Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Curr Top Microbiol Immunol ; 409: 199-227, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27744506

RESUMEN

Skin and soft tissue infections (SSTIs) are among the most common infections worldwide. They range in severity from minor, self-limiting, superficial infections to life-threatening diseases requiring all the resources of modern medicine. Community (CA) and healthcare (HA) acquired SSTIs are most commonly caused by Staphylococcus aureus . They have variable presentations ranging from impetigo and folliculitis to surgical site infections (SSIs). Superficial SSTIs may lead to even more invasive infections such as bacteraemia and osteomyelitis. Here we describe the anatomical localization of the different SSTI associated with S. aureus, the virulence factors known to play a role in these infections, and their current epidemiology. Current prevention and treatment strategies are also discussed. Global epidemiological data show increasing incidence and severity of SSTIs in association with methicillin-resistant S. aureus strains (MRSA). CA-SSTIs are usually less morbid compared to other invasive infections caused by S. aureus, but they have become the most prevalent, requiring a great number of medical interventions, extensive antibiotic use, and therefore a high cost burden. Recurrence of SSTIs is common after initial successful treatment, and decolonization strategies have not been effective in reducing recurrence. Furthermore, decolonization approaches may be contributing to the selection and maintenance of multi-drug resistant strains. Clinical studies from the early 1900s and novel autovaccination approaches suggest an alternative strategy with potential effectiveness: using vaccines to control S. aureus cutaneous infections.


Asunto(s)
Infecciones Comunitarias Adquiridas , Infecciones de los Tejidos Blandos , Infecciones Estafilocócicas , Infecciones Cutáneas Estafilocócicas , Antibacterianos , Humanos , Infecciones de los Tejidos Blandos/epidemiología , Infecciones de los Tejidos Blandos/terapia , Infecciones Estafilocócicas/epidemiología , Infecciones Estafilocócicas/terapia , Staphylococcus aureus
2.
Curr Top Microbiol Immunol ; 409: 491-528, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28197738

RESUMEN

Staphylococcus aureus is a leading pathogen in surgical site, intensive care unit, and skin infections, as well as healthcare-associated pneumonias. These infections are associated with an enormous burden of morbidity, mortality, and increase of hospital length of stay and patient cost. S. aureus is impressively fast in acquiring antibiotic resistance, and multidrug-resistant strains are a serious threat to human health. Due to resistance or insufficient effectiveness, antibiotics and bundle measures leave a tremendous unmet medical need worldwide. There are no licensed vaccines on the market despite the significant efforts done by public and private initiatives. Indeed, vaccines tested in clinical trials in the last two decades have failed to show efficacy. However, they targeted single antigens and contained no adjuvants and efficacy trials were performed in severely ill subjects. Herein, we provide a comprehensive evaluation of potential target populations for efficacy trials taking into account key factors such as population size, incidence of S. aureus infection, disease outcome, primary endpoints, as well as practical advantages and disadvantages. We describe the whole-blood assay as a potential surrogate of protection, and we show the link between phase III clinical trial data of failed vaccines with their preclinical observations. Finally, we give our perspective on how new vaccine formulations and clinical development approaches may lead to successful S. aureus vaccines.


Asunto(s)
Infecciones Estafilocócicas , Vacunas Estafilocócicas , Antibacterianos , Humanos , Staphylococcus aureus
3.
PLoS Pathog ; 8(4): e1002626, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496652

RESUMEN

Clinical isolates of Staphylococcus aureus can express biofilm phenotypes promoted by the major cell wall autolysin and the fibronectin-binding proteins or the icaADBC-encoded polysaccharide intercellular adhesin/poly-N-acetylglucosamine (PIA/PNAG). Biofilm production in methicillin-susceptible S. aureus (MSSA) strains is typically dependent on PIA/PNAG whereas methicillin-resistant isolates express an Atl/FnBP-mediated biofilm phenotype suggesting a relationship between susceptibility to ß-lactam antibiotics and biofilm. By introducing the methicillin resistance gene mecA into the PNAG-producing laboratory strain 8325-4 we generated a heterogeneously resistant (HeR) strain, from which a homogeneous, high-level resistant (HoR) derivative was isolated following exposure to oxacillin. The HoR phenotype was associated with a R602H substitution in the DHHA1 domain of GdpP, a recently identified c-di-AMP phosphodiesterase with roles in resistance/tolerance to ß-lactam antibiotics and cell envelope stress. Transcription of icaADBC and PNAG production were impaired in the 8325-4 HoR derivative, which instead produced a proteinaceous biofilm that was significantly inhibited by antibodies against the mecA-encoded penicillin binding protein 2a (PBP2a). Conversely excision of the SCCmec element in the MRSA strain BH1CC resulted in oxacillin susceptibility and reduced biofilm production, both of which were complemented by mecA alone. Transcriptional activity of the accessory gene regulator locus was also repressed in the 8325-4 HoR strain, which in turn was accompanied by reduced protease production and significantly reduced virulence in a mouse model of device infection. Thus, homogeneous methicillin resistance has the potential to affect agr- and icaADBC-mediated phenotypes, including altered biofilm expression and virulence, which together are consistent with the adaptation of healthcare-associated MRSA strains to the antibiotic-rich hospital environment in which they are frequently responsible for device-related infections in immuno-compromised patients.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Contaminación de Equipos , Resistencia a la Meticilina/fisiología , Staphylococcus aureus Resistente a Meticilina/metabolismo , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Acetilglucosamina/genética , Acetilglucosamina/metabolismo , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Animales , Antibacterianos/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Masculino , Resistencia a la Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Ratones , N-Acetil Muramoil-L-Alanina Amidasa/genética , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Oxacilina/farmacología , Proteínas de Unión a las Penicilinas/genética , Proteínas de Unión a las Penicilinas/metabolismo , Polisacáridos Bacterianos/genética , Polisacáridos Bacterianos/metabolismo
4.
J Infect Dis ; 205(5): 798-806, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22301683

RESUMEN

The difficulty in successfully treating infections caused by methicillin-resistant Staphylococcus aureus (MRSA) has led to them being referred to as highly virulent or pathogenic. In our study of one of the major healthcare-associated MRSA (HA-MRSA) clones, we show that expression of the gene responsible for conferring methicillin resistance (mecA) is also directly responsible for reducing the ability of HA-MRSA to secrete cytolytic toxins. We show that resistance to methicillin induces changes in the cell wall, which affects the bacteria's agr quorum sensing system. This leads to reduced toxin expression and, as a consequence, reduced virulence in a murine model of sepsis. This diminished capacity to cause infection may explain the inability of HA-MRSA to move into the community and help us understand the recent emergence of community-associated MRSA (CA-MRSA). CA-MRSA typically express less penicillin-binding protein 2a (encoded by mecA), allowing them to maintain full virulence and succeed in the community environment.


Asunto(s)
Proteínas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Resistencia a la Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Animales , Proteínas Bacterianas/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Supervivencia Celular , Pared Celular/metabolismo , Infección Hospitalaria , Modelos Animales de Enfermedad , Expresión Génica , Staphylococcus aureus Resistente a Meticilina/metabolismo , Ratones , Proteínas de Unión a las Penicilinas/metabolismo , Péptido Sintasas/metabolismo , Péptidos Cíclicos , Percepción de Quorum/genética , Sepsis/microbiología , Linfocitos T , Transactivadores/efectos de los fármacos , Transactivadores/genética , Transactivadores/metabolismo
5.
Microbiol Spectr ; 11(1): e0257422, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36688711

RESUMEN

Staphylococcus aureus is a clinically important bacterial pathogen that has become resistant to treatment with most routinely used antibiotics. Alternative strategies, such as vaccination and phage therapy, are therefore actively being investigated to prevent or combat staphylococcal infections. Vaccination requires that vaccine targets are expressed at sufficient quantities during infection so that they can be targeted by the host's immune system. While our knowledge of in vitro expression levels of putative vaccine candidates is comprehensive, crucial in vivo expression data are scarce and promising vaccine candidates during in vitro assessment often prove ineffective in preventing S. aureus infection. Here, we show how a newly developed high-throughput quantitative reverse transcription-PCR (qRT-PCR) assay monitoring the expression of 84 staphylococcal genes encoding mostly virulence factors can inform the selection and design of effective vaccine candidates against staphylococcal infections. We show that this assay can accurately quantify mRNA expression levels of these genes in several host organs relying only on very limited amounts of bacterial mRNA in each sample. We selected two highly expressed genes, lukE and lukD, encoding pore-forming leukotoxins, to inform the design of detoxified recombinant proteins and showed that immunization with recombinant genetically detoxified LukED antigens conferred protection against staphylococcal skin infection in mice. Consequently, knowledge of in vivo-expressed virulence determinants can be successfully deployed to identify and select promising candidates for optimized design of effective vaccine antigens against S. aureus. Notably, this approach should be broadly applicable to numerous other pathogens. IMPORTANCE Vaccination is an attractive strategy for preventing bacterial infections in an age of increased antimicrobial resistance. However, vaccine development frequently suffers significant setbacks when candidate antigens that show promising results in in vitro experimentation fail to protect from disease. An alluring strategy is to focus resources on developing bacterial virulence factors that are expressed during disease establishment or maintenance and are critical for bacterial in-host survival as vaccine targets. While expression profiles of many virulence factors have been characterized in detail in vitro, our knowledge of their in vivo expression profiles is still scarce. Here, using a high-throughput qRT-PCR approach, we identified two highly expressed leukotoxins in a murine infection model and showed that genetically detoxified derivatives of these elicited a protective immune response in a murine skin infection model. Therefore, in vivo gene expression can inform the selection of promising candidates for the design of effective vaccine antigens.


Asunto(s)
Infecciones Estafilocócicas , Vacunas , Animales , Ratones , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/metabolismo , Leucocidinas/genética , Leucocidinas/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Vacunas/metabolismo , Infecciones Estafilocócicas/microbiología , Perfilación de la Expresión Génica
6.
Infect Immun ; 79(3): 1153-65, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21189325

RESUMEN

Staphylococcus aureus clinical isolates are capable of producing at least two distinct types of biofilm mediated by the fibronectin-binding proteins (FnBPs) or the icaADBC-encoded polysaccharide intercellular adhesin (PIA). Deletion of the major autolysin gene atl reduced primary attachment rates and impaired FnBP-dependent biofilm production on hydrophilic polystyrene in 12 clinical methicillin-resistant S. aureus (MRSA) isolates but had no effect on PIA-dependent biofilm production by 9 methicillin-susceptible S. aureus (MSSA) isolates. In contrast, Atl was required for both FnBP- and PIA-mediated biofilm development on hydrophobic polystyrene. Here we investigated the role of Atl in biofilm production on hydrophilic polystyrene. The alternative sigma factor σ(B), which represses RNAIII expression and extracellular protease production, was required for FnBP- but not PIA-dependent biofilm development. Furthermore, mutation of the agr locus enhanced FnBP-dependent biofilm development, whereas a sarA mutation, which increases protease production, blocked FnBP-mediated biofilm development. Mutation of sigB in MRSA isolate BH1CC lowered primary attachment rates, in part via reduced atl transcription. Posttranslational activation or inhibition of Atl activity with phenylmethylsulfonyl fluoride and polyanethole sodium sulfonate or mutation of the Atl amidase active site interfered with lytic activity and biofilm development. Consistent with these observations, extracellular DNA was important for the early stages of Atl/FnBP-dependent biofilm development. Further analysis of atl regulation revealed that atlR encodes a transcriptional repressor of the major autolysin and that an atlR::Tc(r) mutation in BH1CC enhanced biofilm-forming capacity. These data reveal an essential role for the major autolysin in the early events of the FnBP-dependent S. aureus biofilm phenotype.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Biopelículas , Staphylococcus aureus Resistente a Meticilina/fisiología , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Staphylococcus aureus/fisiología , Proteínas Bacterianas/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Regulación Bacteriana de la Expresión Génica , Humanos , Fenotipo , Reacción en Cadena de la Polimerasa , Factor sigma/metabolismo , Transactivadores/metabolismo
7.
J Bacteriol ; 190(11): 3835-50, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18375547

RESUMEN

Device-associated infections involving biofilm remain a persistent clinical problem. We recently reported that four methicillin-resistant Staphylococcus aureus (MRSA) strains formed biofilm independently of the icaADBC-encoded exopolysaccharide. Here, we report that MRSA biofilm development was promoted under mildly acidic growth conditions triggered by the addition of glucose to the growth medium. Loss of sortase, which anchors LPXTG-containing proteins to peptidoglycan, reduced the MRSA biofilm phenotype. Furthermore introduction of mutations in fnbA and fnbB, which encode the LPXTG-anchored multifunctional fibrinogen and fibronectin-binding proteins, FnBPA and FnBPB, reduced biofilm formation by several MRSA strains. However, these mutations had no effect on biofilm formation by methicillin-sensitive S. aureus strains. FnBP-promoted biofilm occurred at the level of intercellular accumulation and not primary attachment. Mutation of fnbA or fnbB alone did not substantially affect biofilm, and expression of either gene alone from a complementing plasmid in fnbA fnbB mutants restored biofilm formation. FnBP-promoted biofilm was dependent on the integrity of SarA but not through effects on fnbA or fnbB transcription. Using plasmid constructs lacking regions of FnBPA to complement an fnbAB mutant revealed that the A domain alone and not the domain required for fibronectin binding could promote biofilm. Additionally, an A-domain N304A substitution that abolished fibrinogen binding did not affect biofilm. These data identify a novel S. aureus biofilm phenotype promoted by FnBPA and FnBPB which is apparently independent of the known ligand-binding activities of these multifunctional surface proteins.


Asunto(s)
Adhesinas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Staphylococcus aureus/fisiología , Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Antibacterianos/farmacología , Regulación Bacteriana de la Expresión Génica/fisiología , Concentración de Iones de Hidrógeno , Resistencia a la Meticilina , Fenotipo , Estructura Terciaria de Proteína , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Factores de Tiempo
8.
Res Microbiol ; 157(6): 547-58, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16765569

RESUMEN

In Escherichia coli and most Gram-negative bacteria, KDO (3-deoxy-D-manno-octulosonate), a component of the lipopolysaccharide inner core, is essential for outer membrane biogenesis and cell viability. Two recently identified genes involved in KDO biosynthesis, kdsD and kdsC, belong to the yrbG-yhbG locus where four additional ORFs (yrbG, yrbK, yhbN and yhbG) with unknown function are located. We have constructed six conditional expression mutants in which the arabinose-inducible araBp promoter is respectively located upstream of each gene of the locus. Complementation analysis of these mutants indicates that the locus is organized in at least three operons and that the three distal genes (yrbK, yhbN and yhbG) are essential for E. coli viability. Surprisingly, kdsD and kdsC (encoding a D-arabinose 5-phosphate isomerase and a KDO 8-phosphate phosphatase, respectively) were shown to be non-essential, indicating genetic redundancy for these two functions. A preliminary characterization of the arabinose-dependent mutants under permissive conditions and upon depletion revealed increased sensitivity to hydrophobic toxic chemicals, suggesting that the mutants have a defective outer membrane. These genes may thus be implicated in cell envelope integrity.


Asunto(s)
Proteínas de Escherichia coli/biosíntesis , Escherichia coli/fisiología , Genes Esenciales , Azúcares Ácidos/metabolismo , Isomerasas Aldosa-Cetosa/biosíntesis , Isomerasas Aldosa-Cetosa/genética , Arabinosa/metabolismo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Lipopolisacáridos/metabolismo , Mutación , Sistemas de Lectura Abierta , Monoéster Fosfórico Hidrolasas/biosíntesis , Monoéster Fosfórico Hidrolasas/genética , Regiones Promotoras Genéticas
9.
Clin Vaccine Immunol ; 23(6): 442-50, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27030589

RESUMEN

Staphylococcus aureus alpha-hemolysin (Hla) assembles into heptameric pores on the host cell membrane, causing lysis, apoptosis, and junction disruption. Herein, we present the design of a newly engineered S. aureus alpha-toxin, HlaPSGS, which lacks the predicted membrane-spanning stem domain. This protein is able to form heptamers in aqueous solution in the absence of lipophilic substrata, and its structure, obtained by transmission electron microscopy and single-particle reconstruction analysis, resembles the cap of the wild-type cytolytic Hla pore. HlaPSGS was found to be impaired in binding to host cells and to its receptor ADAM10 and to lack hemolytic and cytotoxic activity. Immunological studies using human sera as well as sera from mice convalescent from S. aureus infection suggested that the heptameric conformation of HlaPSGS mimics epitopes exposed by the cytolytic Hla pore during infection. Finally, immunization with this newly engineered Hla generated high protective immunity against staphylococcal infection in mice. Overall, this study provides unprecedented data on the natural immune response against Hla and suggests that the heptameric HlaPSGS is a highly valuable vaccine candidate against S. aureus.


Asunto(s)
Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Proteínas Hemolisinas/química , Proteínas Hemolisinas/inmunología , Imitación Molecular , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus , Proteína ADAM10/metabolismo , Animales , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/genética , Línea Celular , Citotoxinas , Epítopos/inmunología , Escherichia coli/genética , Proteínas Hemolisinas/administración & dosificación , Proteínas Hemolisinas/genética , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Electrónica de Transmisión , Modelos Moleculares , Ingeniería de Proteínas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Vacunación
10.
FEMS Microbiol Rev ; 39(5): 750-63, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25994610

RESUMEN

Lack of known mechanisms of protection against Staphylococcus aureus in humans is hindering development of efficacious vaccines. Preclinical as well as clinical data suggest that antibodies play an important role against S. aureus. For instance, certain hypogammaglobulinaemic patients are at increased risk of staphylococcal infections. However, development of effective humoral response may be dampened by converging immune-evasion mechanisms of S. aureus. We hypothesize that B-cell proliferation induced by staphylococcal protein A (SpA) and continuous antigen exposure, without the proper T-cell help and cytokine stimuli, leads to antigen-activated B-cell deletion and anergy. Recent findings suggest an important role of type I neutrophils (PMN-I) and conventionally activated macrophages (M1) against S. aureus, while alternatively activated macrophages (M2) favour biofilm persistence and sepsis. In addition, neutrophil-macrophage cooperation promotes extravasation and activation of neutrophils as well as clearance of bacteria ensnared in neutrophil extracellular traps. Activation of these processes is modulated by cytokines and T cells. Indeed, low CD4(+) T-cell counts represent an important risk factor for skin infections and bacteraemia in patients. Altogether, these observations could lead to the identification of predictive correlates of protection and ways for shifting the balance of the response to the benefit of the host through vaccination.


Asunto(s)
Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Supresión Clonal , Humanos , Evasión Inmune , Linfocitos/citología , Linfocitos/inmunología , Fagocitos/inmunología
11.
mBio ; 5(2): e00974-14, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24667709

RESUMEN

Many pathogens produce the ß-(1-6)-linked poly-N-acetylglucosamine (PNAG) surface polysaccharide that is being developed as a broadly protective antimicrobial vaccine. However, it is unknown whether systemically injected PNAG vaccines or antibodies would provide protective immunity against pathogens confined to the gastrointestinal tract such as Shiga toxin (Stx)-producing Escherichia coli (STEC), an important group of gastrointestinal (GI) pathogens for which effective immunotherapeutics are lacking. To ascertain whether systemic IgG antibody to PNAG impacts this infectious situation, a vaccine consisting of a synthetic nonamer of nonacetylated PNAG, 9GlcNH2, conjugated to the Shiga toxin 1b subunit (9GlcNH2-Stx1b) was produced. Rabbit antibodies raised to the conjugate vaccine were tested for bacterial killing and toxin neutralization in vitro and protection against infection in infant mice. Cell surface PNAG was detected on all 9 STEC isolates tested, representing 6 STEC serogroups, including E. coli O157:H7. Antibody to the 9GlcNH2-Stx1b conjugate neutralized Stx1 potently and Stx2 modestly. For O157:H7 and O104:H4 STEC strains, antibodies elicited by the 9GlcNH2-Stx1b conjugate possessed opsonic killing and bactericidal activity. Following intraperitoneal injection, antibodies to both PNAG and Stx were needed for infant mouse protection against O157 STEC. These antibodies also mediated protection against the Stx2-producing O104:H4 strain that was the cause of a recent outbreak in Germany, although sufficient doses of antibody to PNAG alone were protective against this strain in infant mice. Our observations suggest that vaccination against both PNAG and Stx, using a construct such as the 9GlcNH2-Stx1b conjugate vaccine, would be protective against a broad range of STEC serogroups. IMPORTANCE The presence of poly-N-acetylglucosamine (PNAG) on many pathogens presents an opportunity to target this one structure with a multispecies vaccine. Whether antibodies to PNAG can protect against pathogens confined to the gastrointestinal tract is not known. As Shiga toxin (Stx)-producing Escherichia coli (STEC) bacteria are serious causes of infection whose virulence is dependent on elaboration of Stx, we prepared a vaccine containing a synthetic nonamer of PNAG (9GlcNH2) conjugated to Shiga toxin 1b subunit (9GlcNH2-Stx1b) to evaluate bacterial killing, toxin neutralization, and protective efficacy in infant mice. All nine (100%) clinical strains of STEC from different serogroups expressed PNAG. Vaccine-induced antibody mediated in vitro killing of STEC and neutralization of both Stx1 and Stx2. Passive administration of antibody to the conjugate showed protection requiring immunity to both PNAG and Stx for O157 strains, although for an O104 strain, antibody to PNAG alone was protective. Immunity to PNAG may contribute to protection against STEC infections.


Asunto(s)
Infecciones por Escherichia coli/prevención & control , Vacunas contra Escherichia coli/inmunología , Toxina Shiga/inmunología , Escherichia coli Shiga-Toxigénica/inmunología , beta-Glucanos/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Antibacterianos/sangre , Anticuerpos Neutralizantes/sangre , Antitoxinas/sangre , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/inmunología , Vacunas contra Escherichia coli/administración & dosificación , Ratones , Viabilidad Microbiana/inmunología , Proteínas Opsoninas/sangre , Conejos , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/inmunología
12.
PLoS One ; 7(10): e46648, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077517

RESUMEN

Staphylococcus aureus is a major cause of nosocomial and community-acquired infections for which a vaccine is greatly desired. Antigens found on the S. aureus outer surface include the capsular polysaccharides (CP) of serotype 5 (CP5) or 8 (CP8) and/or a second antigen, a ß-(1→6)-polymer of N-acetyl-D-glucosamine (PNAG). Antibodies specific for either CP or PNAG antigens have excellent in vitro opsonic killing activity (OPKA), but when mixed together have potent interference in OPKA and murine protection. To ascertain if this interference could be abrogated by using a synthetic non-acetylated oligosaccharide fragment of PNAG, 9GlcNH(2), in place of chemically partially deacetylated PNAG, three conjugate vaccines consisting of 9GlcNH(2) conjugated to a non-toxic mutant of alpha-hemolysin (Hla H35L), CP5 conjugated to clumping factor B (ClfB), or CP8 conjugated to iron-surface determinant B (IsdB) were used separately to immunize rabbits. Opsonic antibodies mediating killing of multiple S. aureus strains were elicited for all three vaccines and showed carbohydrate antigen-specific reductions in the tissue bacterial burdens in animal models of S. aureus skin abscesses, pneumonia, and nasal colonization. Carrier-protein specific immunity was also shown to be effective in reducing bacterial levels in infected lungs and in nasal colonization. However, use of synthetic 9GlcNH(2) to induce antibody to PNAG did not overcome the interference in OPKA engendered when these were combined with antibody to either CP5 or CP8. Whereas each individual vaccine showed efficacy, combining antisera to CP antigens and PNAG still abrogated individual OPKA activities, indicating difficulty in achieving a multi-valent vaccine targeting both the CP and PNAG antigens.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Staphylococcus aureus/inmunología , Animales , Adhesión Bacteriana , Secuencia de Bases , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Ratones , Nariz/microbiología , Staphylococcus aureus/fisiología
14.
ISME J ; 4(4): 577-84, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20072161

RESUMEN

The prevalence of diverse MRSA (methicillin-resistant Staphylococcus aureus) types in both hospital and community settings is a major health problem worldwide. Here we compare hospital-acquired MRSAs with large type II SCCmec elements with those prevalent in both hospital and community settings with smaller type IV SCCmec elements. We find that the type II but not the type IV SCCmec element causes the bacteria to reduce their levels of costly toxin expression. We compare the relative growth rates of these MRSA types and show that the type II SCCmec carrying MRSAs are more affected than those carrying type IV elements and from this we hypothesize that offsetting the costs associated with antibiotic resistance and toxin expression is why the type II are confined to hospital environments where antibiotic use, the prevalence of immunocompromised individuals and vector-mediated transmission is high. In contrast, those MRSAs that are also successful in the community can maintain their high levels of toxin expression due to a lower fitness burden associated with the smaller SCCmec element.


Asunto(s)
Farmacorresistencia Bacteriana , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Infecciones Comunitarias Adquiridas/microbiología , Infección Hospitalaria/microbiología , ADN Bacteriano/genética , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Infecciones Estafilocócicas/microbiología , Virulencia , Factores de Virulencia/genética
15.
J Clin Microbiol ; 45(5): 1379-88, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17329452

RESUMEN

Production of icaADBC-encoded polysaccharide intercellular adhesin, or poly-N-acetylglucosamine (PIA/PNAG), represents an important biofilm mechanism in staphylococci. We previously described a glucose-induced, ica-independent biofilm mechanism in four methicillin-resistant Staphylococcus aureus (MRSA) isolates. Here, biofilm regulation by NaCl and glucose was characterized in 114 MRSA and 98 methicillin-sensitive S. aureus (MSSA) isolates from diagnosed device-related infections. NaCl-induced biofilm development was significantly more prevalent among MSSA than MRSA isolates, and this association was independent of the isolate's genetic background as assessed by spa sequence typing. Among MSSA isolates, PIA/PNAG production correlated with biofilm development in NaCl, whereas in MRSA isolates grown in NaCl or glucose, PIA/PNAG production was not detected even though icaADBC was transcribed and regulated. Glucose-induced biofilm in MRSA was ica independent and apparently mediated by a protein adhesin(s). Experiments performed with strains that were amenable to genetic manipulation revealed that deletion of icaADBC had no effect on biofilm in a further six MRSA isolates but abolished biofilm in four MSSA isolates. Mutation of sarA abolished biofilm in seven MRSA and eight MSSA isolates. In contrast, mutation of agr in 13 MRSA and 8 MSSA isolates substantially increased biofilm (more than twofold) in only 5 of 21 (23%) isolates and had no significant impact on biofilm in the remaining 16 isolates. We conclude that biofilm development in MRSA is ica independent and involves a protein adhesin(s) regulated by SarA and Agr, whereas SarA-regulated PIA/PNAG plays a more important role in MSSA biofilm development.


Asunto(s)
Biopelículas , Contaminación de Equipos , Equipos y Suministros/efectos adversos , Resistencia a la Meticilina , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Mutación , Staphylococcus aureus/genética , Staphylococcus aureus/fisiología , Transactivadores/genética , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA