Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell Microbiol ; 18(4): 536-45, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26418545

RESUMEN

Neutrophils store large quantities of neutrophil serine proteases (NSPs) that contribute, via multiple mechanisms, to antibacterial immune defences. Even though neutrophils are indispensable in fighting Staphylococcus aureus infections, the importance of NSPs in anti-staphylococcal defence is yet unknown. However, the fact that S. aureus produces three highly specific inhibitors for NSPs [the extracellular adherence proteins (EAPs) Eap, EapH1 and EapH2], suggests that these proteases are important for host defences against this bacterium. In this study we demonstrate that NSPs can inactivate secreted virulence factors of S. aureus and that EAP proteins function to prevent this degradation. Specifically, we find that a large group of S. aureus immune-evasion proteins is vulnerable to proteolytic inactivation by NSPs. In most cases, NSP cleavage leads to functional inactivation of virulence proteins. Interestingly, proteins with similar immune-escape functions appeared to have differential cleavage sensitivity towards NSPs. Using targeted mutagenesis and complementation analyses in S. aureus, we demonstrate that all EAP proteins can protect other virulence factors from NSP degradation in complex bacterial supernatants. These findings show that NSPs inactivate S. aureus virulence factors. Moreover, the protection by EAP proteins can explain why this antibacterial function of NSPs was masked in previous studies. Furthermore, our results indicate that therapeutic inactivation of EAP proteins can help to restore the natural host immune defences against S. aureus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Evasión Inmune , Neutrófilos/enzimología , Serina Proteasas/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , Staphylococcus aureus/inmunología , Factores de Virulencia/metabolismo , Células Cultivadas , Humanos , Neutrófilos/inmunología , Proteolisis , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/fisiología
2.
J Neurooncol ; 123(1): 53-63, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25894595

RESUMEN

Formyl peptide receptor 1 (FPR1) activity in U87 glioblastoma (GBM) cells contributes to tumor cell motility. The present study aimed to evaluate the FPR1 expression in human GBM, the possibility to elicit agonist induced FPR1 activation of GBM cells and inhibit this activation with chemotaxis inhibitory protein of Staphylococcus aureus (CHIPS). Immunohistochemistry was used to assess FPR1 expression in GBM patient samples, which was present in all 178 samples. Also FPR1 mRNA levels measured with quantitative PCR, could be detected in all 25 GBM patient samples tested. Activation of FPR1 in U87 cells, as measured by human mitochondrial-derived agonists, increased calcium mobilization, AKT and ERK1/2 phosphorylation, and ligand-induced migration. Inhibition of all responses could be achieved with CHIPS. Eight early passage human Groningen Glioma (GG) cell lines, isolated from primary GBM tissue were screened for the presence of FPR1. FPR1 mRNA and protein expression as well as receptor activation could not be detected in any of these early passage GG cell lines. However FPR1 was present in ex vivo tumors formed by the same GG cell lines after being implanted in mouse brains. FPR1 is highly expressed in human GBM specimens, it can be activated by human mitochondrial-derived agonists in U87 and inhibited with CHIPS. FPR1 cannot be detected in early passage GG cell lines in vitro, however when engrafted in the mouse brain these cells show FPR1 expression. These results suggest a role of the brain microenvironment in FPR1 expression in GBM.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Interleucina-2/fisiología , Receptores de Formil Péptido/metabolismo , Microambiente Tumoral , Animales , Apoptosis , Biomarcadores de Tumor/genética , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Movimiento Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Glioblastoma/genética , Glioblastoma/patología , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Formil Péptido/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
3.
Cell Microbiol ; 15(8): 1427-37, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23470014

RESUMEN

Staphylococcus aureus community-acquired (CA) MRSA strains are highly virulent and can cause infections in otherwise healthy individuals. The most important mechanism of the host for clearing S. aureus is phagocytosis by neutrophils and subsequent killing of the pathogen. Especially CA-MRSA strains are very efficient in circumventing this neutrophil killing. Interestingly, only a relative small number of virulence factors have been associated with CA-MRSA, one of which are the phenol soluble modulins (PSMs). We have recently shown that the PSMs are functionally inhibited by serum lipoproteins, indicating that PSMs may exert their cytolytic function primarily in the intracellular environment. To further investigate the intracellular role of the PSMs we measured the effect of the α-type and ß-type PSMs on neutrophil killing after phagocytosis. Using fluorescently labelled S. aureus, we measured bacterial survival after phagocytosis in a plate reader, which was employed next to flow cytometry and time-lapse microscopy. Phagocytosis of the CA-MRSA strain MW2 by human neutrophils resulted in rapid host cell death. Using mutant strains of MW2, we demonstrated that in the presence of serum, the intracellular expression of only the psmα operon is both necessary and sufficient for both increased neutrophil cell death and increased survival of S. aureus. Our results identify PSMα peptides as prominent contributors to killing of neutrophils after phagocytosis, a finding with major implications for our understanding of S. aureus pathogenesis and strategies for S. aureus vaccine development.


Asunto(s)
Toxinas Bacterianas/metabolismo , Staphylococcus aureus Resistente a Meticilina/metabolismo , Neutrófilos/patología , Fagocitosis/fisiología , Anticuerpos Antiidiotipos/inmunología , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/uso terapéutico , Muerte Celular/fisiología , Células Cultivadas , Humanos , Neutrófilos/fisiología
4.
Br J Cancer ; 108(3): 587-96, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23322202

RESUMEN

BACKGROUND: High-grade astrocytomas are malignant brain tumours that infiltrate the surrounding brain tissue and have a poor prognosis. Activation of formyl peptide receptor (FPR1) on the human astrocytoma cell line U87 promotes cell motility, growth and angiogenesis. We therefore investigated the FPR1 inhibitor, Chemotaxis Inhibitory Protein of S. aureus (CHIPS), as a potential anti-astrocytoma drug. METHODS AND RESULTS: FPR1 expression was studied immunohistochemically in astrocytomas WHO grades I-IV. With intracellular calcium mobilisation and migration assays, human ligands were tested for their ability to activate FPR1 on U87 cells and on a cell line derived from primary astrocytoma grade IV patient material. Thereafter, we selectively inhibited these ligand-induced responses of FPR1 with an anti-inflammatory compound called Chemotaxis Inhibitory Protein of S. aureus (CHIPS). U87 xenografts in NOD-SCID mice served to investigate the effects of CHIPS in vivo. FPR1 was expressed in 29 out of 32 (90%) of all grades of astrocytomas. Two human mitochondrial-derived formylated peptides, formyl-methionil-leucine-lysine-isoleucine-valine (fMLKLIV) and formyl-methionil-methionil-tyrosine-alanine-leucine-phenylalanine (fMMYALF), were potent activators of FPR1 on tumour cells. Ligand-induced responses of FPR1-expressing tumour cells could be inhibited with FPR1 inhibitor CHIPS. Treatment of tumour-bearing mice with CHIPS slightly reduced tumour growth and improved survival as compared to non-treated animals (P=0.0019). CONCLUSION: Targeting FPR1 with CHIPS reduces cell motility and tumour cell activation, and prolongs the survival of tumour-bearing mice. This strategy could be explored in future research to improve treatment results for astrocytoma patients.


Asunto(s)
Astrocitoma/patología , Astrocitoma/prevención & control , Proteínas Bacterianas/farmacología , Neoplasias Encefálicas/prevención & control , Quimiotaxis/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Formil Péptido/antagonistas & inhibidores , Animales , Astrocitoma/metabolismo , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos NOD , Ratones SCID , Clasificación del Tumor , Receptores de Formil Péptido/metabolismo , Células Tumorales Cultivadas
5.
Microbes Infect ; 7(3): 476-84, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15792635

RESUMEN

Recently we described a novel bacteriophage-encoded pathogenicity island in Staphylococcus aureus that harbors a number of virulence factors that are all involved in the evasion of innate immunity. Here we describe a mechanism by which staphylokinase (SAK), frequently present on this pathogenicity island, interferes with innate immune defenses: SAK is anti-opsonic. By activating human plasminogen (PLG) into plasmin (PL) at the bacterial surface, it creates bacterium-bound serine protease activity that leads to degradation of two major opsonins: human immunoglobulin G (IgG) and human C3b. Incubation of opsonized bacteria with PLG and SAK resulted in removal of anti-staphylococcal IgGs and C3b from the bacterial surface. In phagocytosis assays this proved to be a very efficient mechanism to reduce the opsonic activity of human IgG and serum. The fact that SAK activates human PLG at the bacterial surface and removes IgG as well as C3b makes this protein a unique anti-opsonic molecule.


Asunto(s)
Metaloendopeptidasas/fisiología , Proteínas Opsoninas/metabolismo , Staphylococcus aureus/enzimología , Complemento C3b/inmunología , Complemento C3b/metabolismo , Activación Enzimática , Fibrinolisina , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/metabolismo , Proteínas Opsoninas/sangre , Plasminógeno/metabolismo , Unión Proteica , Staphylococcus aureus/patogenicidad
6.
mBio ; 6(3): e00335, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26045537

RESUMEN

UNLABELLED: Although Staphylococcus aureus is best known for infecting humans, bovine-specific strains are a major cause of mastitis in dairy cattle. The bicomponent leukocidin LukMF', exclusively harbored by S. aureus of ruminant origin, is a virulence factor associated with bovine infections. In this study, the molecular basis of the host specificity of LukMF' is elucidated by identification of chemokine receptor CCR1 as its target. Bovine neutrophils, the major effector cells in the defense against staphylococci, express significant cell surface levels of CCR1, whereas human neutrophils do not. This causes the particular susceptibility of bovine neutrophils to pore formation induced by LukMF'. Bovine S. aureus strains produce high levels of LukMF' in vitro. In culture supernatant of the mastitis field isolate S1444, LukMF' was the most important cytotoxic agent for bovine neutrophils. In a fibrin gel matrix, the effects of the in situ secreted toxins on neutrophils migrating toward S. aureus were visualized. Under these physiological ex vivo conditions, bovine S. aureus S1444 efficiently killed approaching neutrophils at a distance through secretion of LukMF'. Altogether, our findings illustrate the coevolution of pathogen and host, provide new targets for therapeutic and vaccine approaches to treat staphylococcal diseases in the cow, and emphasize the importance of staphylococcal toxins in general. IMPORTANCE: This study explains the mechanism of action of LukMF', a bicomponent toxin found in bovine lineages of S. aureus that is associated with mastitis in cattle. At a molecular level, we describe how LukMF' can specifically kill bovine neutrophils. Here, we demonstrate the contribution of toxins in the determination of host specificity and contribute to the understanding of mechanisms of coevolution of pathogen and host. Our study provides new targets that can be used in therapeutic and vaccine approaches to treat staphylococcal diseases in the cow. We also demonstrate the importance of toxins in specific elimination of immune cells, which has broader implications, especially in human infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Leucocidinas/metabolismo , Mastitis Bovina/microbiología , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Receptores CCR1/metabolismo , Staphylococcus aureus/patogenicidad , Animales , Bovinos , Supervivencia Celular/efectos de los fármacos , Staphylococcus aureus/metabolismo
7.
J Mol Med (Berl) ; 90(10): 1109-20, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22714643

RESUMEN

Toll-like receptors (TLRs) are crucial for our host defense against microbial infections. TLR2 is especially important to fight bacterial infections, as it specifically recognizes bacterial lipoproteins of both Gram-positive and Gram-negative origin. Present on a variety of immune cells, TLR2 is critical for host protection against several bacterial infections, including those caused by Staphylococcus aureus. This major human pathogen causes increasing health care problems due to its increased resistance to antibiotics. S. aureus secretes a wide variety of proteins that inhibit innate immune responses. Recently, several staphylococcal superantigen-like proteins (SSLs) have been described to mediate immune evasive properties. Here, we describe that SSL3 specifically binds and inhibits TLR2 activation on human and murine neutrophils and monocytes. Through binding of the extracellular TLR2 domain, SSL3 inhibits IL-8 production by HEK cells expressing TLR1/2 and TLR2/6 dimers, stimulated with their specific ligands. The SSL3-TLR2 interaction is partially glycan dependent as binding of SSL3 to TLR2 is affected upon removal of sialic acid residues. Moreover, the SSL3(R308A) mutant lacking glycan-binding properties shows lower TLR2 inhibition. An SSL3 mutant, lacking the N-terminal 126 amino acids, still retains full TLR2 inhibiting activity. Of other SSLs tested, only SSL4, which shares the highest homology with SSL3, blocks TLR2 activation. SSL3 is the first-described bacterial protein that blocks TLR2 activation through direct extracellular interaction with the receptor. This unique function of SSL3 adds to the arsenal of immune evasive molecules that S. aureus can employ to subvert both innate and adaptive immunity.


Asunto(s)
Antígenos Bacterianos/inmunología , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Receptor Toll-Like 2/metabolismo , Inmunidad Adaptativa , Animales , Antígenos Bacterianos/farmacología , Antígenos CD/metabolismo , Glicosilación , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Unión Proteica , Ácidos Siálicos/metabolismo , Staphylococcus aureus/fisiología , Superantígenos/farmacología , Receptor Toll-Like 2/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
8.
Clin Exp Immunol ; 140(1): 65-72, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15762876

RESUMEN

Neutrophils can be primed by bacterial lipopolysaccharide (LPS) for an enhanced oxidative burst, which is a key element in the pathogenesis of Gram-negative sepsis. Some serum proteins (e.g. lipopolysaccharide-binding protein) avidly bind LPS and markedly enhance receptor binding and cellular activation while other serum factors (lipoproteins, bactericidal/permeability-increasing protein) neutralize LPS and prevent neutrophil activation. In this paper we examined the kinetics of this priming reaction in whole blood. To study the balance between neutrophil activation and LPS neutralization a sensitive chemiluminescence assay was used in a whole blood system. LPS was able to prime neutrophils for enhanced oxidative burst in whole blood with an optimum incubation time of 25 min. However, LPS was neutralized very rapidly with a t(1/2) of 10 min. After 20 min a second priming factor was already generated, which was shown to be monocyte-derived tumour necrosis factor (TNF).


Asunto(s)
Lipopolisacáridos/inmunología , Neutrófilos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Humanos , Receptores de Lipopolisacáridos/inmunología , Mediciones Luminiscentes/métodos , Monocitos/inmunología , Activación Neutrófila/inmunología , Polimixina B/inmunología , Estallido Respiratorio/inmunología , Salmonella typhimurium
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA