Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 223(12 Suppl 2): S257-S263, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33330904

RESUMEN

Decades of research have failed to define the pathophysiology of necrotizing enterocolitis (NEC), a devastating pediatric gastrointestinal disorder of preterm infants. However, evidence suggests that host-microbiota interactions, in which microbial dysbiosis is followed by loss of barrier integrity, inflammation, and necrosis, are central to NEC development. Thus, greater knowledge of the preterm infant microbiome could accelerate attempts to diagnose, treat, and prevent NEC. In this article, we summarize clinical characteristics of and risk factors for NEC, the structure of the pre-event NEC microbiome, how this community interfaces with host immunology, and microbiome-based approaches that might prevent or lessen the severity of NEC in this very vulnerable population.


Asunto(s)
Enterocolitis Necrotizante/microbiología , Microbioma Gastrointestinal/fisiología , Disbiosis/microbiología , Enterocolitis Necrotizante/terapia , Interacciones Huésped-Patógeno , Humanos , Recién Nacido , Recien Nacido Prematuro , Factores de Riesgo
2.
Angew Chem Int Ed Engl ; 56(49): 15746-15750, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-28906057

RESUMEN

The Staphylococcus aureus ClpXP protease is an important regulator of cell homeostasis and virulence. We utilized a high-throughput screen against the ClpXP complex and identified a specific inhibitor of the ClpX chaperone that disrupts its oligomeric state. Synthesis of 34 derivatives revealed that the molecular scaffold is restrictive for diversification, with only minor changes tolerated. Subsequent analysis of the most active compound revealed strong attenuation of S. aureus toxin production, which was quantified with a customized MS-based assay platform. Transcriptome and whole-proteome studies further confirmed the global reduction of virulence and revealed characteristic signatures of protein expression in the compound-treated cells. Although these partially matched the pattern of ClpX knockout cells, further depletion of toxins was observed, leading to the intriguing perspective that additional virulence pathways may be directly or indirectly addressed by the small molecule.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Endopeptidasa Clp/antagonistas & inhibidores , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Inhibidores de Proteasas/farmacología , Proteínas Bacterianas/metabolismo , Relación Dosis-Respuesta a Droga , Endopeptidasa Clp/deficiencia , Endopeptidasa Clp/metabolismo , Ensayos Analíticos de Alto Rendimiento , Staphylococcus aureus Resistente a Meticilina/metabolismo , Estructura Molecular , Inhibidores de Proteasas/química , Relación Estructura-Actividad , Virulencia
3.
Mol Biol Evol ; 32(11): 2883-96, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26199376

RESUMEN

Symbiotic interactions are indispensable for metazoan function, but their origin and evolution remain elusive. We use a controlled evolution experiment to demonstrate the emergence of novel commensal interactions between Pseudomonas aeruginosa, an initially pathogenic bacterium, and a metazoan host, Caenorhabditis elegans. We show that commensalism evolves through loss of virulence, because it provides bacteria with a double fitness advantage: Increased within-host fitness and a larger host population to infect. Commensalism arises irrespective of host immune status, as the adaptive path in immunocompromised C. elegans knockouts does not differ from that in wild type. Dissection of temporal dynamics of genomic adaptation for 125 bacterial populations reveals highly parallel evolution of incipient commensalism across independent biological replicates. Adaptation is mainly achieved through frame shift mutations in the global regulator lasR and nonsynonymous point mutations in the polymerase gene rpoB that arise early in evolution. Genetic knockouts of lasR not only corroborate its role in virulence attenuation but also show that further mutations are necessary for the fully commensal phenotype. The evolutionary transition from pathogenicity to commensalism as we observe here is facilitated by mutations in global regulators such as lasR, because few genetic changes cause pleiotropic effects across the genome with large phenotypic effects. Finally, we found that nucleotide diversity increased more quickly in bacteria adapting to immunocompromised hosts than in those adapting to immunocompetent hosts. Nevertheless, the outcome of evolution was comparable across host types. Commensalism can thus evolve independently of host immune state solely as a side-effect of bacterial adaptation to novel hosts.


Asunto(s)
Adaptación Fisiológica/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Animales , Proteínas Bacterianas , Evolución Biológica , Evolución Molecular , Regulación Bacteriana de la Expresión Génica , Estudios de Asociación Genética , Variación Genética , Genoma Bacteriano , Interacciones Huésped-Patógeno , Mutación , Simbiosis/genética , Virulencia
4.
EClinicalMedicine ; 71: 102490, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38813445

RESUMEN

Background: Urinary tract infections (UTI) affect approximately 250 million people annually worldwide. Patients often experience a cycle of antimicrobial treatment and recurrent UTI (rUTI) that is thought to be facilitated by a gut reservoir of uropathogenic Escherichia coli (UPEC). Methods: 125 patients with UTI caused by an antibiotic-resistant organism (ARO) were enrolled from July 2016 to May 2019 in a longitudinal, multi-center cohort study. Multivariate statistical models were used to assess the relationship between uropathogen colonization and recurrent UTI (rUTI), controlling for clinical characteristics. 644 stool samples and 895 UPEC isolates were interrogated for taxonomic composition, antimicrobial resistance genes, and phenotypic resistance. Cohort UTI gut microbiome profiles were compared against published healthy and UTI reference microbiomes, as well as assessed within-cohort for timepoint- and recurrence-specific differences. Findings: Risk of rUTI was not independently associated with clinical characteristics. The UTI gut microbiome was distinct from healthy reference microbiomes in both taxonomic composition and antimicrobial resistance gene (ARG) burden, with 11 differentially abundant taxa at the genus level. rUTI and non-rUTI gut microbiomes in the cohort did not generally differ, but gut microbiomes from urinary tract colonized patients were elevated in E. coli abundance 7-14 days post-antimicrobial treatment. Corresponding UPEC gut isolates from urinary tract colonizing lineages showed elevated phenotypic resistance against 11 of 23 tested drugs compared to non-colonizing lineages. Interpretation: The gut microbiome is implicated in UPEC urinary tract colonization during rUTI, serving as an ARG-enriched reservoir for UPEC. UPEC can asymptomatically colonize the gut and urinary tract, and post-antimicrobial blooms of gut E. coli among urinary tract colonized patients suggest that cross-habitat migration of UPEC is an important mechanism of rUTI. Thus, treatment duration and UPEC populations in both the urinary and gastrointestinal tract should be considered in treating rUTI and developing novel therapeutics. Funding: This work was supported in part by awards from the U.S. Centers for Disease Control and Prevention Epicenter Prevention Program (grant U54CK000482; principal investigator, V.J.F.); to J.H.K. from the Longer Life Foundation (an RGA/Washington University partnership), the National Center for Advancing Translational Sciences (grants KL2TR002346 and UL1TR002345), and the National Institute of Allergy and Infectious Diseases (NIAID) (grant K23A1137321) of the National Institutes of Health (NIH); and to G.D. from NIAID (grant R01AI123394) and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (grant R01HD092414) of NIH. R.T.'s research was funded by the Deutsche Forschungsgemeinschaft (DFG; German Research Foundation; grant 402733540). REDCap is Supported by Clinical and Translational Science Award (CTSA) Grant UL1 TR002345 and Siteman Comprehensive Cancer Center and NCI Cancer Center Support Grant P30 CA091842. The content is solely the responsibility of the authors and does not necessarily represent the official views of the funding agencies.

5.
Cell Host Microbe ; 30(5): 675-683, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35550670

RESUMEN

Intestinal host-microbiota interactions during the first year of life are critical for infant development. Early-life antibiotic exposures disrupt stereotypical gut microbiota maturation and adversely affect childhood health. Furthermore, antibiotics increase the abundance of resistant bacteria and enrich the resistome-the compendium of antibiotic resistance genes-within the gut microbiota. Here, we discuss acute and persistent impacts of antibiotic exposure during infancy on pediatric health, the gut microbiome, and, particularly, the resistome. Reviewing our current understanding of antibiotic resistance acquisition and dissemination within and between microbiomes, we highlight open questions, which are imperative to resolve in the face of rising bacterial resistance.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacterias/genética , Niño , Farmacorresistencia Microbiana , Humanos , Lactante
6.
Microbiome ; 10(1): 6, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35039079

RESUMEN

BACKGROUND: The lack of a definition of urinary microbiome health convolutes diagnosis of urinary tract infections (UTIs), especially when non-traditional uropathogens or paucity of bacteria are recovered from symptomatic patients in routine standard-of-care urine tests. Here, we used shotgun metagenomic sequencing to characterize the microbial composition of asymptomatic volunteers in a set of 30 longitudinally collected urine specimens. Using permutation tests, we established a range of asymptomatic microbiota states, and use these to contextualize the microbiota of 122 urine specimens collected from patients with suspected UTIs diagnostically categorized by standard-of-care urinalysis within that range. Finally, we used a standard-of-care culture protocol to evaluate the efficiency of culture-based recovery of the urinary microbiota. RESULTS: The majority of genitourinary microbiota in individals suspected to have UTI overlapped with the spectrum of asymptomatic microbiota states. Longitudinal characterization of the genitourinary microbiome in urine specimens collected from asymptomatic volunteers revealed fluctuations of microbial functions and taxonomy over time. White blood cell counts from urinalysis suggested that urine specimens categorized as 'insignificant', 'contaminated', or 'no-growth' by conventional culture methods frequently showed signs of urinary tract inflammation, but this inflammation is not associated with genitourinary microbiota dysbiosis. Comparison of directly sequenced urine specimens with standard-of-care culturing confirmed that culture-based diagnosis biases genitourinary microbiota recovery towards the traditional uropathogens Escherichia coli and Klebsiella pneumoniae. CONCLUSION: Here, we utilize shotgun metagenomic sequencing to establish a baseline of asymptomatic genitourinary microbiota states. Using this baseline we establish substantial overlap between symptomatic and asymptomatic genitourinary microbiota states. Our results establish that bacterial presence alone does not explain the onset of clinical symptoms. Video Abstract.


Asunto(s)
Microbiota , Infecciones Urinarias , Disbiosis , Humanos , Metagenoma , Metagenómica , Microbiota/genética , Infecciones Urinarias/diagnóstico , Infecciones Urinarias/microbiología
7.
Cell Host Microbe ; 30(7): 1034-1047.e6, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35545083

RESUMEN

Large-scale genomic studies have identified within-host adaptation as a hallmark of bacterial infections. However, the impact of physiological, metabolic, and immunological differences between distinct niches on the pathoadaptation of opportunistic pathogens remains elusive. Here, we profile the within-host adaptation and evolutionary trajectories of 976 isolates representing 119 lineages of uropathogenic Escherichia coli (UPEC) sampled longitudinally from both the gastrointestinal and urinary tracts of 123 patients with urinary tract infections. We show that lineages persisting in both niches within a patient exhibit increased allelic diversity. Habitat-specific selection results in niche-specific adaptive mutations and genes, putatively mediating fitness in either environment. Within-lineage inter-habitat genomic plasticity mediated by mobile genetic elements (MGEs) provides the opportunistic pathogen with a mechanism to adapt to the physiological conditions of either habitat, and reduced MGE richness is associated with recurrence in gut-adapted UPEC lineages. Collectively, our results establish niche-specific adaptation as a driver of UPEC within-host evolution.


Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Adaptación al Huésped , Infecciones Urinarias , Escherichia coli Uropatógena , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Adaptación al Huésped/genética , Humanos , Secuencias Repetitivas Esparcidas , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/genética
8.
Gut Microbes ; 13(1): 1940792, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34264786

RESUMEN

Surgical removal of the intestine, lifesaving in catastrophic gastrointestinal disorders of infancy, can result in a form of intestinal failure known as short bowel syndrome (SBS). Bloodstream infections (BSIs) are a major challenge in pediatric SBS management. BSIs require frequent antibiotic therapy, with ill-defined consequences for the gut microbiome and childhood health. Here, we combine serial stool collection, shotgun metagenomic sequencing, multivariate statistics and genome-resolved strain-tracking in a cohort of 19 patients with surgically-induced SBS to show that antibiotic-driven intestinal dysbiosis in SBS enriches for persistent intestinal colonization with BSI causative pathogens in SBS. Comparing the gut microbiome composition of SBS patients over the first 4 years of life to 19 age-matched term and 18 preterm controls, we find that SBS gut microbiota diversity and composition was persistently altered compared to controls. Commensals including Ruminococcus, Bifidobacterium, Eubacterium, and Clostridium species were depleted in SBS, while pathobionts (Enterococcus) were enriched. Integrating clinical covariates with gut microbiome composition in pediatric SBS, we identified dietary and antibiotic exposures as the main drivers of these alterations. Moreover, antibiotic resistance genes, specifically broad-spectrum efflux pumps, were at a higher abundance in SBS, while putatively beneficial microbiota functions, including amino acid and vitamin biosynthesis, were depleted. Moreover, using strain-tracking we found that the SBS gut microbiome harbors BSI causing pathogens, which can persist intestinally throughout the first years of life. The association between antibiotic-driven gut dysbiosis and enrichment of intestinal pathobionts isolated from BSI suggests that antibiotic treatment may predispose SBS patients to infection. Persistence of pathobionts and depletion of beneficial microbiota and functionalities in SBS highlights the need for microbiota-targeted interventions to prevent infection and facilitate intestinal adaptation.


Asunto(s)
Antibacterianos/uso terapéutico , Disbiosis/tratamiento farmacológico , Disbiosis/etiología , Microbioma Gastrointestinal/efectos de los fármacos , Sepsis/tratamiento farmacológico , Sepsis/etiología , Síndrome del Intestino Corto/complicaciones , Adolescente , Niño , Preescolar , Estudios de Cohortes , Disbiosis/microbiología , Femenino , Humanos , Masculino , Missouri , Síndrome del Intestino Corto/microbiología
9.
Int J Antimicrob Agents ; 55(2): 105864, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31870598

RESUMEN

OBJECTIVES: To evaluate the activity of the reported synergistic and collaterally sensitive antibiotic combination, meropenem/piperacillin/tazobactam (ME/PI/TZ), against a panel of methicillin-resistant Staphylococcus aureus (MRSA) and other methicillin-resistant Staphylococcus species; and to investigate the relationship between ME/PI/TZ susceptibility and the genomic background of clinical isolates of MRSA. METHODS: ME/PI/TZ combination and single drug minimum inhibitory concentrations (MICs) were determined for 207 strains (including 121 MRSA, 4 methicillin-sensitive S. aureus [MSSA], 37 vancomycin-intermediate S. aureus [VISA], 6 ceftaroline non-susceptible MRSA, 29 coagulase-negative staphylococci [CoNS], 5 S. pseudointermedius and 5 vancomycin-resistant Enterococci [VRE]) by broth microdilution. Whole genomes of 168 S. aureus strains were sequenced, assembled, and comparatively analysed. RESULTS: USA300-SCCmec type IV isolates, clonal complex 8 (CC8)-MRSA isolates, including some VISA and ceftaroline (CPT)-intermediate strains, and all tested methicillin-resistant S. epidermidis isolates were highly susceptible to ME/PI/TZ. Isolates with elevated MICs (MICs of >16/16/16 mg/L) clustered with the USA100-SCCmec type II strain. Susceptibility of MRSA to ME/PI/TZ was correlated with susceptibility to ME. No obvious cross-resistance to CPT was observed among high-ME/PI/TZ MIC isolates. CONCLUSIONS: The ME/PI/TZ combination is effective against a variety of clinical MRSA isolates, particularly of the USA300 lineage, which is expanding worldwide. ME/PI/TZ is also effective against drug-resistant CoNS and S. pseudintermedius clinical isolates.


Asunto(s)
Antibacterianos/uso terapéutico , Meropenem/uso terapéutico , Piperacilina/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Tazobactam/uso terapéutico , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Quimioterapia Combinada , Genoma Bacteriano/genética , Genómica , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Pruebas de Sensibilidad Microbiana , Staphylococcus/efectos de los fármacos , Staphylococcus/genética , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/genética , Enterococos Resistentes a la Vancomicina/genética , Secuenciación Completa del Genoma , Inhibidores de beta-Lactamasas/uso terapéutico
10.
Nat Commun ; 10(1): 3846, 2019 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-31451691

RESUMEN

Necrotizing soft tissue infections (NSTIs) are devastating infections caused by either a single pathogen, predominantly Streptococcus pyogenes, or by multiple bacterial species. A better understanding of the pathogenic mechanisms underlying these different NSTI types could facilitate faster diagnostic and more effective therapeutic strategies. Here, we integrate microbial community profiling with host and pathogen(s) transcriptional analysis in patient biopsies to dissect the pathophysiology of streptococcal and polymicrobial NSTIs. We observe that the pathogenicity of polymicrobial communities is mediated by synergistic interactions between community members, fueling a cycle of bacterial colonization and inflammatory tissue destruction. In S. pyogenes NSTIs, expression of specialized virulence factors underlies infection pathophysiology. Furthermore, we identify a strong interferon-related response specific to S. pyogenes NSTIs that could be exploited as a potential diagnostic biomarker. Our study provides insights into the pathophysiology of mono- and polymicrobial NSTIs and highlights the potential of host-derived signatures for microbial diagnosis of NSTIs.


Asunto(s)
Coinfección/patología , Infecciones de los Tejidos Blandos/patología , Infecciones Estreptocócicas/patología , Factores de Virulencia/metabolismo , Adulto , Anciano , Técnicas de Tipificación Bacteriana , Bacteroides/genética , Bacteroides/aislamiento & purificación , Bacteroides/metabolismo , Biopsia , Coinfección/diagnóstico , Coinfección/microbiología , ADN Bacteriano/aislamiento & purificación , Escherichia/genética , Escherichia/aislamiento & purificación , Escherichia/metabolismo , Femenino , Humanos , Masculino , Microbiota/genética , Persona de Mediana Edad , Necrosis/diagnóstico , Necrosis/microbiología , Necrosis/patología , ARN Ribosómico 16S/genética , RNA-Seq , Infecciones de los Tejidos Blandos/diagnóstico , Infecciones de los Tejidos Blandos/microbiología , Staphylococcus/genética , Staphylococcus/aislamiento & purificación , Staphylococcus/metabolismo , Infecciones Estreptocócicas/diagnóstico , Infecciones Estreptocócicas/microbiología , Streptococcus/genética , Streptococcus/aislamiento & purificación , Streptococcus/metabolismo , Factores de Virulencia/genética
11.
mBio ; 10(4)2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455657

RESUMEN

The rise of antimicrobial resistance in uropathogens has complicated the management of urinary tract infections (UTIs), particularly in patients who are afflicted by recurrent episodes of UTIs. Antimicrobial-resistant (AR) uropathogens persistently colonizing individuals at asymptomatic time points have been implicated in the pathophysiology of UTIs. The dynamics of uropathogen persistence following the resolution of symptomatic disease are, however, mostly unclear. To further our understanding, we determined longitudinal AR uropathogen carriage and clonal persistence of uropathogenic Escherichia coli, Proteus mirabilis, and Klebsiella pneumoniae isolates in the intestinal and urinary tracts of patients affected by recurrent and nonrecurrent UTIs. Clonal tracking of isolates in consecutively collected urine and fecal specimens indicated repeated transmission of uropathogens between the urinary tract and their intestinal reservoir. Our results further implicate three independent routes of recurrence of UTIs: (i) following an intestinal bloom of uropathogenic bacteria and subsequent bladder colonization, (ii) reinfection of the urinary tract from an external source, and (iii) bacterial persistence within the urinary tract. Taken together, our observation of clonal persistence following UTIs and uropathogen transmission between the intestinal and urinary tracts warrants further investigations into the connection between the intestinal microbiome and recurrent UTIs.IMPORTANCE The increasing antimicrobial resistance of uropathogens is challenging the continued efficacy of empiric antibiotic therapy for UTIs, which are among the most frequent bacterial infections worldwide. It has been suggested that drug-resistant uropathogens could persist in the intestine after the resolution of UTI and cause recurrences following periurethral contamination. A better understanding of the transmission dynamics between the intestinal and urinary tracts, combined with phenotypic characterization of the uropathogen populations in both habitats, could inform prudent therapies designed to overcome the rising resistance of uropathogens. Here, we integrate genomic surveillance with clinical microbiology to show that drug-resistant clones persist within and are readily transmitted between the intestinal and urinary tracts of patients affected by recurrent and nonrecurrent UTIs. Thus, our results advocate for understanding persistent intestinal uropathogen colonization as part of the pathophysiology of UTIs, particularly in patients affected by recurrent episodes of symptomatic disease.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/genética , Genómica , Klebsiella pneumoniae/genética , Proteus mirabilis/genética , Infecciones Urinarias/microbiología , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Escherichia coli/efectos de los fármacos , Humanos , Klebsiella pneumoniae/efectos de los fármacos , Estudios Longitudinales , Persona de Mediana Edad , Filogenia , Proteus mirabilis/efectos de los fármacos , Recurrencia
12.
Nat Commun ; 8: 14268, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28155859

RESUMEN

The rise of antibiotic resistance calls for alternative strategies to treat bacterial infections. One attractive strategy is to directly target bacterial virulence factors with anti-virulence drugs. The expression of virulence traits by pathogens is, however, not constitutive but rather induced by the level of stress encountered within the host. Here we use dual RNA sequencing (RNA-seq) to show that intrinsic variability in the level of host resistance greatly affects the pathogen's transcriptome in vivo. Through analysis of the transcriptional profiles of host and pathogen during Staphylococcus aureus infection of two mouse strains, shown to be susceptible (A/J) or resistant (C57BL/6) to the pathogen, we demonstrate that the expression of virulence factors is dependent on the encountered host resistance. We furthermore provide evidence that this dependence strongly influences the efficacy of anti-virulence strategies, highlighting a potential limitation for the implementation of these strategies.


Asunto(s)
Interacciones Huésped-Patógeno , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Transcripción Genética , Animales , Antibacterianos/farmacología , Farmacorresistencia Microbiana , Ratones , Ratones Endogámicos C57BL , ARN Bacteriano/genética , Análisis de Secuencia de ARN , Especificidad de la Especie , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/efectos de los fármacos , Transcriptoma , Virulencia , Factores de Virulencia/metabolismo
13.
J Innate Immun ; 9(6): 561-573, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28858870

RESUMEN

Toll-like receptor (TLR) signaling is important in the initiation of immune responses and subsequent instigation of adaptive immunity. TLR2 recognizes bacterial lipoproteins and plays a central role in the host defense against bacterial infections, including those caused by Staphylococcus aureus. Many studies have demonstrated the importance of TLR2 in murine S. aureus infection. S. aureus evades TLR2 activation by secreting two proteins, staphylococcal superantigen-like protein 3 (SSL3) and 4 (SSL4). In this study, we demonstrate that antibodies against SSL3 and SSL4 are found in healthy individuals, indicating that humans are exposed to these proteins during S. aureus colonization or infection. To investigate the TLR2-antagonistic properties of SSL3 and SSL4, we compared the infection with wild-type and SSL3/4 knockout S. aureus strains in an intravenous murine infection model. Direct evaluation of the contribution of SSL3/4 to infection pathogenesis was hindered by the fact that the SSLs were not expressed in the murine system. To circumvent this limitation, an SSL3-overproducing strain (pLukM-SSL3) was generated, resulting in constitutive expression of SSL3. pLukM-SSL3 exhibited increased virulence compared to the parental strain in a murine model that was found to be TLR2 dependent. Altogether, these data indicate that SSL3 contributes to S. aureus virulence in vivo.


Asunto(s)
Proteínas Bacterianas/inmunología , Exotoxinas/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Receptor Toll-Like 2/metabolismo , Factores de Virulencia/inmunología , Animales , Proteínas Bacterianas/genética , Células Cultivadas , Exotoxinas/genética , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal , Staphylococcus aureus/patogenicidad , Receptor Toll-Like 2/antagonistas & inhibidores , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA