Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Nat Chem Biol ; 10(11): 950-6, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25282505

RESUMEN

Severe forms of pneumococcal meningitis, bacteraemia and pneumonia result in more than 1 million deaths each year despite the widespread introduction of carbohydrate-protein conjugate vaccines against Streptococcus pneumoniae. Here we describe a new and highly efficient antipneumococcal vaccine design based on synthetic conjugation of S. pneumoniae capsule polysaccharides to the potent lipid antigen α-galactosylceramide, which stimulates invariant natural killer T (iNKT) cells when presented by the nonpolymorphic antigen-presenting molecule CD1d. Mice injected with the new lipid-carbohydrate conjugate vaccine produced high-affinity IgG antibodies specific for pneumococcal polysaccharides. Vaccination stimulated germinal center formation; accumulation of iNKT cells with a T follicular helper cell phenotype; and increased frequency of carbohydrate-specific, long-lived memory B cells and plasmablasts. This new lipid-carbohydrate vaccination strategy induced potent antipolysaccharide immunity that protected against pneumococcal disease in mice and may also prove effective for the design of carbohydrate-based vaccines against other major bacterial pathogens.


Asunto(s)
Carbohidratos/química , Lípidos/química , Vacunas Neumococicas/síntesis química , Vacunas Neumococicas/inmunología , Streptococcus pneumoniae/inmunología , Vacunas Sintéticas/química , Vacunas Sintéticas/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD1d/inmunología , Antígenos CD1d/metabolismo , Linfocitos B/citología , Linfocitos B/inmunología , Carbohidratos/administración & dosificación , Línea Celular , Centro Germinal/citología , Centro Germinal/inmunología , Células HL-60 , Humanos , Inmunización Pasiva , Memoria Inmunológica/inmunología , Cinética , Lípidos/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/química , Factores de Tiempo , Vacunas Sintéticas/administración & dosificación
2.
Antimicrob Agents Chemother ; 57(1): 333-42, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23114780

RESUMEN

Treatment options are limited for implant-associated infections (IAI) that are mainly caused by biofilm-forming staphylococci. We report here on the activity of the serrulatane compound 8-hydroxyserrulat-14-en-19-oic acid (EN4), a diterpene isolated from the Australian plant Eremophila neglecta. EN4 elicited antimicrobial activity toward various Gram-positive bacteria but not to Gram-negative bacteria. It showed a similar bactericidal effect against logarithmic-phase, stationary-phase, and adherent Staphylococcus epidermidis, as well as against methicillin-susceptible and methicillin-resistant S. aureus with MICs of 25 to 50 µg/ml and MBCs of 50 to 100 µg/ml. The bactericidal activity of EN4 was similar against S. epidermidis and its Δica mutant, which is unable to produce polysaccharide intercellular adhesin-mediated biofilm. In time-kill studies, EN4 exhibited a rapid and concentration-dependent killing of staphylococci, reducing bacterial counts by >3 log(10) CFU/ml within 5 min at concentrations of >50 µg/ml. Investigation of the mode of action of EN4 revealed membranolytic properties and a general inhibition of macromolecular biosynthesis, suggesting a multitarget activity. In vitro-tested cytotoxicity on eukaryotic cells was time and concentration dependent in the range of the MBCs. EN4 was then tested in a mouse tissue cage model, where it showed neither bactericidal nor cytotoxic effects, indicating an inhibition of its activity. Inhibition assays revealed that this was caused by interactions with albumin. Overall, these findings suggest that, upon structural changes, EN4 might be a promising pharmacophore for the development of new antimicrobials to treat IAI.


Asunto(s)
Antibacterianos/farmacología , Diterpenos/farmacología , Eremophila (Planta)/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus epidermidis/efectos de los fármacos , Animales , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Supervivencia Celular/efectos de los fármacos , Recuento de Colonia Microbiana , Diterpenos/química , Diterpenos/aislamiento & purificación , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Extractos Vegetales/química , Prótesis e Implantes/microbiología , Infecciones Relacionadas con Prótesis/microbiología , Albúmina Sérica/química , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/crecimiento & desarrollo
3.
J Immunol ; 186(1): 443-52, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21131426

RESUMEN

Staphylococcus aureus infection elicits through its mature lipoproteins an innate immune response by TLR2-MyD88 signaling, which improves bacterial clearing and disease outcome. The role of dendritic cells (DCs) and T cells in this immune activation and the function of T and B cells in defense against S. aureus infection remain unclear. Therefore, we first evaluated DC and T cell activation after infection with S. aureus wild type (WT) and its isogenic mutant, which is deficient in lipoprotein maturation, in vitro. Lipoproteins in viable S. aureus contributed via TLR2-MyD88 to activation of DCs, which promoted the release of IFN-γ and IL-17 in CD4(+) T cells. This strong effect was independent of superantigens and MHC class II. We next evaluated the function of T cells and their cytokines IFN-γ and IL-17 in infection in vivo. Six days after systemic murine infection IFN-γ, IL-17, and IL-10 production in total spleen cells were MyD88-dependent and their levels increased until day 21. The comparison of CD3(-/-), Rag2(-/-), and C57BL/6 mice after infection revealed that IFN-γ and IL-17 originated from T cells and IL-10 originated from innate immune cells. Furthermore, vaccination of mice to activate T and B cells did not improve eradication of S. aureus from organs. In conclusion, S. aureus enhances DC activation via TLR2-MyD88 and thereby promotes T(H)1 and T(H)17 cell differentiation. However, neither T cells and their MyD88-regulated products, IFN-γ and IL-17, nor B cells affected bacterial clearing from organs and disease outcome.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Subgrupos de Linfocitos T/inmunología , Receptor Toll-Like 2/fisiología , Animales , Subgrupos de Linfocitos B/microbiología , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Inmunidad Innata , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-17/biosíntesis , Lipoproteínas/fisiología , Ratones , Transducción de Señal/inmunología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/crecimiento & desarrollo , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/microbiología
4.
J Immunol ; 186(4): 2592-601, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21217012

RESUMEN

During Gram-negative sepsis and endotoxemia, CD14 is essential for the recognition of LPS by the TLR4 complex and subsequent generation of systemic inflammation. However, CD14-independent responses to LPS have been reported in vitro and in vivo in selected tissues including the skin. As the liver is a key target organ for neutrophil sequestration and inflammatory pathology during sepsis and endotoxemia, we investigated the role of CD14 in the recruitment of neutrophils into the liver in a mouse model of endotoxemia. Using dynamic in vivo imaging of the liver, we observed that neutrophil recruitment within the sinusoids and post-sinusoidal venules occurred equivalently between LPS-treated wild-type and CD14-knockout mice. Neutrophil recruitment within the liver was completely independent of CD14 regardless of whether it was expressed on cells of hematopoietic or nonhematopoietic origin or in serum as soluble CD14. Whereas CD14 expression was essential for activation of circulating neutrophils and for the development of LPS-induced systemic inflammation (pulmonary neutrophil sequestration, leukopenia, and increased serum proinflammatory cytokine levels), deficiency of CD14 did not limit the adhesion strength of neutrophils in vitro. Furthermore, wild-type and CD14-knockout mice displayed identical deposition of serum-derived hyaluronan-associated protein within liver sinusoids in response to LPS, indicating that the sinusoid-specific CD44/hyaluronan/serum-derived hyaluronan-associated protein-dependent pathway of neutrophil adhesion is activated independently of CD14. Therefore, the liver microcirculation possesses a unique CD14-independent mechanism of LPS detection and activation of neutrophil recruitment.


Asunto(s)
Endotoxemia/inmunología , Endotoxemia/patología , Receptores de Lipopolisacáridos/fisiología , Circulación Hepática/inmunología , Microcirculación/inmunología , Infiltración Neutrófila/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Modelos Animales de Enfermedad , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotoxemia/sangre , Femenino , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Inmunidad Innata/genética , Mediadores de Inflamación/fisiología , Receptores de Lipopolisacáridos/sangre , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/administración & dosificación , Circulación Hepática/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/genética , Infiltración Neutrófila/genética , Receptores de Quimiocina/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
5.
PLoS Pathog ; 6(3): e1000804, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20300602

RESUMEN

During long-term cystic fibrosis lung infections, Pseudomonas aeruginosa undergoes genetic adaptation resulting in progressively increased persistence and the generation of adaptive colony morphotypes. This includes small colony variants (SCVs), auto-aggregative, hyper-adherent cells whose appearance correlates with poor lung function and persistence of infection. The SCV morphotype is strongly linked to elevated levels of cyclic-di-GMP, a ubiquitous bacterial second messenger that regulates the transition between motile and sessile, cooperative lifestyles. A genetic screen in PA01 for SCV-related loci identified the yfiBNR operon, encoding a tripartite signaling module that regulates c-di-GMP levels in P. aeruginosa. Subsequent analysis determined that YfiN is a membrane-integral diguanylate cyclase whose activity is tightly controlled by YfiR, a small periplasmic protein, and the OmpA/Pal-like outer-membrane lipoprotein YfiB. Exopolysaccharide synthesis was identified as the principal downstream target for YfiBNR, with increased production of Pel and Psl exopolysaccharides responsible for many characteristic SCV behaviors. An yfi-dependent SCV was isolated from the sputum of a CF patient. Consequently, the effect of the SCV morphology on persistence of infection was analyzed in vitro and in vivo using the YfiN-mediated SCV as a representative strain. The SCV strain exhibited strong, exopolysaccharide-dependent resistance to nematode scavenging and macrophage phagocytosis. Furthermore, the SCV strain effectively persisted over many weeks in mouse infection models, despite exhibiting a marked fitness disadvantage in vitro. Exposure to sub-inhibitory concentrations of antibiotics significantly decreased both the number of suppressors arising, and the relative fitness disadvantage of the SCV mutant in vitro, suggesting that the SCV persistence phenotype may play a more important role during antimicrobial chemotherapy. This study establishes YfiBNR as an important player in P. aeruginosa persistence, and implicates a central role for c-di-GMP, and by extension the SCV phenotype in chronic infections.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , GMP Cíclico/análogos & derivados , Proteínas Periplasmáticas/genética , Liasas de Fósforo-Oxígeno/genética , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Animales , Proteínas de la Membrana Bacteriana Externa/metabolismo , Caenorhabditis elegans , Células Cultivadas , GMP Cíclico/metabolismo , Elementos Transponibles de ADN/genética , Proteínas de Escherichia coli , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mutagénesis/fisiología , Operón/genética , Proteínas Periplasmáticas/metabolismo , Fagocitosis/fisiología , Fenotipo , Liasas de Fósforo-Oxígeno/metabolismo , Neumonía Bacteriana/enzimología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Infecciones por Pseudomonas/enzimología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/metabolismo , Sistemas de Mensajero Secundario/fisiología
6.
Antimicrob Agents Chemother ; 55(7): 3510-6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21576433

RESUMEN

Daptomycin (DAP) is bactericidal against methicillin-resistant Staphylococcus aureus (MRSA) in vitro, but it failed to eradicate MRSA in an experimental model of implant-associated infection. We therefore investigated various factors which could explain treatment failure by evaluating DAP activity, including the role of different cell wall components, adherence, biofilm, and calcium ions (Ca(2+)) in vitro and in vivo. In the tissue cage infection model, DAP was active only prophylactically and against low inocula. To identify the mechanisms of treatment failure, the in vitro activity of DAP against planktonic and adherent growing S. aureus and S. epidermidis mutants, differing in their capacity of biofilm formation and adherence, was determined. For planktonic staphylococci, the MIC was 0.625 µg/ml. For adherent staphylococci, DAP reduced biofilms at 30 µg/ml. However, it did not kill adherent bacteria up to 500 µg/ml, independent of biofilm biosynthesis (the ica mutant strain), nuclease (the nuc1/nuc2 mutant strain), LPXTG-anchored adhesin (the srtA mutant strain), autolysin (the atl mutant strain), or alanyl-LTA (the dltA mutant strain). Resistance of adherent staphylococci was not due to mutations of adherent bacteria, since staphylococci became DAP susceptible after detachment. Phenotypic tolerance was not explained by inactivation of DAP or inability of initial Ca(2+)-DAP complex formation. However, the addition of up to 100 mg/liter (2.5 mmol/liter) Ca(2+) gradually improved bactericidal activity toward adherent staphylococci in vitro and increased the prevention rate in the cage model from 40% to 60%. In summary, adherent staphylococci are resistant to DAP killing unless Ca(2+) is supplemented to physiologic concentrations.


Asunto(s)
Daptomicina/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus/efectos de los fármacos , Animales , Biopelículas/efectos de los fármacos , Daptomicina/farmacología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Implantación de Prótesis/efectos adversos , Infecciones Estafilocócicas/etiología , Staphylococcus/patogenicidad
7.
Microbiology (Reading) ; 157(Pt 12): 3458-3468, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21964732

RESUMEN

Staphylococcus epidermidis is an opportunistic bacterium whose infections often involve the formation of a biofilm on implanted biomaterials. In S. epidermidis, the exopolysaccharide facilitating bacterial adherence in a biofilm is polysaccharide intercellular adhesin (PIA), whose synthesis requires the enzymes encoded within the intercellular adhesin operon (icaADBC). In vitro, the formation of S. epidermidis biofilms is enhanced by conditions that repress tricarboxylic acid (TCA) cycle activity, such as growth in a medium containing glucose. In many Gram-positive bacteria, repression of TCA cycle genes in response to glucose is accomplished by catabolite control protein A (CcpA). CcpA is a member of the GalR-LacI repressor family that mediates carbon catabolite repression, leading us to hypothesize that catabolite control of S. epidermidis biofilm formation is indirectly regulated by CcpA-dependent repression of the TCA cycle. To test this hypothesis, ccpA deletion mutants were constructed in strain 1457 and 1457-acnA and the effects on TCA cycle activity, biofilm formation and virulence were assessed. As anticipated, deletion of ccpA derepressed TCA cycle activity and inhibited biofilm formation; however, ccpA deletion had only a modest effect on icaADBC transcription. Surprisingly, deletion of ccpA in strain 1457-acnA, a strain whose TCA cycle is inactive and where icaADBC transcription is derepressed, strongly inhibited icaADBC transcription. These observations demonstrate that CcpA is a positive effector of biofilm formation and icaADBC transcription and a repressor of TCA cycle activity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Ciclo del Ácido Cítrico/genética , Metabolismo Energético , Regulación Bacteriana de la Expresión Génica , Proteínas Represoras/metabolismo , Staphylococcus epidermidis/fisiología , Animales , Proteínas Bacterianas/genética , Femenino , Eliminación de Gen , Humanos , Ratones , Polisacáridos Bacterianos/metabolismo , Proteínas Represoras/genética , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/crecimiento & desarrollo , Staphylococcus epidermidis/metabolismo , Virulencia
8.
J Immunol ; 182(11): 7110-8, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19454708

RESUMEN

Lipoproteins (Lpp) are ligands of TLR2 and signal by the adaptor MyD88. As part of the bacterial cell envelope, Lpp are mainly involved in nutrient acquisition for Staphylococcus aureus. The impact of Lpp on TLR2-MyD88 activation for S. aureus in systemic infection is unknown. S. aureus strain SA113 deficient in the enzyme encoded by the prolipoprotein diacylglyceryl transferase gene (Deltalgt), which attaches the lipid anchor to pro-Lpp, was used to study benefits and costs of Lpp maturation. Lpp in S. aureus induced early and strong cytokines by TLR2-MyD88 signaling in murine peritoneal macrophages. Lpp contributed via TLR2 to pathogenesis of sepsis in C57BL/6 mice with IL-1beta, chemokine-mediated inflammation, and high bacterial numbers. In the absence of MyD88-mediated inflammation, Lpp allowed bacterial clearing from liver devoid of infiltrating cells, but still conferred a strong growth advantage in mice, which was shown to rely on iron uptake and storage in vitro and in vivo. With iron-restricted bacteria, the Lpp-related growth advantage was evident in infection of MyD88(-/-), but not of C57BL/6, mice. On the other hand, iron overload of the host restored the growth deficit of Deltalgt in MyD88(-/-), but not in immunocompetent C57BL/6 mice. These results indicate that iron acquisition is improved by Lpp of S. aureus but is counteracted by inflammation. Thus, lipid anchoring is an evolutionary advantage for S. aureus to retain essential proteins for better survival in infection.


Asunto(s)
Inflamación/microbiología , Hierro/metabolismo , Lipoproteínas/efectos adversos , Infecciones Estafilocócicas/patología , Staphylococcus aureus/fisiología , Receptor Toll-Like 2/metabolismo , Animales , Crecimiento , Mediadores de Inflamación , Sobrecarga de Hierro/etiología , Macrófagos Peritoneales/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Staphylococcus aureus/química
9.
Antimicrob Agents Chemother ; 54(10): 4208-18, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660682

RESUMEN

Prosthetic joint replacements are used increasingly to alleviate pain and improve mobility of the progressively older and more obese population. Implant infection occurs in about 5% of patients and entails significant morbidity and high social costs. It is most often caused by staphylococci, which are introduced perioperatively. They are a source of prolonged seeding and difficult to treat due to antibiotic resistance; therefore, infection prevention by prosthesis coating with nonantibiotic-type anti-infective substances is indicated. A renewed interest in topically used silver has fostered development of silver nanoparticles, which, however, present a potential health hazard. Here we present new silver coordination polymer networks with tailored physical and chemical properties as nanostructured coatings on metallic implant substrates. These compounds exhibited strong biofilm sugar-independent bactericidal activity on in vitro-grown biofilms and prevented murine Staphylococcus epidermidis implant infection in vivo with slow release of silver ions and limited transient leukocyte cytotoxicity. Furthermore, we describe the biochemical and molecular mechanisms of silver ion action by gene screening and by targeting cell metabolism of S. epidermidis at different levels. We demonstrate that silver ions inactivate enzymes by binding sulfhydryl (thiol) groups in amino acids and promote the release of iron with subsequent hydroxyl radical formation by an indirect mechanism likely mediated by reactive oxygen species. This is the first report investigating the global metabolic effects of silver in the context of a therapeutic application. We anticipate that the compounds presented here open a new treatment field with a high medical impact.


Asunto(s)
Antibacterianos/uso terapéutico , Materiales Biocompatibles Revestidos/uso terapéutico , Radical Hidroxilo/metabolismo , Implantes Experimentales , Polímeros/uso terapéutico , Plata/uso terapéutico , Infecciones Estafilocócicas/prevención & control , Animales , Antibacterianos/química , Biopelículas/efectos de los fármacos , Materiales Biocompatibles Revestidos/química , Transporte de Electrón , Femenino , Ratones , Ratones Endogámicos C57BL , Polímeros/química , Plata/química , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/patogenicidad
10.
PLoS Pathog ; 4(9): e1000164, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18818736

RESUMEN

Capnocytophaga canimorsus, a commensal bacterium of the canine oral flora, has been repeatedly isolated since 1976 from severe human infections transmitted by dog bites. Here, we show that C. canimorsus exhibits robust growth when it is in direct contact with mammalian cells, including phagocytes. This property was found to be dependent on a surface-exposed sialidase allowing C. canimorsus to utilize internal aminosugars of glycan chains from host cell glycoproteins. Although sialidase probably evolved to sustain commensalism, by releasing carbohydrates from mucosal surfaces, it also contributed to bacterial persistence in a murine infection model: the wild type, but not the sialidase-deficient mutant, grew and persisted, both when infected singly or in competition. This study reveals an example of pathogenic bacteria feeding on mammalian cells, including phagocytes by deglycosylation of host glycans, and it illustrates how the adaptation of a commensal to its ecological niche in the host, here the dog's oral cavity, contributes to being a potential pathogen.


Asunto(s)
Capnocytophaga/metabolismo , Células Epiteliales/microbiología , Fagocitos/microbiología , Animales , Capnocytophaga/enzimología , Capnocytophaga/aislamiento & purificación , Células Cultivadas , Infecciones por Bacterias Gramnegativas , Humanos , Ratones , Neuraminidasa/metabolismo , Polisacáridos/metabolismo
11.
Int J Med Microbiol ; 300(2-3): 155-60, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19805005

RESUMEN

Staphylococcus aureus expresses about 50 lipoproteins (Lpp), which are lipid-anchored in the membrane. The processing of the precursor to the mature Lpp is catalyzed by the phosphatidyl glycerol diacylglyceryl transferase (Lgt) and the lipoprotein-specific type II signal peptidase (LspA) leading to diacylated Lpp. Possibly another acyltransferase attaches a third fatty acid leading to triacylated Lpp. Lpp function as binding proteins for transport of nutrients across the microbial membrane and are involved in processing of other proteins, but most Lpp remain of predicted or unknown function. The di- or triacylated lipid structure is sensed by host pattern recognition receptor TLR2 and induces innate immune responses in professional and non-professional phagocytes. In the host, maturation of Lpp confers optimal metal ion - particularly iron - acquisition, it enhances staphylococcal invasion and phagocytosis, intracellular survival and persistence of infections. However, the advantages of Lpp maturation are counterbalanced by the capability to induce inflammation. In this review, we summarize the current knowledge about the role of Lpp in iron acquisition and TLR2 recognition in the host and describe the consequences of Lpp maturation for survival of S. aureus in the host.


Asunto(s)
Lipoproteínas/fisiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Animales , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Hierro/metabolismo , Ratones , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/patogenicidad , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo
12.
Antimicrob Agents Chemother ; 53(10): 4159-66, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19620329

RESUMEN

Brominated furanones from marine algae inhibit multicellular behaviors of gram-negative bacteria such as biofilm formation and quorum sensing (QS) without affecting their growth. The interaction of furanone with QS in gram-positive bacteria is unknown. Staphylococci have two QS systems, agr and luxS, which lower biofilm formation by two different pathways, RNAIII upregulation and bacterial detachment, and polysaccharide intercellular adhesin (PIA) reduction, respectively. We synthesized natural furanone compound 2 [(5Z)-4-bromo-5-(bromomethylene)-3-butyl-2(5H)-furanone] from Delisea pulchra and three analogues to investigate their effect on biofilm formation in gram-positive bacteria. Compound 2, but not the analogues, enhanced the biofilms of Staphylococcus epidermidis 1457 and 047 and of S. aureus Newman at concentrations between 1.25 and 20 microM. We show the growth inhibition of S. epidermidis and S. aureus by free furanone and demonstrate bactericidal activity. An induction of biofilm occurred at concentrations of 10 to 20% of the MIC and correlated with an increase in PIA. The biofilm effect was agr independent. It was due to interference with luxS, as shown by reduced luxS expression in the presence of compound 2 and independence of the strong biofilm formation in a luxS mutant upon furanone addition. Poly(l-lysine)-grafted/poly(ethylene glycol)-grafted furanone was ineffective on biofilm and not bactericidal, indicating the necessity for free furanone. Free furanone was similarly toxic for murine fibroblasts as for staphylococci, excluding a therapeutic application of this compound. In summary, we observed a biofilm enhancement by furanone in staphylococci at subinhibitory concentrations, which was manifested by an increase in PIA and dependent on luxS.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Liasas de Carbono-Azufre/metabolismo , Furanos/farmacología , Staphylococcus/efectos de los fármacos , Staphylococcus/metabolismo , Animales , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Liasas de Carbono-Azufre/genética , Línea Celular , Furanos/química , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/genética , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Polisacáridos Bacterianos/metabolismo , Percepción de Quorum/efectos de los fármacos , Staphylococcus/crecimiento & desarrollo
13.
J Neuroimmunol ; 206(1-2): 28-31, 2009 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-19012971

RESUMEN

TLR2 signaling participates in the pathogenesis of pneumococcal meningitis. In infant rats, the TLR2 agonist Pam(3)CysSK(4) was applied intracisternally (0.5 microg in 10 microl saline) alone or after induction of pneumococcal meningitis to investigate the effect of TLR2 activation on cerebrospinal fluid (CSF) inflammation and hippocampal apoptosis. A dose effect of Pam(3)CysSK(4) on apoptosis was investigated by intracisternal application of 0.5 microg in 10 microl saline and 40 microg in 20 microl saline. Pam(3)CysSK(4) neither induced apoptosis in sham-operated mice nor aggravated apoptosis in acute infection. However, Pam(3)CysSK(4) induced pleocytosis, TNF-alpha and MMP-9 in CSF in sham-infection but not during acute meningitis. We conclude that TLR2 signaling triggered by Pam(3)CysSK(4) at a dosage capable to induce a neuroinflammatory response does not induce hippocampal apoptosis in the infant rat model of experimental pneumococcal meningitis.


Asunto(s)
Encéfalo/efectos de los fármacos , Inflamación/etiología , Lipopéptidos/farmacología , Meningitis Neumocócica/complicaciones , Meningitis Neumocócica/patología , Receptor Toll-Like 2/agonistas , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Inflamación/líquido cefalorraquídeo , Inflamación/tratamiento farmacológico , Leucocitos/efectos de los fármacos , Lipopéptidos/uso terapéutico , Metaloproteinasa 9 de la Matriz/líquido cefalorraquídeo , Meningitis Neumocócica/líquido cefalorraquídeo , Meningitis Neumocócica/mortalidad , Distribución Aleatoria , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/líquido cefalorraquídeo
14.
Infect Immun ; 76(11): 5133-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18779343

RESUMEN

The virulence of SCCmec type IV hospital-acquired methicillin-resistant Staphylococcus aureus (MRSA) clinical isolates belonging to the major sequence type 8 (ST8 [Lyon clone]) and to a minor upcoming clone, ST5, was compared with that of methicillin-susceptible S. aureus (MSSA) isolates of matching sequence types. In vitro adhesion to human airway epithelial cells (HAECs) as an indicator of dissemination and mortality in a murine sepsis model as an indicator of virulence were evaluated. Ten MRSA isolates and 8 MSSA isolates of ST8 and 8 MRSA isolates and 8 MSSA isolates of ST5 were characterized with respect to multilocus sequence type; agr, spa, and capsule typing; in vitro doubling time; toxin and adhesin gene profiles; and adherence to HAECs. Adherence was significantly lower in the MRSA ST5 group than in the ST8 groups. Infections with MRSA and MSSA isolates ST8 and ST5 were compared. No change in virulence related to the presence of SCCmec was observed, since ST8 but not ST5 caused a significantly lower mortality in its presence. Despite their similar genetic backgrounds, individual clonal MRSA and MSSA isolates were heterogeneous in adherence and virulence. No one of these specific virulence factors determined in vitro was related to mouse mortality. In conclusion, in a bacteremic model, mortality was dependent on the ST and was differentially modulated by SCCmec; within an ST, clonality was not associated with a homogenous outcome.


Asunto(s)
Adhesión Bacteriana/fisiología , Infección Hospitalaria/microbiología , Resistencia a la Meticilina/fisiología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Animales , Células Clonales , Células Epiteliales/microbiología , Genotipo , Humanos , Pulmón/citología , Pulmón/microbiología , Ratones , Fenotipo , Mucosa Respiratoria/microbiología , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
15.
Int J Med Microbiol ; 298(7-8): 599-605, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18329335

RESUMEN

Alpha-toxin (Hla, encoded by hla) is a major virulence factor of Staphylococcus aureus. The activity of the hla promoter was analyzed using luxABCDE on an integration vector. The phla-lux construct was introduced in S. aureus Newman and its isogenic sae and sigB regulator mutants. Promoter activity was monitored by bioluminescence in vitro and in the murine tissue-cage model. Hla promoter activity could be followed in real time at repeated time points of infection. The activation of hla in the sigB-deficient strain and the repression to background levels in a sae-deficient strain relative to hla expression in the wild type could be demonstrated in vivo. Subinhibitory concentrations of teicoplanin, imipenem and ciprofloxacin enhanced hla promoter activity in vitro whereas clindamycin and rifampicin did not. Our approach proved to be rapid and adequate to study promoter activity in vitro and in vivo under conditions where high bacterial numbers are reached.


Asunto(s)
Toxinas Bacterianas/biosíntesis , Perfilación de la Expresión Génica/métodos , Proteínas Hemolisinas/biosíntesis , Staphylococcus aureus/fisiología , Factores de Virulencia/biosíntesis , Animales , Antibacterianos/metabolismo , Fusión Artificial Génica , Toxinas Bacterianas/genética , Expresión Génica/efectos de los fármacos , Genes Reporteros , Proteínas Hemolisinas/genética , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Regiones Promotoras Genéticas , Staphylococcus aureus/genética , Factores de Virulencia/genética
16.
J Neurosci ; 26(49): 12826-37, 2006 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-17151286

RESUMEN

Reactive gliosis is a prominent feature of neurodegenerative and neuroinflammatory disease in the CNS, yet the stimuli that drive this response are not known. There is growing appreciation that signaling through Toll-like receptors (TLRs), which is key to generating innate responses to infection, may have pathogen-independent roles. We show that TLR2 was selectively upregulated by microglia in the denervated zones of the hippocampus in response to stereotactic transection of axons in the entorhinal cortex. In mice lacking TLR2, there were transient, selective reductions in lesion-induced expression of cytokines and chemokines. Recruitment of T cells, but not macrophages, was delayed in TLR2-deficient mice, as well as in mice lacking TNFR1 (tumor necrosis factor receptor 1). TLR2 deficiency also affected microglial proliferative expansion, whereas all of these events were unaffected in TLR4-mutant mice. Consistent with the fact that responses in knock-out mice had all returned to wild-type levels by 8 d, there was no evidence for effects on neuronal plasticity at 20 d. These results identify a role for TLR2 signaling in the early glial response to brain injury, acting as an innate bridge to neuroinflammation.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Neuronas/metabolismo , Neuronas/patología , Transducción de Señal/fisiología , Receptor Toll-Like 2/fisiología , Animales , Axones/patología , Lesiones Encefálicas/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Transducción de Señal/genética , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
17.
BMC Infect Dis ; 7: 25, 2007 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-17428319

RESUMEN

BACKGROUND: Streptococcus (S.) pneumoniae meningitis has a high lethality despite antibiotic treatment. Inflammation is a major pathogenetic factor, which is unresponsive to antibiotics. Therefore adjunctive therapies with antiinflammatory compounds have been developed. TNF484 is a TNF-alpha converting enzyme (TACE) inhibitor and has been found efficacious in experimental meningitis. Toll-like receptor 2 (TLR2) contributes to host response in pneumococcal meningitis by enhancing bacterial clearing and downmodulating inflammation. In this study, TNF484 was applied in mice, which lacked TLR2 and exhibited a strong meningeal inflammation. METHODS: 103 CFU S. pneumoniae serotype 3 was inoculated subarachnoidally into C57BL/6 wild type (wt) mice or TLR2-/-, CD14-/- and CD14-/-/TLR2-/- mice. Severity of disease and survival was followed over 9 days. Response to antibiotics (80 mg/kg ceftriaxone i.p. for 5 days) and/or TACE inhibitor treatment (1 mg/kg s.c. twice daily for 4 days) was evaluated. Animals were sacrificed after 12, 24, and 48 h for analysis of bacterial load in cerebrospinal fluid (CSF) and brain and for TNF and leukocyte measurements in CSF. RESULTS: TLR2-/- mice were significantly sicker than the other mouse strains 24 h after infection. All knockout mice showed higher disease severity after 48 h and died earlier than wt mice. TNF release into CSF was significantly more elevated in TLR2-/- than in the other strains after 24 h. Brain bacterial numbers were significantly higher in all knockout than wt mice after 24 h. Modulation of outcome by antibiotic and TACE inhibitor treatment was evaluated. With antibiotic therapy all wt, CD14-/- and TLR2-/-/CD14-/- mice, but only 79% of TLR2-/- mice, were rescued. TACE inhibitor treatment alone did not rescue, but prolonged survival in wt mice, and in TLR2-/- and CD14-/- mice to the values observed in untreated wt mice. By combined antibiotic and TACE inhibitor treatment 95% of TLR2-/- mice were rescued. CONCLUSION: During pneumococcal meningitis strong inflammation in TLR2-deficiency was associated with incomplete responsiveness to antibiotics and complete response to combined antibiotic and TACE inhibitor treatment. TACE inhibitor treatment offers a promising adjuvant therapeutic strategy in pneumococcal meningitis.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Antibacterianos/farmacología , Ácidos Hidroxámicos/farmacología , Meningitis Neumocócica/tratamiento farmacológico , Infecciones Neumocócicas/tratamiento farmacológico , Receptor Toll-Like 2/inmunología , Proteína ADAM17 , Animales , Ceftriaxona/farmacología , Quimioterapia Adyuvante , Femenino , Receptores de Lipopolisacáridos/inmunología , Masculino , Meningitis Neumocócica/enzimología , Meningitis Neumocócica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Neumocócicas/enzimología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/crecimiento & desarrollo , Receptor Toll-Like 2/deficiencia , Factor de Necrosis Tumoral alfa/metabolismo
18.
Microbes Infect ; 19(11): 515-526, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28689009

RESUMEN

Mycobacterium tuberculosis is one of the most successful pathogens known, having infected more than a third of the global population. An important strategy for intracellular survival of pathogenic mycobacteria relies on their capacity to resist delivery to lysosomes, instead surviving within macrophage phagosomes. Several factors of both mycobacterial and host origin have been implicated in this process. However, whether or not this strategy is employed in vivo is not clear. Here we show that in vivo, following intravenous infection, M. tuberculosis and Mycobacterium bovis BCG initially survived by resisting lysosomal transfer. However, after prolonged infection the bacteria were transferred to lysosomes yet continued to proliferate. A M. bovis BCG mutant lacking protein kinase G (PknG), that cannot avoid lysosomal transfer and is readily cleared in vitro, was found to survive and proliferate in vivo. The ability to survive and proliferate in lysosomal organelles in vivo was found to be due to an altered host environment rather than changes in the inherent ability of the bacteria to arrest phagosome maturation. Thus, within an infected host, both M. tuberculosis and M. bovis BCG adapts to infection-specific host responses. These results are important to understand the pathology of tuberculosis and may have implications for the development of effective strategies to combat tuberculosis.


Asunto(s)
Lisosomas/microbiología , Mycobacterium bovis/metabolismo , Mycobacterium tuberculosis/metabolismo , Animales , Bovinos , Interacciones Huésped-Patógeno , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagosomas/microbiología , Tuberculosis/microbiología , Tuberculosis Bovina/metabolismo
19.
J Neurosci ; 25(48): 11125-32, 2005 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-16319312

RESUMEN

The significance of the peripheral immune system in Alzheimer's disease pathogenesis remains controversial. To study the CNS invasion of hematopoietic cells in the course of cerebral amyloidosis, we used a green fluorescence protein (GFP)-bone marrow chimeric amyloid precursor protein transgenic mouse model (APP23 mice). No difference in the number of GFP-positive invading cells was observed between young APP23 mice and nontransgenic control mice. In contrast, in aged, amyloid-depositing APP23 mice, a significant increase in the number of invading ameboid-like GFP-positive cells was found compared with age-matched nontransgenic control mice. Interestingly, independent of the time after transplantation, only a subpopulation of amyloid deposits was surrounded by invading cells. This suggests that not all amyloid plaques are a target for invading cells or, alternatively, all amyloid plaques attract invading cells but only for a limited time, possibly at an early stage of plaque evolution. Immunological and ultrastructural phenotyping revealed that macrophages and T-cells accounted for a significant portion of these ameboid-like invading cells. Macrophages did not show evidence of amyloid phagocytosis at the electron microscopic level, and no obvious signs for T-cell-mediated inflammation or neurodegeneration were observed. The observation that hematopoietic cells invade the brain in response to cerebral amyloidosis may hold an unrecognized therapeutic potential.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Células de la Médula Ósea , Trasplante de Médula Ósea , Encéfalo/metabolismo , Movimiento Celular , Hematopoyesis , Amiloide , Neuropatías Amiloides/metabolismo , Neuropatías Amiloides/patología , Neuropatías Amiloides/fisiopatología , Animales , Células de la Médula Ósea/patología , Encefalopatías/metabolismo , Encefalopatías/patología , Encefalopatías/fisiopatología , Proteínas Fluorescentes Verdes , Macrófagos , Masculino , Ratones , Ratones Transgénicos , Fagocitosis , Linfocitos T/patología , Quimera por Trasplante
20.
J Gastrointest Surg ; 10(6): 837-47, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16769540

RESUMEN

Acute pancreatitis induces liver injury by upregulating Kupffer cell-derived Fas/FasL; on the other hand, acute pancreatitis induces apoptosis of Kupffer cells via NF-kappaB-dependent pathways. The balance between upregulation of Fas/FasL and Fas/FasL-induced apoptosis of its originator cell may determine the severity of pancreatitis-related liver injury. The aim of our study was to determine the role of p65 NF-kappaB/RelA in pancreatitis-induced Kupffer cell apoptosis. Acute pancreatitis was induced in NIH Swiss mice by a choline-deficient ethionine-supplement (CDE) diet. In vitro mouse Kupffer cell line was transfected with p65 siRNA and treated with pancreatic elastase to mimic pancreatitis. CDE pancreatitis upregulated nuclear translocation of p65 NF-kappaB/RelA, Fas/FasL, caspase-3, and DNA fragmentation in mice livers (all P < 0.001). In vitro, pancreatic elastase mimicked CDE-pancreatitis by upregulating nuclear translocation of p65 NF-kappaB/RelA, Fas/FasL, caspase-3, DNA fragmentation, and apoptosis in Kupffer cells (all P < 0.001). Transfection with p65 siRNA attenuated the elastase-induced nuclear translocation of p65 NF-kappaB/RelA, upregulation of Fas/FasL, caspase-3, DNA fragmentation, and apoptosis in Kupffer cells (all P < 0.001). Acute pancreatitis activates p65 NF-kappaB/RelA and induces apoptosis of Kupffer cells. Inhibition of p65NF-kappaB/RelA attenuates elastase-induced upregulation of proapoptotic pathways and apoptosis in Kupffer cells. The ability of Kupffer cells to autoregulate their stress response by inducing self-apoptosis warrants further investigation.


Asunto(s)
Apoptosis/fisiología , Macrófagos del Hígado/fisiología , Pancreatitis/fisiopatología , Factor de Transcripción ReIA/fisiología , Enfermedad Aguda , Animales , Caspasa 3 , Caspasas/metabolismo , Fragmentación del ADN , Ensayo de Inmunoadsorción Enzimática , Proteína Ligando Fas , Femenino , Citometría de Flujo , Homeostasis/fisiología , Immunoblotting , Macrófagos del Hígado/patología , Glicoproteínas de Membrana/metabolismo , Ratones , Pancreatitis/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Translocación Genética , Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba/fisiología , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA