Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 209(6): 1146-1155, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36002230

RESUMO

IgG molecules are crucial for the human immune response against bacterial infections. IgGs can trigger phagocytosis by innate immune cells, like neutrophils. To do so, IgGs should bind to the bacterial surface via their variable Fab regions and interact with Fcγ receptors and complement C1 via the constant Fc domain. C1 binding to IgG-labeled bacteria activates the complement cascade, which results in bacterial decoration with C3-derived molecules that are recognized by complement receptors on neutrophils. Next to FcγRs and complement receptors on the membrane, neutrophils also express the intracellular neonatal Fc receptor (FcRn). We previously reported that staphylococcal protein A (SpA), a key immune-evasion protein of Staphylococcus aureus, potently blocks IgG-mediated complement activation and killing of S. aureus by interfering with IgG hexamer formation. SpA is also known to block IgG-mediated phagocytosis in absence of complement, but the mechanism behind it remains unclear. In this study, we demonstrate that SpA blocks IgG-mediated phagocytosis and killing of S. aureus and that it inhibits the interaction of IgGs with FcγRs (FcγRIIa and FcγRIIIb, but not FcγRI) and FcRn. Furthermore, our data show that multiple SpA domains are needed to effectively block IgG1-mediated phagocytosis. This provides a rationale for the fact that SpA from S. aureus contains four to five repeats. Taken together, our study elucidates the molecular mechanism by which SpA blocks IgG-mediated phagocytosis and supports the idea that in addition to FcγRs, the intracellular FcRn is also prevented from binding IgG by SpA.


Assuntos
Imunoglobulina G , Fagocitose , Receptores de IgG , Proteína Estafilocócica A , Staphylococcus aureus , Complemento C1 , Humanos , Imunoglobulina G/imunologia , Receptores de Complemento , Receptores de IgG/metabolismo , Proteína Estafilocócica A/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33563762

RESUMO

Immunoglobulin (Ig) G molecules are essential players in the human immune response against bacterial infections. An important effector of IgG-dependent immunity is the induction of complement activation, a reaction that triggers a variety of responses that help kill bacteria. Antibody-dependent complement activation is promoted by the organization of target-bound IgGs into hexamers that are held together via noncovalent Fc-Fc interactions. Here we show that staphylococcal protein A (SpA), an important virulence factor and vaccine candidate of Staphylococcus aureus, effectively blocks IgG hexamerization and subsequent complement activation. Using native mass spectrometry and high-speed atomic force microscopy, we demonstrate that SpA blocks IgG hexamerization through competitive binding to the Fc-Fc interaction interface on IgG monomers. In concordance, we show that SpA interferes with the formation of (IgG)6:C1q complexes and prevents downstream complement activation on the surface of S. aureus. Finally, we demonstrate that IgG3 antibodies against S. aureus can potently induce complement activation and opsonophagocytic killing even in the presence of SpA. Together, our findings identify SpA as an immune evasion protein that specifically blocks IgG hexamerization.


Assuntos
Ativação do Complemento , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/metabolismo , Multimerização Proteica , Proteína Estafilocócica A/metabolismo , Sítios de Ligação , Células Cultivadas , Humanos , Fagócitos/imunologia , Fagocitose , Ligação Proteica , Staphylococcus aureus/imunologia
3.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34155115

RESUMO

Complement is an important effector mechanism for antibody-mediated clearance of infections and tumor cells. Upon binding to target cells, the antibody's constant (Fc) domain recruits complement component C1 to initiate a proteolytic cascade that generates lytic pores and stimulates phagocytosis. The C1 complex (C1qr2s2) consists of the large recognition protein C1q and a heterotetramer of proteases C1r and C1s (C1r2s2). While interactions between C1 and IgG-Fc are believed to be mediated by the globular heads of C1q, we here find that C1r2s2 proteases affect the capacity of C1q to form an avid complex with surface-bound IgG molecules (on various 2,4-dinitrophenol [DNP]-coated surfaces and pathogenic Staphylococcus aureus). The extent to which C1r2s2 contributes to C1q-IgG stability strongly differs between human IgG subclasses. Using antibody engineering of monoclonal IgG, we reveal that hexamer-enhancing mutations improve C1q-IgG stability, both in the absence and presence of C1r2s2 In addition, hexamer-enhanced IgGs targeting S. aureus mediate improved complement-dependent phagocytosis by human neutrophils. Altogether, these molecular insights into complement binding to surface-bound IgGs could be important for optimal design of antibody therapies.


Assuntos
Membrana Celular/metabolismo , Complemento C1q/metabolismo , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Imunoglobulina G/metabolismo , Ativação do Complemento , Humanos , Microscopia de Força Atômica , Mutação/genética , Fagocitose , Ligação Proteica , Multimerização Proteica , Estabilidade Proteica , Staphylococcus aureus/imunologia
4.
J Immunol ; 204(4): 954-966, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31915259

RESUMO

Neutrophils are critical to the generation of effective immune responses and for killing invading microbes. Paired immune receptors provide important mechanisms to modulate neutrophil activation thresholds and effector functions. Expression of the leukocyte Ig-like receptor (LILR)A6 (ILT8/CD85b) and LILRB3 (ILT5/CD85a) paired-receptor system on human neutrophils has remained unclear because of the lack of specific molecular tools. Additionally, there is little known of their possible functions in neutrophil biology. The objective of this study was to characterize expression of LILRA6/LILRB3 receptors during human neutrophil differentiation and activation, and to assess their roles in modulating Fc receptor-mediated effector functions. LILRB3, but not LILRA6, was detected in human neutrophil lysates following immunoprecipitation by mass spectrometry. We demonstrate high LILRB3 expression on the surface of resting neutrophils and release from the surface following neutrophil activation. Surface expression was recapitulated in a human PLB-985 cell model of neutrophil-like differentiation. Continuous ligation of LILRB3 inhibited key IgA-mediated effector functions, including production of reactive oxygen species, phagocytic uptake, and microbial killing. This suggests that LILRB3 provides an important checkpoint to control human neutrophil activation and their antimicrobial effector functions during resting and early-activation stages of the neutrophil life cycle.


Assuntos
Antígenos CD/metabolismo , Neutrófilos/imunologia , Receptores Fc/metabolismo , Receptores Imunológicos/metabolismo , Infecções Estafilocócicas/imunologia , Antígenos CD/genética , Antígenos CD/isolamento & purificação , Diferenciação Celular/imunologia , Linhagem Celular , Regulação para Baixo/imunologia , Humanos , Ativação de Neutrófilo , Neutrófilos/metabolismo , Fagocitose , Cultura Primária de Células , Espécies Reativas de Oxigênio/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/isolamento & purificação , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus capitis/imunologia
5.
FASEB J ; 33(3): 3807-3824, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30509126

RESUMO

Staphylococcus aureus Panton-Valentine leukocidin is a pore-forming toxin targeting the human C5a receptor (hC5aR), enabling this pathogen to battle the immune response by destroying phagocytes through targeted lysis. The mechanisms that contribute to rapid cell lysis are largely unexplored. Here, we show that cell lysis may be enabled by a process of toxins targeting receptor clusters and present indirect evidence for receptor "recycling" that allows multiple toxin pores to be formed close together. With the use of live cell single-molecule super-resolution imaging, Förster resonance energy transfer and nanoscale total internal reflection fluorescence colocalization microscopy, we visualized toxin pore formation in the presence of its natural docking ligand. We demonstrate disassociation of hC5aR from toxin complexes and simultaneous binding of new ligands. This effect may free mobile receptors to amplify hyperinflammatory reactions in early stages of microbial infections and have implications for several other similar bicomponent toxins and the design of new antibiotics.-Haapasalo, K., Wollman, A. J. M., de Haas, C. J. C., van Kessel, K. P. M., van Strijp, J. A. G., Leake, M. C. Staphylococcus aureus toxin LukSF dissociates from its membrane receptor target to enable renewed ligand sequestration.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Leucocidinas/metabolismo , Receptores de Superfície Celular/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Linhagem Celular , Humanos , Ligantes , Fagócitos , Receptor da Anafilatoxina C5a/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(35): 9439-9444, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28808028

RESUMO

Staphylococcus aureus is highly adapted to its host and has evolved many strategies to resist opsonization and phagocytosis. Even after uptake by neutrophils, S. aureus shows resistance to killing, which suggests the presence of phagosomal immune evasion molecules. With the aid of secretome phage display, we identified a highly conserved protein that specifically binds and inhibits human myeloperoxidase (MPO), a major player in the oxidative defense of neutrophils. We have named this protein "staphylococcal peroxidase inhibitor" (SPIN). To gain insight into inhibition of MPO by SPIN, we solved the cocrystal structure of SPIN bound to a recombinant form of human MPO at 2.4-Å resolution. This structure reveals that SPIN acts as a molecular plug that prevents H2O2 substrate access to the MPO active site. In subsequent experiments, we observed that SPIN expression increases inside the neutrophil phagosome, where MPO is located, compared with outside the neutrophil. Moreover, bacteria with a deleted gene encoding SPIN showed decreased survival compared with WT bacteria after phagocytosis by neutrophils. Taken together, our results demonstrate that S. aureus secretes a unique proteinaceous MPO inhibitor to enhance survival by interfering with MPO-mediated killing.


Assuntos
Peroxidase/antagonistas & inibidores , Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica , Humanos , Modelos Moleculares , Neutrófilos/fisiologia , Fagocitose , Ligação Proteica , Conformação Proteica , Staphylococcus aureus/metabolismo , Regulação para Cima
8.
J Biol Chem ; 293(7): 2260-2271, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29306874

RESUMO

The heme-containing enzyme myeloperoxidase (MPO) is critical for optimal antimicrobial activity of human neutrophils. We recently discovered that the bacterium Staphylococcus aureus expresses a novel immune evasion protein, called SPIN, that binds tightly to MPO, inhibits MPO activity, and contributes to bacterial survival following phagocytosis. A co-crystal structure of SPIN bound to MPO suggested that SPIN blocks substrate access to the catalytic heme by inserting an N-terminal ß-hairpin into the MPO active-site channel. Here, we describe a series of experiments that more completely define the structure/function relationships of SPIN. Whereas the SPIN N terminus adopts a ß-hairpin confirmation upon binding to MPO, the solution NMR studies presented here are consistent with this region of SPIN being dynamically structured in the unbound state. Curiously, whereas the N-terminal ß-hairpin of SPIN accounts for ∼55% of the buried surface area in the SPIN-MPO complex, its deletion did not significantly change the affinity of SPIN for MPO but did eliminate the ability of SPIN to inhibit MPO. The flexible nature of the SPIN N terminus rendered it susceptible to proteolytic degradation by a series of chymotrypsin-like proteases found within neutrophil granules, thereby abrogating SPIN activity. Degradation of SPIN was prevented by the S. aureus immune evasion protein Eap, which acts as a selective inhibitor of neutrophil serine proteases. Together, these studies provide insight into MPO inhibition by SPIN and suggest possible functional synergy between two distinct classes of S. aureus immune evasion proteins.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Peroxidase/química , Peroxidase/metabolismo , Infecções Estafilocócicas/enzimologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/genética , Cristalografia por Raios X , Humanos , Espectroscopia de Ressonância Magnética , Peroxidase/genética , Ligação Proteica , Staphylococcus aureus/química , Staphylococcus aureus/genética
9.
Cell Microbiol ; 20(11): e12941, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30098280

RESUMO

Staphylococcal superantigen-like (SSL) proteins, one of the major virulence factor families produced by Staphylococcus aureus, were previously demonstrated to be immune evasion molecules that interfere with a variety of innate immune defences. However, in contrast to characterised SSLs, which inhibit immune functions, we show that SSL13 is a strong activator of neutrophils via the formyl peptide receptor 2 (FPR2). Moreover, our data show that SSL13 acts as a chemoattractant and induces degranulation and oxidative burst in neutrophils. As with many other staphylococcal immune evasion proteins, SSL13 shows a high degree of human specificity. SSL13 is not able to efficiently activate mouse neutrophils, hampering in vivo experiments. In conclusion, SSL13 is a neutrophil chemoattractant and activator that acts via FPR2. Therefore, SSL13 is a unique SSL member that does not belong to the immune evasion class but is a pathogen alarming molecule. Our study provides a new concept of SSLs; SSLs not only inhibit host immune processes but also recruit human neutrophils to the site of infection. This new insight allows us to better understand complex interactions between host and S. aureus pathological processes.


Assuntos
Proteínas de Bactérias/metabolismo , Neutrófilos/microbiologia , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Staphylococcus aureus/patogenicidade , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Degranulação Celular , Fatores Quimiotáticos/metabolismo , Feminino , Células HL-60 , Humanos , Evasão da Resposta Imune , Camundongos Endogâmicos , Ativação de Neutrófilo , Neutrófilos/fisiologia , Peritonite/metabolismo , Peritonite/microbiologia , Explosão Respiratória , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia
10.
Arch Biochem Biophys ; 645: 1-11, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29524428

RESUMO

Staphylococcus aureus and related species are highly adapted to their hosts and have evolved numerous strategies to evade the immune system. S. aureus shows resistance to killing following uptake into the phagosome, which suggests that the bacterium evades intracellular killing mechanisms used by neutrophils. We recently discovered an S. aureus protein (SPIN for Staphylococcal Peroxidase INhibitor) that binds to and inhibits myeloperoxidase (MPO), a major player in the oxidative defense of neutrophils. To allow for comparative studies between multiple SPIN sequences, we identified a panel of homologs from species closely related to S. aureus. Characterization of these proteins revealed that SPIN molecules from S. agnetis, S. delphini, S. schleiferi, and S. intermedius all bind human MPO with nanomolar affinities, and that those from S. delphini, S. schleiferi, and S. intermedius inhibit human MPO in a dose-dependent manner. A 2.4 Šresolution co-crystal structure of SPIN-delphini bound to recombinant human MPO allowed us to identify conserved structural features of SPIN proteins, and to propose sequence-dependent physical explanations for why SPIN-aureus binds human MPO with higher affinity than SPIN-delphini. Together, these studies expand our understanding of MPO binding and inhibition by a recently identified component of the staphylococcal innate immune evasion arsenal.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Peroxidase/antagonistas & inibidores , Staphylococcus/química , Sequência de Aminoácidos , Inibidores Enzimáticos/metabolismo , Humanos , Modelos Moleculares , Peroxidase/química , Peroxidase/metabolismo , Conformação Proteica
11.
Proc Natl Acad Sci U S A ; 112(35): 11018-23, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26283364

RESUMO

Toll-like receptors (TLRs) are crucial in innate recognition of invading micro-organisms and their subsequent clearance. Bacteria are not passive bystanders and have evolved complex evasion mechanisms. Staphylococcus aureus secretes a potent TLR2 antagonist, staphylococcal superantigen-like protein 3 (SSL3), which prevents receptor stimulation by pathogen-associated lipopeptides. Here, we present crystal structures of SSL3 and its complex with TLR2. The structure reveals that formation of the specific inhibitory complex is predominantly mediated by hydrophobic contacts between SSL3 and TLR2 and does not involve interaction of TLR2-glycans with the conserved Lewis(X) binding site of SSL3. In the complex, SSL3 partially covers the entrance to the lipopeptide binding pocket in TLR2, reducing its size by ∼50%. We show that this is sufficient to inhibit binding of agonist Pam2CSK4 effectively, yet allows SSL3 to bind to an already formed TLR2-Pam2CSK4 complex. The binding site of SSL3 overlaps those of TLR2 dimerization partners TLR1 and TLR6 extensively. Combined, our data reveal a robust dual mechanism in which SSL3 interferes with TLR2 activation at two stages: by binding to TLR2, it blocks ligand binding and thus inhibits activation. Second, by interacting with an already formed TLR2-lipopeptide complex, it prevents TLR heterodimerization and downstream signaling.


Assuntos
Endotoxinas/fisiologia , Staphylococcus aureus/fisiologia , Receptor 2 Toll-Like/antagonistas & inibidores , Dimerização , Endotoxinas/química , Endotoxinas/genética , Estrutura Molecular , Mutagênese , Ligação Proteica , Receptor 2 Toll-Like/química
12.
J Immunol ; 195(3): 1034-43, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26091719

RESUMO

Staphylococcus aureus is well adapted to the human host. Evasion of the host phagocyte response is critical for successful infection. The staphylococcal bicomponent pore-forming toxins Panton-Valentine leukocidin LukSF-PV (PVL) and γ-hemolysin CB (HlgCB) target human phagocytes through interaction with the complement receptors C5aR1 and C5aR2. Currently, the apparent redundancy of both toxins cannot be adequately addressed in experimental models of infection because mice are resistant to PVL and HlgCB. The molecular basis for species specificity of the two toxins in animal models is not completely understood. We show that PVL and HlgCB feature distinct activity toward neutrophils of different mammalian species, where activity of PVL is found to be restricted to fewer species than that of HlgCB. Overexpression of various mammalian C5a receptors in HEK cells confirms that cytotoxicity toward neutrophils is driven by species-specific interactions of the toxins with C5aR1. By taking advantage of the species-specific engagement of the toxins with their receptors, we demonstrate that PVL and HlgCB differentially interact with human C5aR1 and C5aR2. In addition, binding studies illustrate that different parts of the receptor are involved in the initial binding of the toxin and the subsequent formation of lytic pores. These findings allow a better understanding of the molecular mechanism of pore formation. Finally, we show that the toxicity of PVL, but not of HlgCB, is neutralized by various C5aR1 antagonists. This study offers directions for the development of improved preclinical models for infection, as well as for the design of drugs antagonizing leukocidin toxicity.


Assuntos
Proteínas de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Exotoxinas/imunologia , Proteínas Hemolisinas/imunologia , Leucocidinas/imunologia , Receptor da Anafilatoxina C5a/imunologia , Receptores de Quimiocinas/imunologia , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Células HEK293 , Humanos , Evasão da Resposta Imune/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Neutrófilos/imunologia , Fagócitos/imunologia , Ligação Proteica , Estrutura Terciária de Proteína , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Receptores de Quimiocinas/antagonistas & inibidores , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/patogenicidade
13.
Proc Natl Acad Sci U S A ; 111(36): 13187-92, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25161283

RESUMO

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.


Assuntos
Neutrófilos/metabolismo , Inibidores de Serina Proteinase/metabolismo , Staphylococcus aureus/metabolismo , Animais , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Biocatálise , Espaço Extracelular/metabolismo , Feminino , Humanos , Elastase de Leucócito/antagonistas & inibidores , Elastase de Leucócito/metabolismo , Camundongos Endogâmicos C57BL , Modelos Moleculares , Infecções Estafilocócicas/patologia
14.
EMBO J ; 31(17): 3607-19, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22850671

RESUMO

The CXC chemokine receptor 2 (CXCR2) on neutrophils, which recognizes chemokines produced at the site of infection, plays an important role in antimicrobial host defenses such as neutrophil activation and chemotaxis. Staphylococcus aureus is a successful human pathogen secreting a number of proteolytic enzymes, but their influence on the host immune system is not well understood. Here, we identify the cysteine protease Staphopain A as a chemokine receptor blocker. Neutrophils treated with Staphopain A are unresponsive to activation by all unique CXCR2 chemokines due to cleavage of the N-terminal domain, which can be neutralized by specific protease inhibitors. Moreover, Staphopain A inhibits neutrophil migration towards CXCR2 chemokines. By comparing a methicillin-resistant S. aureus (MRSA) strain with an isogenic Staphopain A mutant, we demonstrate that Staphopain A is the only secreted protease with activity towards CXCR2. Although the inability to cleave murine CXCR2 limits in-vivo studies, our data indicate that Staphopain A is an important immunomodulatory protein that blocks neutrophil recruitment by specific cleavage of the N-terminal domain of human CXCR2.


Assuntos
Proteínas de Bactérias/imunologia , Cisteína Endopeptidases/imunologia , Neutrófilos/imunologia , Receptores de Interleucina-8B/imunologia , Animais , Células Cultivadas , Quimiotaxia de Leucócito/imunologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ativação de Neutrófilo/imunologia , Infiltração de Neutrófilos/imunologia , Receptores de Interleucina-8B/antagonistas & inibidores , Células U937
15.
Microbiology (Reading) ; 162(7): 1185-1194, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27112346

RESUMO

Staphylococcus aureus has developed many mechanisms to escape from human immune responses. To resist phagocytic clearance, S. aureus expresses a polysaccharide capsule, which effectively masks the bacterial surface and surface-associated proteins, such as opsonins, from recognition by phagocytic cells. Additionally, secretion of the extracellular fibrinogen binding protein (Efb) potently blocks phagocytic uptake of the pathogen. Efb creates a fibrinogen shield surrounding the bacteria by simultaneously binding complement C3b and fibrinogen at the bacterial surface. By means of neutrophil phagocytosis assays with fluorescently labelled encapsulated serotype 5 (CP5) and serotype 8 (CP8) strains we compare the immune-modulating function of these shielding mechanisms. The data indicate that, in highly encapsulated S. aureus strains, the polysaccharide capsule is able to prevent phagocytic uptake at plasma concentrations <10 %, but loses its protective ability at higher concentrations of plasma. Interestingly, Efb shows a strong inhibitory effect on both capsule-negative and encapsulated strains at all tested plasma concentrations. Furthermore, the results suggest that both shielding mechanisms can exist simultaneously and collaborate to provide optimal protection against phagocytosis at a broad range of plasma concentrations. As opsonizing antibodies will be shielded from recognition by either mechanism, incorporating both capsular polysaccharides and Efb in future vaccines could be of great importance.


Assuntos
Cápsulas Bacterianas/metabolismo , Proteínas de Bactérias/metabolismo , Fibrinogênio/metabolismo , Neutrófilos/imunologia , Fagocitose/imunologia , Polissacarídeos Bacterianos/metabolismo , Staphylococcus aureus/imunologia , Anticorpos Antibacterianos/imunologia , Complemento C3b/metabolismo , Corantes Fluorescentes , Humanos , Imunomodulação/imunologia , Microscopia Confocal , Proteínas Opsonizantes/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/metabolismo
16.
Int J Mol Sci ; 17(7)2016 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-27399672

RESUMO

Matrix metalloproteinases (MMPs) are endopeptidases that degrade components of the extracellular matrix, but also modulate inflammation. During bacterial infections, MMPs are important in the recruitment and migration of inflammatory cells. Besides facilitating cell migration by degrading extracellular matrix components, they potentiate the action of several inflammatory molecules, including cytokines, chemokines, and antimicrobial peptides. Staphylococcus aureus secretes an arsenal of immune evasion molecules that interfere with immune cell functioning and hamper proper immune responses. An earlier study identified staphylococcal superantigen-like protein 5 (SSL5) as an MMP9 inhibitor. Since multiple MMPs are involved in neutrophil recruitment, we set up an in-depth search for additional MMP inhibitors by testing a panel of over 70 secreted staphylococcal proteins on the inhibition of the two main neutrophil MMPs: MMP8 (neutrophil collagenase) and MMP9 (neutrophil gelatinase B). We identified SSL1 and SSL5 as potent inhibitors of both neutrophil MMPs and show that they are actually broad range MMP inhibitors. SSL1 and SSL5 prevent MMP-induced cleavage and potentiation of IL-8 and inhibit the migration of neutrophils through collagen. Thus, through MMP-inhibition, SSL1 and SSL5 interfere with neutrophil activation, chemotaxis, and migration, all vital neutrophil functions in bacterial clearance. Studies on MMP-SSL interactions can have therapeutic potential and SSL based derivatives might prove useful in treatment of cancer and destructive inflammatory diseases.


Assuntos
Proteínas de Bactérias/metabolismo , Metaloproteinases da Matriz/metabolismo , Staphylococcus aureus/metabolismo , Proteínas de Bactérias/farmacologia , Movimento Celular/efeitos dos fármacos , Quimiotaxia , Ensaio de Imunoadsorção Enzimática , Humanos , Imunidade Inata/efeitos dos fármacos , Inibidores de Metaloproteinases de Matriz/farmacologia , Metaloproteinases da Matriz/química , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Ligação Proteica , Células U937
17.
PLoS Pathog ; 9(12): e1003816, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24348255

RESUMO

Upon contact with human plasma, bacteria are rapidly recognized by the complement system that labels their surface for uptake and clearance by phagocytic cells. Staphylococcus aureus secretes the 16 kD Extracellular fibrinogen binding protein (Efb) that binds two different plasma proteins using separate domains: the Efb N-terminus binds to fibrinogen, while the C-terminus binds complement C3. In this study, we show that Efb blocks phagocytosis of S. aureus by human neutrophils. In vitro, we demonstrate that Efb blocks phagocytosis in plasma and in human whole blood. Using a mouse peritonitis model we show that Efb effectively blocks phagocytosis in vivo, either as a purified protein or when produced endogenously by S. aureus. Mutational analysis revealed that Efb requires both its fibrinogen and complement binding residues for phagocytic escape. Using confocal and transmission electron microscopy we show that Efb attracts fibrinogen to the surface of complement-labeled S. aureus generating a 'capsule'-like shield. This thick layer of fibrinogen shields both surface-bound C3b and antibodies from recognition by phagocytic receptors. This information is critical for future vaccination attempts, since opsonizing antibodies may not function in the presence of Efb. Altogether we discover that Efb from S. aureus uniquely escapes phagocytosis by forming a bridge between a complement and coagulation protein.


Assuntos
Proteínas de Bactérias/metabolismo , Complemento C3b/metabolismo , Fibrinogênio/metabolismo , Evasão da Resposta Imune , Fagocitose/imunologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Animais , Fatores de Coagulação Sanguínea/metabolismo , Células Cultivadas , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo
18.
Int J Med Microbiol ; 305(1): 55-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25466204

RESUMO

Due to substantial therapy failure and the emergence of antibiotic-resistant Staphylococcus aureus strains, alternatives for antibiotic treatment of S. aureus infections are urgently needed. Passive immunization using S. aureus-specific monoclonal antibodies (mAb) could be such an alternative to prevent and treat severe S. aureus infections. The invariantly expressed immunodominant staphylococcal antigen A (IsaA) is a promising target for passive immunization. Here we report the development of the human anti-IsaA IgG1 mAb 1D9, which was shown to bind to all 26 S. aureus isolates tested. These included both methicillin-susceptible and methicillin-resistant S. aureus (MSSA and MRSA, respectively). Immune complexes consisting of IsaA and 1D9 stimulated human as well as murine neutrophils to generate an oxidative burst. In a murine bacteremia model, the prophylactic treatment with a single dose of 5 mg/kg 1D9 improved the survival of mice challenged with S. aureus isolate P (MSSA) significantly, while therapeutic treatment with the same dose did not influence animal survival. Neither prophylactic nor therapeutic treatment with 5 mg/kg 1D9 resulted in improved survival of mice with S. aureus USA300 (MRSA) bacteremia. Importantly, our studies show that healthy S. aureus carriers elicit an immune response which is sufficient to generate protective mAbs against invariant staphylococcal surface antigens. Human mAb 1D9, possibly conjugated to for example another antibody, antibiotics, cytokines or chemokines, may be valuable to fight S. aureus infections in patients.


Assuntos
Anticorpos Antibacterianos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antígenos de Bactérias/metabolismo , Bacteriemia/prevenção & controle , Infecções Estafilocócicas/prevenção & controle , Fatores de Virulência/antagonistas & inibidores , Animais , Antígenos de Bactérias/imunologia , Bacteriemia/microbiologia , Modelos Animais de Doenças , Feminino , Imunização Passiva/métodos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/microbiologia , Análise de Sobrevida , Resultado do Tratamento , Fatores de Virulência/imunologia
19.
J Immunol ; 191(1): 353-62, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23740955

RESUMO

To evade opsonophagocytosis, Staphylococcus aureus secretes various immunomodulatory molecules that interfere with effective opsonization by complement and/or IgG. Immune-evasion molecules targeting the phagocyte receptors for these opsonins have not been described. In this study, we demonstrate that S. aureus escapes from FcγR-mediated immunity by secreting a potent FcγR antagonist, FLIPr, or its homolog FLIPr-like. Both proteins were previously reported to function as formyl peptide receptor inhibitors. Binding of FLIPr was mainly restricted to FcγRII receptors, whereas FLIPr-like bound to different FcγR subclasses, and both competitively blocked IgG-ligand binding. They fully inhibited FcγR-mediated effector functions, including opsonophagocytosis and subsequent intracellular killing of S. aureus by neutrophils and Ab-dependent cellular cytotoxicity of tumor cells by both neutrophils and NK cells. In vivo, treatment of mice with FLIPr-like prevented the development of an immune complex-mediated FcγR-dependent Arthus reaction. This study reveals a novel immune-escape function for S. aureus-secreted proteins that may lead to the development of new therapeutic agents in FcγR-mediated diseases.


Assuntos
Proteínas de Bactérias/fisiologia , Receptores de IgG/antagonistas & inibidores , Staphylococcus aureus/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação de Anticorpos/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Leucemia P388/imunologia , Leucemia P388/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/imunologia , Ligação Proteica/imunologia , Receptores de IgG/química , Receptores de IgG/fisiologia , Homologia de Sequência de Aminoácidos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/patogenicidade
20.
J Exp Med ; 204(10): 2461-71, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17893203

RESUMO

To combat the human immune response, bacteria should be able to divert the effectiveness of the complement system. We identify four potent complement inhibitors in Staphylococcus aureus that are part of a new immune evasion cluster. Two are homologues of the C3 convertase modulator staphylococcal complement inhibitor (SCIN) and function in a similar way as SCIN. Extracellular fibrinogen-binding protein (Efb) and its homologue extracellular complement-binding protein (Ecb) are identified as potent complement evasion molecules, and their inhibitory mechanism was pinpointed to blocking C3b-containing convertases: the alternative pathway C3 convertase C3bBb and the C5 convertases C4b2aC3b and C3b2Bb. The potency of Efb and Ecb to block C5 convertase activity was demonstrated by their ability to block C5a generation and C5a-mediated neutrophil activation in vitro. Further, Ecb blocks C5a-dependent neutrophil recruitment into the peritoneal cavity in a mouse model of immune complex peritonitis. The strong antiinflammatory properties of these novel S. aureus-derived convertase inhibitors make these compounds interesting drug candidates for complement-mediated diseases.


Assuntos
Proteínas de Bactérias/metabolismo , Convertases de Complemento C3-C5/antagonistas & inibidores , Proteínas do Sistema Complemento/metabolismo , Staphylococcus aureus/metabolismo , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Movimento Celular , Convertases de Complemento C3-C5/metabolismo , Proteínas do Sistema Complemento/genética , Enteropeptidase/metabolismo , Humanos , Imunidade Inata/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Neutrófilos/citologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA