapídicos/metabolismo , Exotoxinas/metabolismo , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Conformação Proteica , Convertases de Complemento C3-C5/química , Complemento C5/química , Cristalografia , Venenos Elapídicos/química , Exotoxinas/química , Humanos , Complexos Multiproteicos/química
16.
J Immunol ; 184(1): 420-5, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19949103

RESUMO

The human pathogen Staphylococcus aureus produces several complement-evasion molecules that enable the bacterium to withstand the host immune response. The human-specific staphylococcal complement inhibitor (SCIN) blocks the central C3 convertase enzymes that trigger critical complement functions, such as C3b deposition, phagocytosis, and C5a generation. SCIN effectively blocks the conversion of C3 by alternative pathway C3 convertases (C3bBb), but also induces dimerization of these enzymes. In this study, we show that formation of dimeric convertases by SCIN is important for S. aureus immune evasion because it modulates complement recognition by phagocytic receptors. Dimeric, but not monomeric, SCIN convertases showed an impaired binding to complement receptor 1 and the complement receptor of the Ig superfamily. The dimerization site of SCIN is essential for its strong antiphagocytic properties. These studies provide critical insights into the unique immune-evasion strategies used by S. aureus.


Assuntos
Antígenos de Bactérias/imunologia , Convertases de Complemento C3-C5/imunologia , Fagocitose/imunologia , Infecções Estafilocócicas/imunologia , Animais , Antígenos de Bactérias/metabolismo , Células CHO , Complemento C3/imunologia , Complemento C3/metabolismo , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/metabolismo , Cricetinae , Cricetulus , Dimerização , Eletroforese em Gel de Poliacrilamida , Humanos , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/imunologia
17.
EMBO J ; 28(16): 2469-78, 2009 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-19574954

RESUMO

Immune protection by the complement system critically depends on assembly of C3 convertases on the surface of pathogens and altered host cells. These short-lived protease complexes are formed through pro-convertases, which for the alternative pathway consist of the complement component C3b and the pro-enzyme factor B (FB). Here, we present the crystal structure at 2.2-A resolution, small-angle X-ray scattering and electron microscopy (EM) data of the pro-convertase formed by human FB and cobra venom factor (CVF), a potent homologue of C3b that generates more stable convertases. FB is loaded onto CVF through its pro-peptide Ba segment by specific contacts, which explain the specificity for the homologous C3b over the native C3 and inactive products iC3b and C3c. The protease segment Bb binds the carboxy terminus of CVF through the metal-ion dependent adhesion site of the Von Willebrand factor A-type domain. A possible dynamic equilibrium between a 'loading' and 'activation' state of the pro-convertase may explain the observed difference between the crystal structure of CVFB and the EM structure of C3bB. These insights into formation of convertases provide a basis for further development of complement therapeutics.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Fator B do Complemento/química , Fator B do Complemento/metabolismo , Venenos Elapídicos/química , Venenos Elapídicos/metabolismo , Elapidae/metabolismo , Animais , Linhagem Celular , Convertases de Complemento C3-C5/química , Fator B do Complemento/genética , Cristalografia por Raios X , Venenos Elapídicos/genética , Venenos Elapídicos/isolamento & purificação , Ativação Enzimática , Expressão Gênica , Humanos , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína
18.
Nat Immunol ; 10(7): 721-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19503103

RESUMO

Activation of the complement system generates potent chemoattractants and leads to the opsonization of cells for immune clearance. Short-lived protease complexes cleave complement component C3 into anaphylatoxin C3a and opsonin C3b. Here we report the crystal structure of the C3 convertase formed by C3b and the protease fragment Bb, which was stabilized by the bacterial immune-evasion protein SCIN. The data suggest that the proteolytic specificity and activity depend on the formation of dimers of C3 with C3b of the convertase. SCIN blocked the formation of a productive enzyme-substrate complex. Irreversible dissociation of the complex of C3b and Bb is crucial to complement regulation and was determined by slow binding kinetics of the Mg(2+)-adhesion site in Bb. Understanding the mechanistic basis of the central complement-activation step and microbial immune evasion strategies targeting this step will aid in the development of complement therapeutics.


Assuntos
Proteínas de Bactérias/química , C3 Convertase da Via Alternativa do Complemento/química , Proteínas Inativadoras do Complemento/química , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Catálise , Domínio Catalítico , Complemento C3/química , Complemento C3/metabolismo , C3 Convertase da Via Alternativa do Complemento/metabolismo , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/química , Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Via Alternativa do Complemento/imunologia , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Staphylococcus aureus/química , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Especificidade por Substrato , Ressonância de Plasmônio de Superfície
19.
Dev Comp Immunol ; 33(1): 105-16, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18760301

RESUMO

Cobra venom factor (CVF) is a structural and functional analog of complement C3 isolated from cobra venom. Both CVF and C3b can bind factor B and subsequently form the bimolecular C3/C5 convertases CVF,Bb or C3b,Bb, respectively. The two homologous enzymes exhibit several differences of which the difference in physico-chemical stability is most important, allowing continuous activation of C3 and C5 by CVF,Bb, leading to serum complement depletion. Here we describe the detailed functional properties of two hybrid proteins in which the 113 or 315 C-terminal residues of C3 were replaced with corresponding CVF sequences. Both hybrid proteins formed stable convertases that exhibited C3-cleaving activity, although at different rates. Neither convertase cleaved C5. Both convertases showed partial resistance to inactivation by factors H and I, allowing them to deplete complement in human serum. These data demonstrate that functionally important structural differences between CVF and C3 are located in the very C-terminal region of both homologous proteins, and that small substitutions in human C3 with homologous CVF sequence result in C3 derivatives with CVF-like functions. Such hybrid proteins are important tools to study the structure/function relationships in both C3 and CVF, and these "humanized CVF" proteins may become reagents for therapeutic complement depletion.


Assuntos
Complemento C3/química , Venenos Elapídicos/química , Animais , Clonagem Molecular , Complemento C3/genética , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/genética , Fator H do Complemento/química , Venenos Elapídicos/genética , Fibrinogênio/química , Hemólise , Humanos , Modelos Moleculares , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Ovinos
20.
J Biol Chem ; 282(25): 18552-18562, 2007 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-17395591

RESUMO

Focused complement activation on foreign targets depends on regulatory proteins that decay the bimolecular C3 convertases. Although this process is central to complement control, how the convertases engage and disassemble is not established. The second and third complement control protein (CCP) modules of the cell surface regulator, decay-accelerating factor (DAF, CD55), comprise the simplest structure mediating this activity. Positioning the functional effects of 31 substitution mutants of DAF CCP2 to -4 on partial structures was previously reported. In light of the high resolution crystal structure of the DAF four-CCP functional region, we now reexamine the effects of these and 40 additional mutations. Moreover, we map six monoclonal antibody epitopes and overlap their effects with those of the amino acid substitutions. The data indicate that the interaction of DAF with the convertases is mediated predominantly by two patches approximately 13 A apart, one centered around Arg69 and Arg96 on CCP2 and the other around Phe148 and Leu171 on CCP3. These patches on the same face of the adjacent modules bracket an intermodular linker of critical length (16 A.) Although the key DAF residues in these patches are present or there are conservative substitutions in all other C3 convertase regulators that mediate decay acceleration and/or provide factor I-cofactor activity, the linker region is highly conserved only in the former. Intra-CCP regions also differ. Linker region comparisons suggest that the active CCPs of the decay accelerators are extended, whereas those of the cofactors are tilted. Intra-CCP comparisons suggest that the two classes of regulators bind different regions on their respective ligands.


Assuntos
Antígenos CD55/química , Convertases de Complemento C3-C5/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Antígenos CD55/metabolismo , Convertases de Complemento C3-C5/metabolismo , Cristalografia por Raios X , Epitopos/química , Humanos , Leucina/química , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Fenilalanina/química , Ligação Proteica , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...