Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(14)2022 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-35886967

RESUMEN

The human gut symbiont Lacticaseibacillus (L.) casei (previously Lactobacillus casei) is under intense research due to its wide range of immunomodulatory effects on the human host. Dendritic cells (DCs) are crucial players in the direct and indirect communication with lactobacilli in the gastrointestinal tract. Here, we demonstrate that human monocyte-derived DCs (moDCs) are able to engulf L. casei BL23, in which the intact bacterial cell wall and morphology have a key role. The absence of the bacterial cell-wall-degrading enzyme, Lc-p75, in L. casei cells causes remarkable morphological changes, which have important consequences in the phagocytosis of L. casei by moDCs. Our results showed that the Lc-p75 mutation induced defective internalization and impaired proinflammatory and T-cell-polarizing cytokine secretion by bacteria-exposed moDCs. The T helper (Th) 1 and Th17 cell activating capacity of moDCs induced by the mutant L. casei was consequently reduced. Moreover, inhibition of the phagocytosis of wild-type bacteria showed similar results. Taken together, these data suggested that formation of short bacterial chains helps to exert the potent immunomodulatory properties of L. casei BL23.


Asunto(s)
Células Dendríticas , Lacticaseibacillus casei , N-Acetil Muramoil-L-Alanina Amidasa , Células Dendríticas/inmunología , Humanos , Lacticaseibacillus casei/genética , Lacticaseibacillus casei/inmunología , Lacticaseibacillus casei/fisiología , Monocitos/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/biosíntesis , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Fagocitosis
2.
Sci Rep ; 11(1): 13865, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226629

RESUMEN

Staphylococcus aureus is an opportunistic pathogen causing high morbidity and mortality. Since multi-drug resistant S. aureus lineages are nowadays omnipresent, alternative tools for preventive or therapeutic interventions, like immunotherapy, are urgently needed. However, there are currently no vaccines against S. aureus. Surface-exposed and secreted proteins are regarded as potential targets for immunization against S. aureus infections. Yet, many potential staphylococcal antigens of this category do not elicit protective immune responses. To obtain a better understanding of this problem, we compared the binding of serum IgGs from healthy human volunteers, highly S. aureus-colonized patients with the genetic blistering disease epidermolysis bullosa (EB), or immunized mice to the purified S. aureus peptidoglycan hydrolases Sle1, Aly and LytM and their different domains. The results show that the most abundant serum IgGs from humans and immunized mice target the cell wall-binding domain of Sle1, and the catalytic domains of Aly and LytM. Interestingly, in a murine infection model, these particular IgGs were not protective against S. aureus bacteremia. In contrast, relatively less abundant IgGs against the catalytic domain of Sle1 and the N-terminal domains of Aly and LytM were almost exclusively detected in sera from EB patients and healthy volunteers. These latter IgGs may contribute to the protection against staphylococcal infections, as previous studies suggest that serum IgGs protect EB patients against severe S. aureus infection. Together, these observations focus attention on the use of particular protein domains for vaccination to direct potentially protective immune responses towards the most promising epitopes within staphylococcal antigens.


Asunto(s)
Inmunoglobulina G/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Dominio Catalítico/genética , Dominio Catalítico/inmunología , Pared Celular/genética , Pared Celular/inmunología , Epítopos/genética , Epítopos/inmunología , Humanos , Inmunoglobulina G/genética , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , N-Acetil Muramoil-L-Alanina Amidasa/química , Peptidoglicano/genética , Peptidoglicano/inmunología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/prevención & control
3.
Immunol Lett ; 212: 125-131, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30496765

RESUMEN

Staphylococcus aureus is a leading infectious cause of life-threatening diseases in human beings, with no effective vaccine available to date against this bacterium. Treatment of methicillin-resistant S. aureus (MRSA) infections has become increasingly difficult because of the emergence of multidrug-resistant isolates. Immunotherapy represents a potential approach to prevent S. aureus-related infections. Autolysin is one of the virulence factors, which controls the growth, cell lysis, daughter-cell separation, and biofilm formation. Our study focused on passive immunization against MRSA infection. Herein, rabbit polyclonal IgG was produced following the preparation of r-autolysin. Specificity of IgG against r-autolysin was investigated by ELISA and western blotting assays. IgG fraction was prepared using sulfate ammonium precipitation, and the ability of antiserum to promote phagocytosis of bacteria was assessed by opsonophagocytosis assay. Then, passive immunization of mice was carried out with polyclonal IgG fraction and, mice were sacrificed three days after challenge and their kidneys, liver, and spleen were collected. Results exhibited that the passive immunization with rabbit polyclonal anti-IgG fraction tremendously improved survival rates of mice challenged by S. aureus as well as vancomycin treatment compared with the negative control groups. In addition, a remarkable decrease in bacterial numbers was observed in mice treated with rabbit polyclonal anti-IgG. Importantly, our findings demonstrated that passive immunotherapy and antibiotic therapy lead to decreased histopathological damage in mice infected by S. aureus as compared with control groups. Our results suggested that the passive immunization may result in the introduction of excellent strategies to control infections caused by MRSA, like antibiotic therapy.


Asunto(s)
Antibacterianos/uso terapéutico , Inmunización Pasiva/métodos , Inmunoglobulina G/administración & dosificación , Staphylococcus aureus Resistente a Meticilina/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Infecciones Estafilocócicas/terapia , Animales , Proteínas Bacterianas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulina G/inmunología , Staphylococcus aureus Resistente a Meticilina/genética , Ratones , N-Acetil Muramoil-L-Alanina Amidasa/genética , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Resultado del Tratamiento , Factores de Virulencia/inmunología
4.
Mol Immunol ; 91: 1-7, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28858628

RESUMEN

The methicillin-resistant Staphylococcus aureus infection is a hot topic area in microbiology research. Here a novel vaccine candidate consisting of recombinant PBP2a and autolysin proteins were used. The proteins over expressed in E.coli BL21 (DE3) cells, and purified by the Ni-NTA affinity column and conjugated using EDAC and ADH as a linker and spacer, respectively. To investigate the immunogenicity and protective effects of recombinant proteins, 5 and 20µg of proteins in various formulations were subcutaneously injected in different groups. Two booster vaccinations were carried out in three-week intervals and blood samples were collected three weeks after each injection. To evaluate the immune response, total IgG, IgG1, IgG2a, and IgG2b were analyzed. Immunization of mice with r-autolysin and r-autolysin-PBP2a mixture raised total IgGantibody. Additionally, both IgG1 and IgG2a responses induced. Opsonophagocytosis assay showed that anti r-PBP2a and r-autolysin IgG not only promoted phagocytosis of S.aureus, but also decreased the number of viable bacterial cells. Furthermore, survival rate of experimental mice increased in the bacteremia infection. Our results demonstrated that active vaccination with a mixture of r-PBP2a/r-autolysin and conjugate form vaccine reduced the mortality rate and protected mice against lethal MRSA challenge as well as single proteins.


Asunto(s)
Proteínas Bacterianas/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Proteínas de Unión a las Penicilinas/inmunología , Infecciones Estafilocócicas/prevención & control , Vacunas Estafilocócicas/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Femenino , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones Estafilocócicas/inmunología , Vacunación
5.
Mol Immunol ; 82: 10-18, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28006655

RESUMEN

Staphylococcus aureus (MRSA) is an opportunistic pathogen which causes a variety of clinical diseases and leads to high rates of morbidity and mortality. Development of an effective vaccine appears to be a useful strategy to control the infection. Here, the internal region of atl was cloned into the pET24a plasmid and expressed in E. coli BL21 (DE3). Cloning of atl was confirmed by colony-PCR, enzymatic digestion and sequencing. Protein expressed in E coli, BL21 DE3 and was confirmed with SDS-PAGE and western blot analysis. Subsequently, BALB/c mice were injected subcutaneously three times with 20µg of the recombinant autolysin. After Bleeding, autolysin-specific total IgG antibodies and isotypes were evaluated using ELISA. Opsonophagocytic killing assay was performed and experimental challenge was done by intraperitoneal injection with sub lethal doses of MRSA in mice and also survival rate was regularly monitored. Results showed that vaccinated mice could exhibit higher levels of autolysin-specific antibodies (P<0.0001) with a predominant IgG1 response versus control group. Results from in vitro experiments indicated that S. aureus opsonized with immunized-mice sera displayed significantly increased phagocytic uptake and effective intracellular killing versus non-immunized mice. The number of viable bacteria in the kidney of immunized mice showed 1000 times less than the control mice; additionally, an increased survival rate was found after immunization with the candidate vaccine versus control group. Results from this study demonstrated that the autolysin is a valuable target for the development of immunotherapeutic strategies against S. aureus and candidate vaccines.


Asunto(s)
Antígenos Bacterianos/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Infecciones Estafilocócicas/inmunología , Vacunas Estafilocócicas/inmunología , Animales , Proteínas Bacterianas/inmunología , Western Blotting , Clonación Molecular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos BALB C , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/aislamiento & purificación , Vacunas Sintéticas
6.
Vaccine ; 34(50): 6148-6157, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27840016

RESUMEN

The cell wall glucosaminidase LytB of Streptococcus pneumoniae is a surface exposed protein involved in daughter cell separation, biofilm formation and contributes to different aspects of the pathogenesis process. In this study we have characterized the antibody responses after immunization of mice with LytB in the presence of alhydrogel as an adjuvant. Enzyme-linked immunosorbent assays measuring different subclasses of immunoglobulin G, demonstrated that the antibody responses to LytB were predominantly IgG1 and IgG2b, followed by IgG3 and IgG2a subclasses. Complement-mediated immunity against two different pneumococcal serotypes was investigated using sera from immunized mice. Immunization with LytB increased the recognition of S. pneumoniae by complement components C1q and C3b demonstrating that anti-LytB antibodies trigger activation of the classical pathway. Phagocytosis assays showed that serum containing antibodies to LytB stimulates neutrophil-mediated phagocytosis against S. pneumoniae. Animal models of infection including invasive pneumonia and sepsis were performed with two different clinical isolates. Vaccination with LytB increased bacterial clearance and induced protection demonstrating that LytB might be a good candidate to be considered in a future protein-based vaccine against S. pneumoniae.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Fagocitosis , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/inmunología , Neumonía Bacteriana/prevención & control , Sepsis/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Hidróxido de Aluminio/administración & dosificación , Animales , Anticuerpos Antibacterianos/sangre , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunoglobulina G/sangre , Factores Inmunológicos/metabolismo , Masculino , Ratones Endogámicos BALB C , N-Acetil Muramoil-L-Alanina Amidasa/administración & dosificación , Neutrófilos/inmunología , Infecciones Neumocócicas/inmunología , Vacunas Neumococicas/administración & dosificación , Neumonía Bacteriana/inmunología , Sepsis/inmunología , Streptococcus pneumoniae/inmunología , Resultado del Tratamiento
7.
Science ; 353(6306): 1434-1437, 2016 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-27708039

RESUMEN

The intestinal microbiome modulates host susceptibility to enteric pathogens, but the specific protective factors and mechanisms of individual bacterial species are not fully characterized. We show that secreted antigen A (SagA) from Enterococcus faecium is sufficient to protect Caenorhabditis elegans against Salmonella pathogenesis by promoting pathogen tolerance. The NlpC/p60 peptidoglycan hydrolase activity of SagA is required and generates muramyl-peptide fragments that are sufficient to protect C. elegans against Salmonella pathogenesis in a tol-1-dependent manner. SagA can also be heterologously expressed and secreted to improve the protective activity of probiotics against Salmonella pathogenesis in C. elegans and mice. Our study highlights how protective intestinal bacteria can modify microbial-associated molecular patterns to enhance pathogen tolerance.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Enterococcus faecium/inmunología , Microbioma Gastrointestinal/inmunología , Interacciones Huésped-Patógeno/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Infecciones por Salmonella/prevención & control , Salmonella typhimurium/inmunología , Animales , Caenorhabditis elegans/inmunología , Caenorhabditis elegans/microbiología , Proteínas de Caenorhabditis elegans , Enterococcus faecium/enzimología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas del Tejido Nervioso , Probióticos
8.
Cell Microbiol ; 17(2): 147-63, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25225110

RESUMEN

Inducing long-term protective memory CD8(+) T-cells is a desirable goal for vaccines against intracellular pathogens. However, the mechanisms of differentiation of CD8(+) T-cells into long-lived memory cells capable of mediating protection of immunized hosts remain incompletely understood. We have developed an experimental system using mice immunized with wild type (WT) or mutants of the intracellular bacterium Listeria monocytogenes (Lm) that either do or do not develop protective memory CD8(+) T-cells. We previously reported that mice immunized with Lm lacking functional SecA2, an auxiliary secretion system of gram-positive bacteria, did not differentiate functional memory CD8(+) T-cells that protected against a challenge infection with WT Lm. Herein we hypothesized that the p60 and NamA autolysins of Lm, which are major substrates of the SecA2 pathway, account for this phenotype. We generated Lm genetically deficient for genes encoding for the p60 and NamA proteins, ΔiapΔmurA Lm, and further characterized this mutant. Δp60ΔNamA Lm exhibited a strong filamentous phenotype, inefficiently colonized host tissues, and grew mostly outside cells. When Δp60ΔNamA Lm was made single unit, cell invasion was restored to WT levels during vaccination, yet induced memory T-cells still did not protect immunized hosts against recall infection. Recruitment of blood phagocytes and antigen-presenting cell activation was close to that of mice immunized with ΔActA Lm, which develop protective memory. However, key inflammatory factors involved in optimal T-cell programming such as IL-12 and type I IFN (IFN-I) were lacking, suggesting that cytokine signals may largely account for the observed phenotype. Thus, altogether, these results establish that p60 and NamA secreted by Lm promote primary host cell invasion, the inflammatory response and the differentiation of functional memory CD8(+) T-cells, by preventing Lm filamentation during growth and subsequent triggering of innate sensing mechanisms.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Memoria Inmunológica , Listeria monocytogenes/inmunología , Listeriosis/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Animales , Proteínas Bacterianas/genética , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Eliminación de Gen , Listeria monocytogenes/genética , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , N-Acetil Muramoil-L-Alanina Amidasa/genética , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo
9.
PLoS One ; 8(2): e55098, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23390515

RESUMEN

Listeria monocytogenes serotype 4b is responsible for a high percentage of fatal cases of food-borne infection. In a previous study, we created 15 monoclonal antibodies (MAbs) against a ≈ 77 kDa antigen that is associated with the cell surface of live L. monocytogenes serotype 4b cells. Here we report an extensive characterization of these MAbs to further their development as diagnostic reagents. The ≈ 77 kDa target antigen was identified by mass spectrometry and N-terminal sequencing to be IspC, a novel surface associated autolysin. Epitope localization experiments revealed that each of the 15 MAbs recognized the C-terminal cell-wall binding domain of IspC. The presence of IspC was shown to be highly conserved within L. monocytogenes serotype 4b, as evidenced by a strong reaction between anti-IspC MAbs and all 4b isolates. To determine the range of cross-reactivity with other L. monocytogenes serotypes ELISA was used to test each MAb against multiple isolates from each of the L. monocytogenes serotypes. Of the 15 MAbs, five: M2774, M2775, M2780, M2790 and M2797, showed specificity for L. monocytogenes serotype 4b and only cross reacted with serotype 4ab isolates. The kinetics of the interaction between each of the MAbs and IspC was measured using surface plasmon resonance. The MAbs M2773, M2792, M2775, M2797 and M2781 each had very low dissociation constants (4.5 × 10(-9) to 1.2 × 10(-8) M). While several of these antibodies have properties which could be useful in diagnostic tests, the combined high fidelity and affinity of M2775 for the IspC protein and serotype 4b isolates, makes it a particularly promising candidate for use in the development of a specific L. monocytogenes serotype 4b diagnostic test.


Asunto(s)
Anticuerpos Monoclonales , Antígenos Bacterianos/análisis , Proteínas Bacterianas/análisis , Pared Celular/química , Epítopos/química , Listeria monocytogenes/química , N-Acetil Muramoil-L-Alanina Amidasa/análisis , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Sitios de Unión , Pared Celular/inmunología , Reacciones Cruzadas , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Cinética , Listeria monocytogenes/inmunología , Espectrometría de Masas , Datos de Secuencia Molecular , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Unión Proteica , Alineación de Secuencia , Serotipificación , Resonancia por Plasmón de Superficie
10.
Hum Vaccin Immunother ; 8(6): 799-805, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22495112

RESUMEN

We prospectively compared serum antibody levels of 5 Streptococcus pneumoniae (Spn) proteins: PcpA PhtD, PhtE Ply and LytB associated with nasopharyngeal (NP) colonization and acute otitis media (AOM) infection in a cohort of 6-30 mo old children. Antigen-specific antibody titers were determined by ELISA. A total of 731 visits among 168 children were studied. There were 301 Spn NP colonization episodes documented in 109 (65%) children and 42 Spn AOM episodes in 34 (20%) children. IgG antibody titers to the 5 proteins were significantly different among children over time (p < 0.001), with a rank order as follows: PcpA > PhtE = PhtD > Ply > LytB Characterization of IgG and IgM acute and convalescent serum antibody levels of Spn AOM infection showed the kinetics of the response differed among children, with the same rank order of antibody levels over time. Individual data showed that some children responded to AOM with an antibody increase to one or more of these Spn proteins but some children failed to respond. We conclude that antibody levels to Spn proteins PcpA PhtD, PhtE, Ply and LytB, all rise over time in children age 6 to 30 mo following natural exposure to Spn after NP colonization and AOM; however, there were significant differences in quantity of antibody elicited among these potential vaccine antigens.


Asunto(s)
Formación de Anticuerpos/inmunología , Proteínas Bacterianas/inmunología , Proteínas Portadoras/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Nasofaringe/microbiología , Otitis Media/microbiología , Streptococcus pneumoniae/inmunología , Estreptolisinas/inmunología , Anticuerpos Antibacterianos/inmunología , Preescolar , Femenino , Humanos , Lactante , Péptidos y Proteínas de Señalización Intracelular , Masculino
11.
Vet Microbiol ; 157(3-4): 439-47, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22342493

RESUMEN

Staphylococcus aureus is an opportunistic pathogen in dairy ruminants where it is found in healthy carriage and can be a major cause of mastitis. A better knowledge of the host-pathogen interactions is needed to tackle this serious animal health problem. This study aimed at identifying S. aureus proteins differentially expressed by S. aureus in nasal colonization versus mastitis. Serological proteome analysis (SERPA) was used to examine protein samples prepared from culture supernatants of S. aureus strains originally isolated from gangrenous mastitis and nasal carriage (O11) or subclinical mastitis (O46) and to compare patterns of immune-reactive proteins. These staphylococcal proteins were revealed by sera obtained from ewes suffering from S. aureus mastitis and by sera obtained from healthy nulliparous ewes (i.e. no lactation and no mastitis or other symptoms) that were nasally colonized by S. aureus. Altogether 49 staphylococcal immune-reactive proteins were identified in this study. Patterns of proteins revealed by sera from infected- or healthy carrier- animals were comparable and analysis singled out one immune-reactive protein, N-acetylmuramyl-L-alanine amidase, which was recognized by each of the 6 sera from infected animals, when tested individually, and not by the sera of healthy carriers. This is the first study that compares the S. aureus seroproteome in colonization versus mastitis context in ruminants. These results open avenues for studies aiming at a better understanding of the balance between infection and commensal lifestyle in this opportunistic pathogen and at new prevention strategies.


Asunto(s)
Proteínas Bacterianas/inmunología , Portador Sano , Mastitis/inmunología , Enfermedades de las Ovejas/inmunología , Oveja Doméstica/inmunología , Infecciones Estafilocócicas/veterinaria , Animales , Proteínas Bacterianas/metabolismo , Femenino , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Nariz/microbiología , Proteoma , Ovinos , Enfermedades de las Ovejas/microbiología , Oveja Doméstica/microbiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo
12.
Pediatr Pulmonol ; 46(2): 179-83, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20963842

RESUMEN

BACKGROUND: Empyema is a complication of pneumonia, commonly caused by Streptococcus pneumoniae. AIMS: To validate the utility of an immunochromatographic test for the detection of S. pneumoniae antigen in the pleural fluid of children with empyema. METHODS: Empyema patients had blood and pleural fluid cultured, and polymerase chain reaction (PCR) to detect the S. pneumoniae autolysin gene, lytA, in pleural fluid. Pleural fluid was tested using the Binax NOW S. pneumoniae antigen detection assay and compared with lytA PCR results and/or culture in blood or pleural fluid. RESULTS: S. pneumoniae was detected by PCR in pleural fluid of 68 of 137 (49.6%) patients, by culture in 11 of 135 (8.1%) pleural specimens and 16 of 120 (13.3%) blood specimens. Pleural fluid Binax NOW testing from 130 patients demonstrated a sensitivity of 83.8% and specificity of 93.5% (positive predictive value of 93.4% and negative predictive value of 84.1%). CONCLUSIONS: In pediatric empyema, high predictive values of pleural fluid Binax NOW S. pneumoniae antigen test suggest that this test may help rationalize antibiotic choice in these patients.


Asunto(s)
Empiema/diagnóstico , Empiema/microbiología , Infecciones Neumocócicas/diagnóstico , Sistemas de Atención de Punto , Streptococcus pneumoniae/aislamiento & purificación , Adolescente , Antígenos Bacterianos/sangre , Antígenos Bacterianos/genética , Niño , Preescolar , Empiema/inmunología , Femenino , Humanos , Lactante , Masculino , N-Acetil Muramoil-L-Alanina Amidasa/análisis , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Derrame Pleural/inmunología , Derrame Pleural/microbiología , Infecciones Neumocócicas/inmunología , Reacción en Cadena de la Polimerasa/métodos , Sensibilidad y Especificidad , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/inmunología
13.
Artículo en Ruso | MEDLINE | ID: mdl-21384595

RESUMEN

Streptococcus pneumoniae cell wall and cytoplasmic proteins contribute directly to pathogenesis of pneumococcal infection. Protective effect of pneumococcal proteins such as pneumolysin (Ply), muramylamidase (LytA) and pneumococcal surface protein A (PspA). There is discussion in the literature about development of conjugared pneumococcal vaccines, which should include polysaccharides of invasive serotypes of pneumococci as well as protein antigens of this pathogen, for prevention of infections caused by S. pneumoniae. Researches suggest that such hybrid vaccines will be effective, first of all, for children < 2 years of age and elderly > 65 years old because immune response to polysaccharide vaccines either do not form at all or insufficient for prevention of pneumococcal infection.


Asunto(s)
Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Pared Celular/inmunología , Citoplasma/inmunología , Humanos , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Polisacáridos/inmunología , Estreptolisinas/química , Estreptolisinas/inmunología , Vacunas Conjugadas/inmunología , Factores de Virulencia/química , Factores de Virulencia/inmunología
14.
Clin Vaccine Immunol ; 16(6): 916-23, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19403781

RESUMEN

This study focuses on the immunogenicity of the following three pneumococcal vaccine candidate proteins in Filipino infants, all inducing protection in animal models: pneumococcal histidine triad protein D (PhtD), choline binding protein A (CbpA), and the lysozyme LytC. The immunoglobulin G antibody concentrations to PhtD, its putative, protective, and exposed C-terminal fragment (PhtD C), CbpA, and LytC were measured by enzyme immunoassay in 52 serum samples from pregnant women, 39 cord blood samples, and consecutive serum samples (n = 263) from 52 newborns between 6 weeks and 10 months of age scheduled to be taken at six time points. A nasopharyngeal swab to detect pneumococcal carriage was taken parallel to the serum samples. The antibody concentrations in the cord blood samples were similar to those in the samples from the mothers. In infant sera, the geometric mean antibody concentrations (GMCs) for all three proteins decreased until the age of 18 weeks and started to increase after that age, suggesting that the infants' own antibody production started close to the age of 4 to 5 months. The increase in GMCs by age, most clear-cut for CbpA, was associated with pneumococcal carriage. Anti-PhtD concentrations were higher than anti-PhtD C concentrations but correlated well (r of 0.89 at 10.5 months), suggesting that antibodies are directed to the supposedly exposed and protective C-terminal part of PhtD. Our results show that young children are able to develop an antibody response to PhtD, CbpA, and LytC and encourage the development of pneumococcal protein vaccines for this age group.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Proteínas Bacterianas/inmunología , Portador Sano/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Factores de Edad , Animales , Femenino , Sangre Fetal/inmunología , Humanos , Lactante , Masculino , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Nasofaringe/microbiología , Filipinas , Embarazo , Mujeres Embarazadas
15.
J Innate Immun ; 1(2): 88-97, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19319201

RESUMEN

Peptidoglycan (PGN) is a major component of the bacterial cell envelope in both Gram-positive and Gram-negative bacteria. These muropeptides can be produced or modified by the activity of bacterial glycolytic and peptidolytic enzymes referred to as PGN hydrolases and autolysins. Some of these bacterial enzymes are crucial for bacterial pathogenicity and have been shown to modulate muropeptide release and/or host innate immune responses. The ability of muropeptides to modulate host responses is due to the fact that eukaryotes do not produce PGN and have instead evolved numerous strategies to detect intact PGN and PGN fragments (muropeptides). Here we review the structure of PGN and introduce the various bacterial enzymes known to degrade or modify bacterial PGN. Host factors involved in PGN and muropeptide detection are also briefly discussed, as are examples of how specific bacterial pathogens use PGN degradation and modification to subvert host innate immunity.


Asunto(s)
Bacterias/enzimología , Interacciones Huésped-Patógeno/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Peptidoglicano/inmunología , Secuencia de Aminoácidos , Animales , Bacterias/patogenicidad , Infecciones Bacterianas/inmunología , Humanos , Inmunidad Innata , Ratones , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Peptidoglicano/metabolismo , Estructura Terciaria de Proteína
16.
J Immunol ; 178(4): 2407-14, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17277147

RESUMEN

Both peptidoglycan and muropeptides potently modulate inflammatory and innate immune responses. The secreted Listeria monocytogenes p60 autolysin digests peptidoglycan and promotes bacterial infection in vivo. Here, we report that p60 contributes to bacterial subversion of NK cell activation and innate IFN-gamma production. L. monocytogenes deficient for p60 (Deltap60) competed well for expansion in mice doubly deficient for IFNAR1 and IFN-gammaR1 or singly deficient for IFN-gammaR1, but not in wild-type, IFNAR1(-/-), or TLR2(-/-) mice. The restored competitiveness of p60-deficient bacteria suggested a specific role for p60 in bacterial subversion of IFN-gamma-mediated immune responses, since in vivo expansion of three other mutant L. monocytogenes strains (DeltaActA, DeltaNamA, and DeltaPlcB) was not complemented in IFN-gammaR1(-/-) mice. Bacterial expression of p60 was not required to induce socs1, socs3, and il10 expression in infected mouse bone marrow macrophages but did correlate with enhanced production of IL-6, IL-12p70, and most strikingly IFN-gamma. The primary source of p60-dependent innate IFN-gamma was NK cells, whereas bacterial p60 expression did not significantly alter innate IFN-gamma production by T cells. The mechanism for p60-dependent NK cell stimulation was also indirect, given that treatment with purified p60 protein failed to directly activate NK cells for IFN-gamma production. These data suggest that p60 may act on infected cells to indirectly enhance NK cell activation and increase innate IFN-gamma production, which presumably promotes early bacterial expansion through its immunoregulatory effects on bystander cells. Thus, the simultaneous induction of IFN-gamma production and factors that inhibit IFN-gamma signaling may be a common strategy for misdirection of early antibacterial immunity.


Asunto(s)
Proteínas Bacterianas/inmunología , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Activación de Linfocitos/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/microbiología , Efecto Espectador/inmunología , Regulación Bacteriana de la Expresión Génica/inmunología , Inmunidad Innata/genética , Interferón gamma/biosíntesis , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Interleucina-6/biosíntesis , Interleucina-6/inmunología , Células Asesinas Naturales/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutación , N-Acetil Muramoil-L-Alanina Amidasa/biosíntesis , N-Acetil Muramoil-L-Alanina Amidasa/genética , Receptores de Interferón/deficiencia , Receptores de Interferón/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
17.
Crit Rev Microbiol ; 32(3): 139-53, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16893751

RESUMEN

Streptococcus pneumoniae is a causative agent for community acquired pneumonia, bacteremia, acute otitis media, and meningitis. Recent emergence of multi-drug resistant clinical isolates prompts the need of effective vaccine for the prevention of disease. The licensed polysaccharide-based pneumococcal vaccines only elicit protective antibodies against the infection of serotypes that are included in the vaccine. To broaden the protection, the use of pneumococcal proteins will be a feasible and preferable alternative. This communication provides a review on the biochemical properties of these protein candidates, their immunization results in animal studies, and perspectives on the development of protein-based pneumococcal vaccine.


Asunto(s)
Proteínas Bacterianas/administración & dosificación , Infecciones Neumocócicas/prevención & control , Streptococcus pneumoniae/química , Adhesinas Bacterianas/administración & dosificación , Adhesinas Bacterianas/inmunología , Animales , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Modelos Animales de Enfermedad , Lipoproteínas/administración & dosificación , Lipoproteínas/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/administración & dosificación , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Neuraminidasa/administración & dosificación , Neuraminidasa/inmunología , Vacunas Neumococicas/administración & dosificación , Estreptolisinas/administración & dosificación , Estreptolisinas/inmunología
18.
J Infect Dis ; 188(7): 1040-8, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14513425

RESUMEN

The Streptococcus pneumoniae cell-wall components peptidoglycan and lipoteichoic acid activate Toll-like receptor 2 (TLR2), which transduces an inflammatory response. After exposure to penicillin, type 2 S. pneumoniae strain D39, but not the isogenic autolysin-deficient mutant AL2, induced significantly enhanced interleukin-8 promoter activity in TLR2-transfected HeLa cells. Lag-phase D39 exhibited enhanced TLR2 activation after exposure to penicillin at levels below the minimum inhibitory concentration (MIC); in contrast, early log-phase S. pneumoniae were most active when exposed to the MIC. This enhancement was not ablated by heat treatment but was attenuated by autolysin inhibitors. The antimicrobial activity of moxifloxacin and erythromycin was not associated with TLR2 activation by S. pneumoniae. These data show that penicillin treatment of S. pneumoniae releases proinflammatory cell-wall components that activate TLR2 and that this activity is dependent on autolysin, the growth phase of the organism, and the antibiotic concentration.


Asunto(s)
Compuestos Aza , Fluoroquinolonas , Glicoproteínas de Membrana/inmunología , Penicilinas/farmacología , Infecciones Neumocócicas/inmunología , Quinolinas , Receptores de Superficie Celular/inmunología , Streptococcus pneumoniae/inmunología , Antiinfecciosos/farmacología , Colina/farmacología , Recuento de Colonia Microbiana , Inhibidores Enzimáticos/farmacología , Eritromicina/farmacología , Etanolamina/farmacología , Células HeLa , Humanos , Interleucina-8/inmunología , Interleucina-8/metabolismo , Moxifloxacino , N-Acetil Muramoil-L-Alanina Amidasa/antagonistas & inhibidores , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Infecciones Neumocócicas/tratamiento farmacológico , Streptococcus pneumoniae/efectos de los fármacos , Receptor Toll-Like 2 , Receptores Toll-Like , Transfección
19.
Blood ; 90(3): 1246-54, 1997 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9242559

RESUMEN

N-acetylmuramyl-L-alanine amidase (NAMLAA) specifically degrades peptidoglycan, which is a major component of bacterial cell walls with strong inflammatory properties. For instance, peptidoglycan is capable of stimulating peripheral blood cells to release pro-inflammatory cytokines and is capable of inducing chronic arthritis in an animal model. In a previous study we found that degradation of peptidoglycan by purified NAMLAA reduced its inflammatory effects. To determine where NAMLAA is located in tissues, monoclonal antibodies against purified NAMLAA were produced for use in immunohistochemistry, immunoelectron microscopy, flow cytometric analysis, and Western blotting. The immunohistochemical studies showed NAMLAA-positive cells in human spleen, liver, arthritic synovial tissues, and lymph nodes. In flow cytometric studies of blood and bone marrow, neutrophilic and eosinophilic granulocytes proved to be positive. Monocytes were negative, although they do contain lysozyme, the other important peptidoglycan-degrading enzyme. However, mature macrophages obtained by bronchoalveolar lavage and subsequent selection based on autofluorescence did possess NAMLAA. In immunocytochemical staining of blood smears, thrombocytes were also positive for NAMLAA. Western blot analysis and immunoelectron microscopy of neutrophils and eosinophils showed that NAMLAA is located in azurophilic granules of neutrophils and in secretory vesicles and crystalloid-containing granules of eosinophils. Flow cytometric analysis of blood and bone marrow from different French-American-British-classified acute myeloid leukemia (AML) patients showed that AML-M2 myeloblasts were the first in the granulocyte maturation lineage that were positive for NAMLAA. The more immature AML, such as AML-M0 and AML-M1, did not express NAMLAA. CD15- and CD13-negative megakaryoblasts, corresponding to AML-M7, were also positive for NAMLAA. The expression pattern of NAMLAA in the myeloid lineage suggests that the monoclonal antibody AAA4, recognizing NAMLAA, is useful for discrimination between AML in the monocyte lineage and in the granulocyte lineage.


Asunto(s)
Plaquetas/enzimología , Pared Celular/metabolismo , Granulocitos/enzimología , Macrófagos Alveolares/enzimología , N-Acetil Muramoil-L-Alanina Amidasa/biosíntesis , Enfermedad Aguda , Anticuerpos Monoclonales/inmunología , Artritis/enzimología , Artritis/patología , Biomarcadores , Plaquetas/ultraestructura , Médula Ósea/enzimología , Líquido del Lavado Bronquioalveolar , Diferenciación Celular , Eosinófilos/enzimología , Eosinófilos/ultraestructura , Granulocitos/ultraestructura , Humanos , Técnicas Inmunológicas , Leucemia Mieloide/enzimología , Leucemia Mieloide/patología , Tejido Linfoide/enzimología , Macrófagos Alveolares/ultraestructura , N-Acetil Muramoil-L-Alanina Amidasa/análisis , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/ultraestructura , Neutrófilos/enzimología , Neutrófilos/ultraestructura , Especificidad de Órganos , Peptidoglicano/metabolismo , Membrana Sinovial/enzimología
20.
J Bacteriol ; 178(6): 1565-71, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8626282

RESUMEN

atl is a newly discovered autolysin gene in Staphylococcus aureus. The gene product, ATL, is a unique, bifunctional protein that has an amidase domain and a glucosaminidase domain. It undergoes proteolytic processing to generate two extracellular peptidoglycan hydrolases, a 59-kDa endo-beta-N-acetylglucosaminidase and a 62-kDa N-acetylmuramyl-L-alanine amidase. It has been suggested that these enzymes are involved in the separation of daughter cells after cell division. We recently demonstrated that atl gene products are cell associated (unpublished data). The cell surface localization of the atl gene products was investigated by immunoelectron microscopy using anti-62-kDa N-acetylmuramyl-L-alanine amidase or anti-51-kDa endo-beta-N-acetylglucosaminidase immunoglobulin G. Protein A-gold particles reacting with the antigen-antibody complex were found to form a ring structure on the cell surface at the septal region for the next cell division site. Electron microscopic examination of an ultrathin section of the preembedded sample revealed preferential distribution of the gold particles at the presumptive sites for cell separation where the new septa had not been completed. The distribution of the gold particles on the surface of protoplast cells and the association of the gold particles with fibrous materials extending from the cells suggested that some atl gene products were associated with a cellular component extending from the cell membrane, such as lipoteichoic acid. The formation of a ring structure of atl gene products may be required for efficient partitioning of daughter cells after cell division.


Asunto(s)
Acetilglucosaminidasa/aislamiento & purificación , Compartimento Celular , N-Acetil Muramoil-L-Alanina Amidasa/aislamiento & purificación , Staphylococcus aureus/crecimiento & desarrollo , Acetilglucosaminidasa/inmunología , División Celular , Oro , Microscopía Inmunoelectrónica , Modelos Biológicos , N-Acetil Muramoil-L-Alanina Amidasa/inmunología , Staphylococcus aureus/enzimología , Staphylococcus aureus/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...