Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 40(7): e106103, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522633

RESUMEN

Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed ß protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in ß represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.


Asunto(s)
Adhesinas Bacterianas/química , Antígenos CD/química , Antígeno Carcinoembrionario/química , Moléculas de Adhesión Celular/química , Adhesinas Bacterianas/metabolismo , Animales , Antígenos CD/metabolismo , Sitios de Unión , Células CHO , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Cricetinae , Cricetulus , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Células HeLa , Humanos , Unión Proteica , Streptococcus agalactiae/metabolismo
2.
J Immunol ; 209(6): 1146-1155, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36002230

RESUMEN

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.


Asunto(s)
Inmunoglobulina G , Fagocitosis , Receptores de IgG , Proteína Estafilocócica A , Staphylococcus aureus , Complemento C1 , Humanos , Inmunoglobulina G/inmunología , Receptores de Complemento , Receptores de IgG/metabolismo , Proteína Estafilocócica A/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33563762

RESUMEN

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.


Asunto(s)
Activación de Complemento , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Multimerización de Proteína , Proteína Estafilocócica A/metabolismo , Sitios de Unión , Células Cultivadas , Humanos , Fagocitos/inmunología , Fagocitosis , Unión Proteica , Staphylococcus aureus/inmunología
4.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34155115

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Complemento C1q/metabolismo , Complemento C1r/metabolismo , Complemento C1s/metabolismo , Inmunoglobulina G/metabolismo , Activación de Complemento , Humanos , Microscopía de Fuerza Atómica , Mutación/genética , Fagocitosis , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica , Staphylococcus aureus/inmunología
5.
J Cell Sci ; 134(5)2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33589501

RESUMEN

Staphylococcus aureus infects ∼30% of the human population and causes a spectrum of pathologies ranging from mild skin infections to life-threatening invasive diseases. The strict host specificity of its virulence factors has severely limited the accuracy of in vivo models for the development of vaccines and therapeutics. To resolve this, we generated a humanised zebrafish model and determined that neutrophil-specific expression of the human C5a receptor conferred susceptibility to the S. aureus toxins PVL and HlgCB, leading to reduced neutrophil numbers at the site of infection and increased infection-associated mortality. These results show that humanised zebrafish provide a valuable platform to study the contribution of human-specific S. aureus virulence factors to infection in vivo that could facilitate the development of novel therapeutic approaches and essential vaccines.


Asunto(s)
Staphylococcus aureus , Factores de Virulencia , Animales , Humanos , Receptor de Anafilatoxina C5a/genética , Staphylococcus aureus/genética , Virulencia , Factores de Virulencia/genética , Pez Cebra
6.
FASEB J ; 35(10): e21875, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34533845

RESUMEN

Signal inhibitory receptor on leukocytes-1 (SIRL-1) is a negative regulator of myeloid cell function and dampens antimicrobial responses. We here show that different species of the genus Staphylococcus secrete SIRL-1-engaging factors. By screening a library of single-gene transposon mutants in Staphylococcus aureus, we identified these factors as phenol-soluble modulins (PSMs). PSMs are amphipathic α-helical peptides involved in multiple aspects of staphylococcal virulence and physiology. They are cytotoxic and activate the chemotactic formyl peptide receptor 2 (FPR2) on immune cells. Human cathelicidin LL-37 is also an amphipathic α-helical peptide with antimicrobial and chemotactic activities, structurally and functionally similar to α-type PSMs. We demonstrate that α-type PSMs from multiple staphylococcal species as well as human cathelicidin LL-37 activate SIRL-1, suggesting that SIRL-1 recognizes α-helical peptides with an amphipathic arrangement of hydrophobicity, although we were not able to show direct binding to SIRL-1. Upon rational peptide design, we identified artificial peptides in which the capacity to ligate SIRL-1 is segregated from cytotoxic and FPR2-activating properties, allowing specific engagement of SIRL-1. In conclusion, we propose staphylococcal PSMs and human LL-37 as a potential new class of natural ligands for SIRL-1.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Toxinas Bacterianas/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Sirtuina 1/metabolismo , Staphylococcus aureus/metabolismo , Humanos , Percepción de Quorum , Catelicidinas
7.
J Immunol ; 204(4): 954-966, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31915259

RESUMEN

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.


Asunto(s)
Antígenos CD/metabolismo , Neutrófilos/inmunología , Receptores Fc/metabolismo , Receptores Inmunológicos/metabolismo , Infecciones Estafilocócicas/inmunología , Antígenos CD/genética , Antígenos CD/aislamiento & purificación , Diferenciación Celular/inmunología , Línea Celular , Regulación hacia Abajo/inmunología , Humanos , Activación Neutrófila , Neutrófilos/metabolismo , Fagocitosis , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus capitis/inmunología
8.
Nat Immunol ; 10(7): 721-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19503103

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , C3 Convertasa de la Vía Alternativa del Complemento/química , Proteínas Inactivadoras de Complemento/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Sitios de Unión , Catálisis , Dominio Catalítico , Complemento C3/química , Complemento C3/metabolismo , C3 Convertasa de la Vía Alternativa del Complemento/metabolismo , Convertasas de Complemento C3-C5/química , Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/química , Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento/inmunología , Proteínas Inactivadoras de Complemento/metabolismo , Vía Alternativa del Complemento/inmunología , Cristalografía por Rayos X , Humanos , Cinética , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Staphylococcus aureus/química , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Especificidad por Sustrato , Resonancia por Plasmón de Superficie
9.
PLoS Pathog ; 14(10): e1007348, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30321240

RESUMEN

Human Group IIA secreted phospholipase A2 (hGIIA) is an acute phase protein with bactericidal activity against Gram-positive bacteria. Infection models in hGIIA transgenic mice have suggested the importance of hGIIA as an innate defense mechanism against the human pathogens Group A Streptococcus (GAS) and Group B Streptococcus (GBS). Compared to other Gram-positive bacteria, GAS is remarkably resistant to hGIIA activity. To identify GAS resistance mechanisms, we exposed a highly saturated GAS M1 transposon library to recombinant hGIIA and compared relative mutant abundance with library input through transposon-sequencing (Tn-seq). Based on transposon prevalence in the output library, we identified nine genes, including dltA and lytR, conferring increased hGIIA susceptibility. In addition, seven genes conferred increased hGIIA resistance, which included two genes, gacH and gacI that are located within the Group A Carbohydrate (GAC) gene cluster. Using GAS 5448 wild-type and the isogenic gacI mutant and gacI-complemented strains, we demonstrate that loss of the GAC N-acetylglucosamine (GlcNAc) side chain in the ΔgacI mutant increases hGIIA resistance approximately 10-fold, a phenotype that is conserved across different GAS serotypes. Increased resistance is associated with delayed penetration of hGIIA through the cell wall. Correspondingly, loss of the Lancefield Group B Carbohydrate (GBC) rendered GBS significantly more resistant to hGIIA-mediated killing. This suggests that the streptococcal Lancefield antigens, which are critical determinants for streptococcal physiology and virulence, are required for the bactericidal enzyme hGIIA to exert its bactericidal function.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/metabolismo , Fosfolipasas A2 Grupo II/inmunología , Inmunidad Innata/efectos de los fármacos , Polisacáridos Bacterianos/farmacología , Infecciones Estreptocócicas/microbiología , Streptococcus/inmunología , Actividad Bactericida de la Sangre , Fosfolipasas A2 Grupo II/sangre , Fosfolipasas A2 Grupo II/genética , Interacciones Huésped-Patógeno , Humanos , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/enzimología , Streptococcus/patogenicidad
10.
FASEB J ; 33(3): 3807-3824, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30509126

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Leucocidinas/metabolismo , Receptores de Superficie Celular/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Línea Celular , Humanos , Ligandos , Fagocitos , Receptor de Anafilatoxina C5a/metabolismo
11.
Cell Microbiol ; 21(10): e13072, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31219660

RESUMEN

Staphylococcus aureus is a common skin commensal but is also associated with various skin and soft tissue pathologies. Upon invasion, S. aureus is detected by resident innate immune cells through pattern-recognition receptors (PRRs), although a comprehensive understanding of the specific molecular interactions is lacking. Recently, we demonstrated that the PRR langerin (CD207) on epidermal Langerhans cells senses the conserved ß-1,4-linked N-acetylglucosamine (GlcNAc) modification on S. aureus wall teichoic acid (WTA), thereby increasing skin inflammation. Interestingly, the S. aureus ST395 lineage as well as certain species of coagulase-negative staphylococci (CoNS) produce a structurally different WTA molecule, consisting of poly-glycerolphosphate with α-O-N-acetylgalactosamine (GalNAc) residues, which are attached by the glycosyltransferase TagN. Here, we demonstrate that S. aureus ST395 strains interact with the human Macrophage galactose-type lectin (MGL; CD301) receptor, which is expressed by dendritic cells and macrophages in the dermis. MGL bound S. aureus ST395 in a tagN- and GalNAc-dependent manner but did not interact with different tagN-positive CoNS species. However, heterologous expression of Staphylococcus lugdunensis tagN in S. aureus conferred phage infection and MGL binding, confirming the role of this CoNS enzyme as GalNAc-transferase. Functionally, the detection of GalNAc on S. aureus ST395 WTA by human monocyte-derived dendritic cells significantly enhanced cytokine production. Together, our findings highlight differential recognition of S. aureus glycoprofiles by specific human innate receptors, which may affect downstream adaptive immune responses and pathogen clearance.


Asunto(s)
Pared Celular/metabolismo , Células Dendríticas/inmunología , Glicosiltransferasas/metabolismo , Lectinas Tipo C/inmunología , Staphylococcus aureus/enzimología , Ácidos Teicoicos/química , Acetilgalactosamina/análogos & derivados , Acetilgalactosamina/química , Citocinas/metabolismo , Dermis/inmunología , Dermis/microbiología , Glicerofosfatos/química , Glicosiltransferasas/genética , Interacciones Huésped-Patógeno , Humanos , Macrófagos/inmunología , Mutación , Staphylococcus aureus/química , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Staphylococcus lugdunensis/química , Staphylococcus lugdunensis/enzimología
13.
Proc Natl Acad Sci U S A ; 114(35): 9439-9444, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28808028

RESUMEN

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.


Asunto(s)
Peroxidasa/antagonistas & inhibidores , Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica , Humanos , Modelos Moleculares , Neutrófilos/fisiología , Fagocitosis , Unión Proteica , Conformación Proteica , Staphylococcus aureus/metabolismo , Regulación hacia Arriba
14.
J Biol Chem ; 293(7): 2260-2271, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29306874

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Peroxidasa/química , Peroxidasa/metabolismo , Infecciones Estafilocócicas/enzimología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Cristalografía por Rayos X , Humanos , Espectroscopía de Resonancia Magnética , Peroxidasa/genética , Unión Proteica , Staphylococcus aureus/química , Staphylococcus aureus/genética
15.
J Biol Chem ; 293(12): 4468-4477, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29414776

RESUMEN

Staphylococcus aureus is a versatile pathogen capable of causing a broad range of diseases in many different hosts. S. aureus can adapt to its host through modification of its genome (e.g. by acquisition and exchange of mobile genetic elements that encode host-specific virulence factors). Recently, the prophage φSaeq1 was discovered in S. aureus strains from six different clonal lineages almost exclusively isolated from equids. Within this phage, we discovered a novel variant of staphylococcal complement inhibitor (SCIN), a secreted protein that interferes with activation of the human complement system, an important line of host defense. We here show that this equine variant of SCIN, eqSCIN, is a potent blocker of equine complement system activation and subsequent phagocytosis of bacteria by phagocytes. Mechanistic studies indicate that eqSCIN blocks equine complement activation by specific inhibition of the C3 convertase enzyme (C3bBb). Whereas SCIN-A from human S. aureus isolates exclusively inhibits human complement, eqSCIN represents the first animal-adapted SCIN variant that functions in a broader range of hosts (horses, humans, and pigs). Binding analyses suggest that the human-specific activity of SCIN-A is related to amino acid differences on both sides of the SCIN-C3b interface. These data suggest that modification of this phage-encoded complement inhibitor plays a role in the host adaptation of S. aureus and are important to understand how this pathogen transfers between different hosts.


Asunto(s)
Convertasas de Complemento C3-C5/metabolismo , Complemento C3b/antagonistas & inhibidores , Proteínas Inactivadoras de Complemento/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Factores de Virulencia/metabolismo , Animales , Complemento C3b/metabolismo , Proteínas Inactivadoras de Complemento/química , Hemólisis , Caballos , Especificidad del Huésped , Humanos , Fagocitosis , Unión Proteica , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/aislamiento & purificación , Porcinos , Factores de Virulencia/química
16.
Cell Microbiol ; 20(11): e12941, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30098280

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Neutrófilos/microbiología , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Degranulación de la Célula , Factores Quimiotácticos/metabolismo , Femenino , Células HL-60 , Humanos , Evasión Inmune , Ratones Endogámicos , Activación Neutrófila , Neutrófilos/fisiología , Peritonitis/metabolismo , Peritonitis/microbiología , Estallido Respiratorio , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología
17.
Annu Rev Microbiol ; 67: 629-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23834243

RESUMEN

The pathogen Staphylococcus aureus is well adapted to its human host. Neutrophil-mediated killing is a crucial defense system against S. aureus; however, the pathogen has evolved many strategies to resist killing. We first describe the discrete steps of neutrophil activation and migration to the site of infection and the killing of microbes by neutrophils in general. We then highlight the different approaches utilized by S. aureus to resist the different steps of neutrophil attack. Various molecules are discussed in their evolutionary context. Most of the molecules secreted by S. aureus to combat neutrophil attacks at the site of infection show clear human specificity. Many elements of human neutrophil defenses appear redundant, and so the evasion strategies of staphylococci display redundant functions as well. All efforts by S. aureus to resist neutrophil-mediated killing stress the importance of these mechanisms in the pathophysiology of staphylococcal diseases. However, the highly human-specific nature of most host-pathogen interactions hinders the in vivo establishment of their contribution to staphylococcal pathophysiology.


Asunto(s)
Evasión Inmune , Neutrófilos/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Interacciones Huésped-Patógeno , Humanos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología
18.
Curr Top Microbiol Immunol ; 409: 441-489, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-26919864

RESUMEN

Staphylococcus aureus is a successful human and animal pathogen. Its pathogenicity is linked to its ability to secrete a large amount of virulence factors. These secreted proteins interfere with many critical components of the immune system, both innate and adaptive, and hamper proper immune functioning. In recent years, numerous studies have been conducted in order to understand the molecular mechanism underlying the interaction of evasion molecules with the host immune system. Structural studies have fundamentally contributed to our understanding of the mechanisms of action of the individual factors. Furthermore, such studies revealed one of the most striking characteristics of the secreted immune evasion molecules: their conserved structure. Despite high-sequence variability, most immune evasion molecules belong to a small number of structural categories. Another remarkable characteristic is that S. aureus carries most of these virulence factors on mobile genetic elements (MGE) or ex-MGE in its accessory genome. Coevolution of pathogen and host has resulted in immune evasion molecules with a highly host-specific function and prevalence. In this review, we explore how these shared structures and genomic locations relate to function and host specificity. This is discussed in the context of therapeutic options for these immune evasion molecules in infectious as well as in inflammatory diseases.


Asunto(s)
Evasión Inmune , Animales , Humanos , Infecciones Estafilocócicas , Staphylococcus aureus
19.
Arch Biochem Biophys ; 645: 1-11, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29524428

RESUMEN

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.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Peroxidasa/antagonistas & inhibidores , Staphylococcus/química , Secuencia de Aminoácidos , Inhibidores Enzimáticos/metabolismo , Humanos , Modelos Moleculares , Peroxidasa/química , Peroxidasa/metabolismo , Conformación Proteica
20.
Vet Res ; 49(1): 115, 2018 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-30486901

RESUMEN

Staphylococcus aureus is a versatile opportunistic pathogen, causing disease in human and animal species. Its pathogenicity is linked to the ability of S. aureus to secrete immunomodulatory molecules. These evasion proteins bind to host receptors or their ligands, resulting in inhibitory effects through high affinity protein-protein interactions. Staphylococcal evasion molecules are often species-specific due to differences in host target proteins between species. We recently solved the crystal structure of murine TLR2 in complex with immunomodulatory molecule staphylococcal superantigen-like protein 3 (SSL3), which revealed the essential residues within SSL3 for TLR2 inhibition. In this study we aimed to investigate the molecular basis of the interaction on the TLR2 side. The SSL3 binding region on murine TLR2 was compared to that of other species through sequence alignment and homology modeling, which identified interspecies differences. To examine whether this resulted in altered SSL3 activity on the corresponding TLR2s, bovine, equine, human, and murine TLR2 were stably expressed in HEK293T cells and the ability of SSL3 to inhibit TLR2 was assessed. We found that SSL3 was unable to inhibit bovine TLR2. Subsequent loss and gain of function mutagenesis showed that the lack of inhibition is explained by the absence of two tyrosine residues in bovine TLR2 that play a prominent role in the SSL3-TLR2 interface. We found no evidence for the existence of allelic SSL3 variants that have adapted to the bovine host. Thus, within this paper we reveal the molecular determinants of the TLR2-SSL3 interaction which adds to our understanding of staphylococcal host specificity.


Asunto(s)
Proteínas Bacterianas/farmacología , Superantígenos/farmacología , Receptor Toll-Like 2/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Bovinos , Clonación Molecular , Simulación por Computador , Proteínas de Unión al ADN , Células HEK293 , Caballos , Humanos , Modelos Químicos , Modelos Moleculares , Conformación Proteica , Especificidad de la Especie , Staphylococcus aureus/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA