Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
EMBO J ; 35(10): 1133-49, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27013439

RESUMO

Regulators of complement activation (RCA) inhibit complement-induced immune responses on healthy host tissues. We present crystal structures of human RCA (MCP, DAF, and CR1) and a smallpox virus homolog (SPICE) bound to complement component C3b. Our structural data reveal that up to four consecutive homologous CCP domains (i-iv), responsible for inhibition, bind in the same orientation and extended arrangement at a shared binding platform on C3b. Large sequence variations in CCP domains explain the diverse C3b-binding patterns, with limited or no contribution of some individual domains, while all regulators show extensive contacts with C3b for the domains at the third site. A variation of ~100° rotation around the longitudinal axis is observed for domains binding at the fourth site on C3b, without affecting the overall binding mode. The data suggest a common evolutionary origin for both inhibitory mechanisms, called decay acceleration and cofactor activity, with variable C3b binding through domains at sites ii, iii, and iv, and provide a framework for understanding RCA disease-related mutations and immune evasion.


Assuntos
Complemento C3b/química , Complemento C3b/metabolismo , Sítios de Ligação , Antígenos CD55/química , Antígenos CD55/metabolismo , Ativação do Complemento , Humanos , Proteína Cofatora de Membrana/química , Proteína Cofatora de Membrana/metabolismo , Domínios Proteicos , Receptores de Complemento 3b/química , Receptores de Complemento 3b/metabolismo , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/metabolismo
2.
J Biol Chem ; 289(1): 450-63, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24214979

RESUMO

To survive and replicate within the human host, malaria parasites must invade erythrocytes. Invasion can be mediated by the P. falciparum reticulocyte-binding homologue protein 4 (PfRh4) on the merozoite surface interacting with complement receptor type 1 (CR1, CD35) on the erythrocyte membrane. The PfRh4 attachment site lies within the three N-terminal complement control protein modules (CCPs 1-3) of CR1, which intriguingly also accommodate binding and regulatory sites for the key complement activation-specific proteolytic products, C3b and C4b. One of these regulatory activities is decay-accelerating activity. Although PfRh4 does not impact C3b/C4b binding, it does inhibit this convertase disassociating capability. Here, we have employed ELISA, co-immunoprecipitation, and surface plasmon resonance to demonstrate that CCP 1 contains all the critical residues for PfRh4 interaction. We fine mapped by homologous substitution mutagenesis the PfRh4-binding site on CCP 1 and visualized it with a solution structure of CCPs 1-3 derived by NMR and small angle x-ray scattering. We cross-validated these results by creating an artificial PfRh4-binding site through substitution of putative PfRh4-interacting residues from CCP 1 into their homologous positions within CCP 8; strikingly, this engineered binding site had an ∼30-fold higher affinity for PfRh4 than the native one in CCP 1. These experiments define a candidate site on CR1 by which P. falciparum merozoites gain access to human erythrocytes in a non-sialic acid-dependent pathway of merozoite invasion.


Assuntos
Proteínas de Membrana/metabolismo , Merozoítos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Receptores de Complemento 3b/metabolismo , Sítios de Ligação , Complemento C3b/química , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C4b/química , Complemento C4b/genética , Complemento C4b/metabolismo , Eritrócitos/química , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Células HEK293 , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Merozoítos/química , Mutagênese , Ressonância Magnética Nuclear Biomolecular , Plasmodium falciparum/química , Plasmodium falciparum/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Receptores de Complemento 3b/química , Receptores de Complemento 3b/genética , Espalhamento a Baixo Ângulo , Ressonância de Plasmônio de Superfície , Difração de Raios X
3.
Blood ; 118(7): 1923-33, 2011 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-21685372

RESUMO

The Plasmodium falciparum adhesin PfRh4 binds to complement receptor type-1 (CR1) on human erythrocytes and mediates a glycophorin-independent invasion pathway. CR1 is a complement regulator and immune-adherence receptor on erythrocytes required for shuttling of C3b/C4b-opsonized particles to liver and spleen for phagocytosis. Using recombinant CR1 constructs, we mapped the recognition site for PfRh4 to complement control protein modules 1 to 3 (CCP1-3) at the membrane-distal amino terminus of CR1. This region of CR1 binds to C4b and C3b and accelerates decay of both classic pathway and alternative pathway C3 and C5 convertases. CCP1-3 competed for PfRh4 binding to erythroid CR1 and inhibited the PfRh4-CR1 invasion pathways across a wide range of P falciparum strains. PfRh4 did not bind significantly to other CR1 constructs, including CCP15-17, which is 85% identical to CCP1-3. PfRh4 binding to CR1 did not affect its C3b/C4b binding capability, and we show evidence for a ternary complex between CCP1-3, C4b, and PfRh4. PfRh4 binding specifically inhibited CR1's convertase decay-accelerating activity, whereas there was no effect on factor H-mediated decay-accelerating activity. These results increase our understanding of the functional implications of CR1 engagement with PfRh4 and highlight the interplay between complement regulation and infection.


Assuntos
Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Malária Falciparum/parasitologia , Proteínas de Membrana/metabolismo , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Receptores de Complemento/metabolismo , Humanos , Ligação Proteica , Proteínas Recombinantes/metabolismo
4.
J Immunol ; 187(1): 424-33, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21642539

RESUMO

The complement system plays a pivotal protective role in the innate immune response to many pathogens including flaviviruses. Flavivirus nonstructural protein 1 (NS1) is a secreted nonstructural glycoprotein that accumulates in plasma to high levels and is displayed on the surface of infected cells but absent from viral particles. Previous work has defined an immune evasion role of flavivirus NS1 in limiting complement activation by forming a complex with C1s and C4 to promote cleavage of C4 to C4b. In this study, we demonstrate a second mechanism, also involving C4 and its active fragment C4b, by which NS1 antagonizes complement activation. Dengue, West Nile, or yellow fever virus NS1 directly associated with C4b binding protein (C4BP), a complement regulatory plasma protein that attenuates the classical and lectin pathways. Soluble NS1 recruited C4BP to inactivate C4b in solution and on the plasma membrane. Mapping studies revealed that the interaction sites of NS1 on C4BP partially overlap with the C4b binding sites. Together, these studies further define the immune evasion potential of NS1 in reducing the functional capacity of C4 in complement activation and control of flavivirus infection.


Assuntos
Ativação do Complemento/imunologia , Flavivirus/imunologia , Antígenos de Histocompatibilidade/imunologia , Proteínas não Estruturais Virais/imunologia , Animais , Linhagem Celular , Complemento C4b/antagonistas & inibidores , Complemento C4b/metabolismo , Proteína de Ligação ao Complemento C4b , Cricetinae , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Flavivirus/patogenicidade , Antígenos de Histocompatibilidade/metabolismo , Humanos , Ligação Proteica/imunologia , Proteínas não Estruturais Virais/metabolismo , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Vírus da Febre Amarela/imunologia , Vírus da Febre Amarela/patogenicidade
5.
J Immunol ; 183(5): 3150-9, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19667083

RESUMO

Although smallpox was eradicated as a global illness more than 30 years ago, variola virus and other related pathogenic poxviruses, such as monkeypox, remain potential bioterrorist weapons or could re-emerge as natural infections. Poxviruses express virulence factors that down-modulate the host's immune system. We previously compared functional profiles of the poxviral complement inhibitors of smallpox, vaccinia, and monkeypox known as SPICE, VCP (or VICE), and MOPICE, respectively. SPICE was the most potent regulator of human complement and attached to cells via glycosaminoglycans. The major goals of the present study were to further characterize the complement regulatory and heparin binding sites of SPICE and to evaluate a mAb that abrogates its function. Using substitution mutagenesis, we established that (1) elimination of the three heparin binding sites severely decreases but does not eliminate glycosaminoglycan binding, (2) there is a hierarchy of activity for heparin binding among the three sites, and (3) complement regulatory sites overlap with each of the three heparin binding motifs. By creating chimeras with interchanges of SPICE and VCP residues, a combination of two SPICE amino acids (H77 plus K120) enhances VCP activity approximately 200-fold. Also, SPICE residue L131 is critical for both complement regulatory function and accounts for the electrophoretic differences between SPICE and VCP. An evolutionary history for these structure-function adaptations of SPICE is proposed. Finally, we identified and characterized a mAb that inhibits the complement regulatory activity of SPICE, MOPICE, and VCP and thus could be used as a therapeutic agent.


Assuntos
Enzimas Ativadoras do Complemento/antagonistas & inibidores , Enzimas Ativadoras do Complemento/metabolismo , Vírus da Varíola/imunologia , Proteínas da Matriz Viral/antagonistas & inibidores , Proteínas da Matriz Viral/fisiologia , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/metabolismo , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Sítios de Ligação de Anticorpos , Células CHO , Enzimas Ativadoras do Complemento/genética , Complemento C3b/metabolismo , Cricetinae , Cricetulus , Glicosaminoglicanos/antagonistas & inibidores , Glicosaminoglicanos/metabolismo , Heparina/metabolismo , Humanos , Hibridomas , Camundongos , Dados de Sequência Molecular , Mutação Puntual , Vírus da Varíola/genética , Vírus da Varíola/patogenicidade , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/genética , Fatores de Virulência/fisiologia
6.
Blood ; 112(13): 4948-52, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18796626

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a disease of complement dysregulation. In approximately 50% of patients, mutations have been described in the genes encoding the complement regulators factor H, MCP, and factor I or the activator factor B. We report here mutations in the central component of the complement cascade, C3, in association with aHUS. We describe 9 novel C3 mutations in 14 aHUS patients with a persistently low serum C3 level. We have demonstrated that 5 of these mutations are gain-of-function and 2 are inactivating. This establishes C3 as a susceptibility factor for aHUS.


Assuntos
Complemento C3/genética , Síndrome Hemolítico-Urêmica/genética , Mutação , Adolescente , Adulto , Criança , Pré-Escolar , Códon sem Sentido , Complemento C3/análise , Análise Mutacional de DNA , Predisposição Genética para Doença , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/imunologia , Heterozigoto , Humanos , Lactente , Mutação de Sentido Incorreto , Adulto Jovem
7.
J Immunol ; 181(6): 4199-207, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768877

RESUMO

Despite eradication of smallpox three decades ago, public health concerns remain due to its potential use as a bioterrorist weapon. Smallpox and other orthopoxviruses express virulence factors that inhibit the host's complement system. In this study, our goals were to characterize the ability of the smallpox inhibitor of complement enzymes, SPICE, to regulate human complement on the cell surface. We demonstrate that SPICE binds to a variety of cell types and that the heparan sulfate and chondroitin sulfate glycosaminoglycans serve as attachment sites. A transmembrane-engineered version as well as soluble recombinant SPICE inhibited complement activation at the C3 convertase step with equal or greater efficiency than that of the related host regulators. Moreover, SPICE attached to glycosaminoglycans was more efficient than transmembrane SPICE. We also demonstrate that this virulence activity of SPICE on cells could be blocked by a mAb to SPICE. These results provide insights related to the complement inhibitory activities of poxviral inhibitors of complement and describe a mAb with therapeutic potential.


Assuntos
Membrana Celular/imunologia , Enzimas Ativadoras do Complemento/antagonistas & inibidores , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento/fisiologia , Vírus da Varíola/imunologia , Proteínas da Matriz Viral/fisiologia , Proteínas Virais/fisiologia , Fatores de Virulência/fisiologia , Ligação Viral , Animais , Células CHO , Membrana Celular/enzimologia , Membrana Celular/genética , Membrana Celular/virologia , Enzimas Ativadoras do Complemento/metabolismo , Ativação do Complemento/genética , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/metabolismo , Convertases de Complemento C3-C5/fisiologia , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Cricetinae , Cricetulus , Glicosaminoglicanos/metabolismo , Células HeLa , Humanos , Ligação Proteica/genética , Ligação Proteica/imunologia , Estrutura Terciária de Proteína/genética , Vírus da Varíola/patogenicidade , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
8.
Mol Immunol ; 45(1): 95-105, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17597211

RESUMO

Recent studies have identified mutations in the complement regulatory gene factor I (CFI) that predispose to atypical hemolytic uremic syndrome (aHUS). CFI is a two-chain serine protease in which the light chain carries the catalytic domain while the heavy chain's function is unclear. It downregulates the alternative and classical complement pathways by cleaving the alpha' chains of C3b and C4b in the presence of cofactor proteins (known as cofactor activity). Many CFI mutations in aHUS result in low CFI levels with a consequent quantitative defect in complement regulation. In others, the mutant protein is present in normal amounts but the presumed functional deficiency has not yet been defined. In this report we examine the nature of the functional defect in aHUS-associated CFI mutations. The I322T, D501N and D506V mutations reside in the serine protease domain of CFI and result in secreted proteins that lack C3b and C4b cofactor activity. The delTTCAC (1446-1450) mutant leads to a protein that is not secreted. The R299W mutant lies in a region of the CFI heavy chain of no known function. Our assessments demonstrate decreased C3b and C4b cofactor activity, providing evidence that this region is important for cofactor activity. In two other heavy chain mutants and one probable polymorphic variant, no functional deficiency was identified. These defective mutant proteins will result in an inability to appropriately control the complement cascade at sites of endothelial cell injury. The excessive complement activation for a given degree of damage may result in generation of a procoagulant state and aHUS.


Assuntos
Fator I do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação/genética , Adulto , Linhagem Celular , Pré-Escolar , Complemento C3b/metabolismo , Complemento C4b/metabolismo , Fator I do Complemento/química , Feminino , Deleção de Genes , Humanos , Lactente , Masculino , Proteínas Mutantes/metabolismo , Polimorfismo Genético , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo
9.
PLoS One ; 11(2): e0149792, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26914463

RESUMO

Chronic activation of the complement system and induced inflammation are associated with neuropathology in Alzheimer's disease (AD). Recent large genome wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) in the C3b/C4b receptor (CR1 or CD35) that are associated with late onset AD. Here, anti-CR1 antibodies (Abs) directed against different epitopes of the receptor, were used to localize CR1 in brain, and relative binding affinities of the CR1 ligands, C1q and C3b, were assessed by ELISA. Most Abs tested stained red blood cells in blood vessels but showed no staining in brain parenchyma. However, two monoclonal anti-CR1 Abs labeled astrocytes in all of the cases tested, and this reactivity was preabsorbed by purified recombinant human CR1. Human brain-derived astrocyte cultures were also reactive with both mAbs. The amount of astrocyte staining varied among the samples, but no consistent difference was conferred by diagnosis or the GWAS-identified SNPs rs4844609 or rs6656401. Plasma levels of soluble CR1 did not correlate with diagnosis but a slight increase was observed with rs4844609 and rs6656401 SNP. There was also a modest but statistically significant increase in relative binding activity of C1q to CR1 with the rs4844609 SNP compared to CR1 without the SNP, and of C3b to CR1 in the CR1 genotypes containing the rs6656401 SNP (also associated with the larger isoform of CR1) regardless of clinical diagnosis. These results suggest that it is unlikely that astrocyte CR1 expression levels or C1q or C3b binding activity are the cause of the GWAS identified association of CR1 variants with AD. Further careful functional studies are needed to determine if the variant-dictated number of CR1 expressed on red blood cells contributes to the role of this receptor in the progression of AD, or if another mechanism is involved.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Regulação da Expressão Gênica , Polimorfismo de Nucleotídeo Único , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/sangue , Astrócitos/metabolismo , Encéfalo/patologia , Complemento C1q/metabolismo , Complemento C3b/metabolismo , Eritrócitos/metabolismo , Feminino , Predisposição Genética para Doença/genética , Humanos , Masculino , Transporte Proteico , Receptores de Complemento 3b/sangue
10.
Mol Immunol ; 69: 62-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26675068

RESUMO

Complement, a major component of innate immunity, presents a rapid and robust defense of the intravascular space. While regulatory proteins protect host cells from complement attack, when these measures fail, unrestrained complement activation may trigger self-tissue injury, leading to pathologic conditions. Of the three complement activation pathways, the alternative pathway (AP) in particular has been implicated in numerous disease and injury states. Consequently, the AP components represent attractive targets for therapeutic intervention. The common hard-bodied ticks from the family Ixodidae derive nourishment from the blood of their mammalian hosts. During its blood meal the tick is exposed to host immune effectors, including the complement system. In defense, the tick produces salivary proteins that can inhibit host immune functions. The Salp20 salivary protein of Ixodes scapularis inhibits the host AP pathway by binding properdin and dissociating C3bBbP, the active C3 convertase. In these studies we examined Salp20 activity in various complement-mediated pathologies. Our results indicate that Salp20 can inhibit AP-dependent pathogenesis in the mouse. Its efficacy may be part in due to synergic effects it provides with the endogenous AP regulator, factor H. While Salp20 itself would be expected to be highly immunogenic and therefore inappropriate for therapeutic use, its emergence speaks for the potential development of a non-immunogenic Salp20 mimic that replicates its anti-properdin activity.


Assuntos
Via Alternativa do Complemento/imunologia , Ixodes/imunologia , Infestações por Carrapato/imunologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas e Peptídeos Salivares/imunologia , Transfecção
11.
PLoS One ; 7(4): e34820, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22506052

RESUMO

Complement receptor-type 1 (CR1, CD35) is the immune-adherence receptor, a complement regulator, and an erythroid receptor for Plasmodium falciparum during merozoite invasion and subsequent rosette formation involving parasitized and non-infected erythrocytes. The non-uniform geographical distribution of Knops blood group CR1 alleles Sl1/2 and McC(a/b) may result from selective pressures exerted by differential exposure to infectious hazards. Here, four variant short recombinant versions of CR1 were produced and analyzed, focusing on complement control protein modules (CCPs) 15-25 of its ectodomain. These eleven modules encompass a region (CCPs 15-17) key to rosetting, opsonin recognition and complement regulation, as well as the Knops blood group polymorphisms in CCPs 24-25. All four CR1 15-25 variants were monomeric and had similar axial ratios. Modules 21 and 22, despite their double-length inter-modular linker, did not lie side-by-side so as to stabilize a bent-back architecture that would facilitate cooperation between key functional modules and Knops blood group antigens. Indeed, the four CR1 15-25 variants had virtually indistinguishable affinities for immobilized complement fragments C3b (K(D) = 0.8-1.1 µM) and C4b (K(D) = 5.0-5.3 µM). They were all equally good co-factors for factor I-catalysed cleavage of C3b and C4b, and they bound equally within a narrow affinity range, to immobilized C1q. No differences between the variants were observed in assays for inhibition of erythrocyte invasion by P. falciparum or for rosette disruption. Neither differences in complement-regulatory functionality, nor interactions with P. falciparum proteins tested here, appear to have driven the non-uniform geographic distribution of these alleles.


Assuntos
Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Malária/genética , Malária/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Alelos , Animais , Complemento C3b/genética , Complemento C3b/metabolismo , Complemento C4b/genética , Complemento C4b/metabolismo , Eritrócitos/metabolismo , Humanos , Malária/sangue , Parasitos/genética , Parasitos/metabolismo , Pichia/genética , Pichia/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Polimorfismo Genético
12.
Virology ; 413(2): 253-64, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21429549

RESUMO

Dengue virus (DENV) NS1 is a versatile non-structural glycoprotein that is secreted as a hexamer, binds to the cell surface of infected and uninfected cells, and has immune evasive functions. DENV NS1 displays two conserved N-linked glycans at N130 and N207. In this study, we examined the role of these two N-linked glycans on NS1 secretion, stability, and function. Because some groups have reported reduced yields of infectious DENV when N130 and N207 are changed, we analyzed glycosylation-deficient NS1 phenotypes using a transgenic expression system. We show that the N-linked glycan at position 130 is required for stabilization of the secreted hexamer whereas the N-linked glycan at residue 207 facilitates secretion and extracellular protein stability. Moreover, NS1 mutants lacking an N-linked glycan at N130 did not interact efficiently with complement components C1s and C4. In summary, our results elucidate the contribution of N-linked glycosylation to the function of DENV NS1.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Vírus da Dengue/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Proteínas não Estruturais Virais/metabolismo , Vírus da Dengue/classificação , Glicosilação , Humanos , Mutação , Polissacarídeos , Sindbis virus , Proteínas não Estruturais Virais/genética , Eliminação de Partículas Virais
13.
mBio ; 2(6)2011.
Artigo em Inglês | MEDLINE | ID: mdl-22167226

RESUMO

Mannose-binding lectin (MBL) is a key soluble pathogen recognition protein of the innate immune system that binds specific mannose-containing glycans on the surfaces of microbial agents and initiates complement activation via the lectin pathway. Prior studies showed that MBL-dependent activation of the complement cascade neutralized insect cell-derived West Nile virus (WNV) in cell culture and restricted pathogenesis in mice. Here, we investigated the antiviral activity of MBL in infection by dengue virus (DENV), a related flavivirus. Using a panel of naïve sera from mouse strains deficient in different complement components, we showed that inhibition of infection by insect cell- and mammalian cell-derived DENV was primarily dependent on the lectin pathway. Human MBL also bound to DENV and neutralized infection of all four DENV serotypes through complement activation-dependent and -independent pathways. Experiments with human serum from naïve individuals with inherent variation in the levels of MBL in blood showed a direct correlation between the concentration of MBL and neutralization of DENV; samples with high levels of MBL in blood neutralized DENV more efficiently than those with lower levels. Our studies suggest that allelic variation of MBL in humans may impact complement-dependent control of DENV pathogenesis. IMPORTANCE Dengue virus (DENV) is a mosquito-transmitted virus that causes a spectrum of clinical disease in humans ranging from subclinical infection to dengue hemorrhagic fever and dengue shock syndrome. Four serotypes of DENV exist, and severe illness is usually associated with secondary infection by a different serotype. Here, we show that mannose-binding lectin (MBL), a pattern recognition molecule that initiates the lectin pathway of complement activation, neutralized infection of all four DENV serotypes through complement activation-dependent and -independent pathways. Moreover, we observed a direct correlation with the concentration of MBL in human serum and neutralization of DENV infection. Our studies suggest that common genetic polymorphisms that result in disparate levels and function of MBL in humans may impact DENV infection, pathogenesis, and disease severity.


Assuntos
Proteínas do Sistema Complemento/imunologia , Vírus da Dengue/imunologia , Lectina de Ligação a Manose/imunologia , Inativação de Vírus , Animais , Ativação do Complemento , Humanos , Camundongos , Testes de Neutralização
14.
J Exp Med ; 207(4): 793-806, 2010 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-20308361

RESUMO

The complement system plays an essential protective role in the initial defense against many microorganisms. Flavivirus NS1 is a secreted nonstructural glycoprotein that accumulates in blood, is displayed on the surface of infected cells, and has been hypothesized to have immune evasion functions. Herein, we demonstrate that dengue virus (DENV), West Nile virus (WNV), and yellow fever virus (YFV) NS1 attenuate classical and lectin pathway activation by directly interacting with C4. Binding of NS1 to C4 reduced C4b deposition and C3 convertase (C4b2a) activity. Although NS1 bound C4b, it lacked intrinsic cofactor activity to degrade C4b, and did not block C3 convertase formation or accelerate decay of the C3 and C5 convertases. Instead, NS1 enhanced C4 cleavage by recruiting and activating the complement-specific protease C1s. By binding C1s and C4 in a complex, NS1 promotes efficient degradation of C4 to C4b. Through this mechanism, NS1 protects DENV from complement-dependent neutralization in solution. These studies define a novel immune evasion mechanism for restricting complement control of microbial infection.


Assuntos
Complemento C4/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Animais , Biocatálise/efeitos dos fármacos , Células CHO , Complemento C1/metabolismo , Proteína Inibidora do Complemento C1/metabolismo , Complemento C1s/agonistas , Complemento C1s/antagonistas & inibidores , Complemento C1s/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/metabolismo , Complemento C4/metabolismo , Complemento C4b/metabolismo , Fator I do Complemento/metabolismo , Ensaio de Atividade Hemolítica de Complemento , Via Clássica do Complemento/efeitos dos fármacos , Via Clássica do Complemento/imunologia , Lectina de Ligação a Manose da Via do Complemento/efeitos dos fármacos , Lectina de Ligação a Manose da Via do Complemento/imunologia , Cricetinae , Cricetulus , Vírus da Dengue/imunologia , Precursores Enzimáticos/metabolismo , Cobaias , Humanos , Cinética , Testes de Neutralização , Ligação Proteica/imunologia , Proteínas não Estruturais Virais/farmacologia
15.
J Immunol ; 176(6): 3725-34, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16517741

RESUMO

The outbreak of monkeypox in the Unites States in the summer of 2003 was the first occurrence of this smallpox-like disease outside of Africa. This limited human epidemic resulted from cross-infection of prairie dogs by imported African rodents. Although there were no human fatalities, this outbreak illustrates that monkeypox is an emerging natural infection and a potential biological weapon. We characterized a virulence factor expressed by monkeypox (monkeypox inhibitor of complement enzymes or MOPICE). We also compared its structure and regulatory function to homologous complement regulatory proteins of variola (SPICE) and vaccinia (VCP). In multiple expression systems, 5-30% of MOPICE, SPICE, and VCP consisted of function-enhancing disulfide-linked homodimers. Mammalian cells infected with vaccinia virus also expressed VCP dimers. MOPICE bound human C3b/C4b intermediate to that of SPICE and VCP. Cofactor activity of MOPICE was similar to VCP, but both were approximately 100-fold less efficient than SPICE. SPICE and VCP, but not MOPICE, possessed decay-accelerating activity for the C3 and C5 convertases of the classical pathway. Additionally, all three regulators possessed heparin-binding capability. These studies demonstrate that MOPICE regulates human complement and suggest that dimerization is a prominent feature of these virulence factors. Thus, our data add novel information relative to the functional repertoire of these poxviral virulence factors. Furthermore, targeting and neutralizing these complement regulatory active sites via mAbs is a therapeutic approach that may enhance protection against smallpox.


Assuntos
Complemento C3b/antagonistas & inibidores , Complemento C4b/antagonistas & inibidores , Monkeypox virus/química , Monkeypox virus/imunologia , Vaccinia virus/química , Vírus da Varíola/química , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , Enzimas Ativadoras do Complemento/metabolismo , Complemento C3b/imunologia , Complemento C3b/metabolismo , Complemento C4b/imunologia , Complemento C4b/metabolismo , Cricetinae , Dimerização , Heparina/metabolismo , Humanos , Dados de Sequência Molecular , Monkeypox virus/genética , Ligação Proteica , Estrutura Quaternária de Proteína , Vaccinia virus/imunologia , Vírus da Varíola/imunologia
16.
J Immunol ; 175(7): 4528-35, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16177096

RESUMO

The extracellular domain of the complement receptor type 1 (CR1; CD35) consists entirely of 30 complement control protein repeats (CCPs). CR1 has two distinct functional sites, site 1 (CCPs 1-3) and two copies of site 2 (CCPs 8-10 and CCPs 15-17). In this report we further define the structural requirements for decay-accelerating activity (DAA) for the classical pathway (CP) C3 and C5 convertases and, using these results, generate more potent decay accelerators. Previously, we demonstrated that both sites 1 and 2, tandemly arranged, are required for efficient DAA for C5 convertases. We show that site 1 dissociates the CP C5 convertase, whereas the role of site 2 is to bind the C3b subunit. The intervening CCPs between two functional sites are required for optimal DAA, suggesting that a spatial orientation of the two sites is important. DAA for the CP C3 convertase is increased synergistically if two copies of site 1, particularly those carrying DAA-increasing mutations, are contained within one protein. DAA in such constructs may exceed that of long homologous repeat A (CCPs 1-7) by up to 58-fold. To explain this synergy, we propose a dimeric structure for the CP C3 convertase on cell surfaces. We also extended our previous studies of the amino acid requirements for DAA of site 1 and found that the CCP 1/CCP 2 junction is critical and that Phe82 may contact the C3 convertases. These observations increase our understanding of the mechanism of DAA. In addition, a more potent decay-accelerating form of CR1 was generated.


Assuntos
Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/fisiologia , Proteínas Inativadoras do Complemento/metabolismo , Via Clássica do Complemento , Receptores de Complemento 3b/fisiologia , Sítios de Ligação , Antígenos CD55/fisiologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/química , Via Clássica do Complemento/genética , Glicina/genética , Glicina/metabolismo , Humanos , Mutação , Fenilalanina/genética , Fenilalanina/metabolismo , Estrutura Terciária de Proteína , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/metabolismo
17.
J Am Chem Soc ; 127(1): 405-11, 2005 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-15631491

RESUMO

This paper presents polychromatic selective polarization inversion (PC-SPI) as an alternative to the polarization transfer methods recently developed for the application of NMR to large biological molecules. Theoretical and numerical considerations indicate that PC-SPI has the potential for more efficient polarization transfer under conditions of rapid transverse relaxation compared to J coupling- and cross-correlated relaxation-based transfers. The main advantage offered by the method presented here is the maintenance of near-optimal trajectories of inversion of the individual components of the spin magnetization while using broadband optimized pulses. A 2D experiment was implemented combining PC-SPI with TROSY-based chemical shift correlation. The experiment was applied to detect (15)N-(1)H chemical shift correlation spectra of a 200 kDa complex consisting of an 80% (2)H- and uniformly (15)N,(13)C-labeled 22 kDa portion of complement receptor type 1 and unlabeled C3b of complement (180 kDa).


Assuntos
Complemento C3b/química , Ressonância Magnética Nuclear Biomolecular/métodos , Receptores de Complemento 3b/química , Sítios de Ligação , Deutério , Modelos Moleculares , Isótopos de Nitrogênio , Fragmentos de Peptídeos/química
18.
Cell ; 108(6): 769-80, 2002 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-11955431

RESUMO

Complement receptor type 1 (CR1 or CD35) is a multiple modular protein that mediates the immune adherence phenomenon, a fundamental event for destroying microbes and initiating an immunological response. It fulfills this role through binding C3b/C4b-opsonized foreign antigens. The structure of the principal C3b/C4b binding site (residues 901-1095) of CR1 is reported, revealing three complement control protein modules (modules 15-17) in an extended head-to-tail arrangement with flexibility at the 16-17 junction. Structure-guided mutagenesis identified a positively charged surface region on module 15 that is critical for C4b binding. This patch, together with basic side chains of module 16 exposed on the same face of CR1, is required for C3b binding. These studies reveal the initial structural details of one of the first receptor-ligand interactions to be identified in immunobiology.


Assuntos
Complemento C3b/metabolismo , Reação de Imunoaderência , Receptores de Complemento 3b/química , Receptores de Complemento 3b/metabolismo , Sítios de Ligação , Humanos , Espectroscopia de Ressonância Magnética , Mutagênese , Estrutura Terciária de Proteína , Receptores de Complemento 3b/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA