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










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 19(9): e1011666, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37733817

RESUMEN

Prior infection can generate protective immunity against subsequent infection, although the efficacy of such immunity can vary considerably. Live-attenuated vaccines (LAVs) are one of the most effective methods for mimicking this natural process, and analysis of their efficacy has proven instrumental in the identification of protective immune mechanisms. Here, we address the question of what makes a LAV efficacious by characterising immune responses to a LAV, termed TAS2010, which is highly protective (80-90%) against lethal murine salmonellosis, in comparison with a moderately protective (40-50%) LAV, BRD509. Mice vaccinated with TAS2010 developed immunity systemically and were protected against gut-associated virulent infection in a CD4+ T cell-dependent manner. TAS2010-vaccinated mice showed increased activation of Th1 responses compared with their BRD509-vaccinated counterparts, leading to increased Th1 memory populations in both lymphoid and non-lymphoid organs. The optimal development of Th1-driven immunity was closely correlated with the activation of CD11b+Ly6GnegLy6Chi inflammatory monocytes (IMs), the activation of which can be modulated proportionally by bacterial load in vivo. Upon vaccination with the LAV, IMs expressed T cell chemoattractant CXCL9 that attracted CD4+ T cells to the foci of infection, where IMs also served as a potent source of antigen presentation and Th1-promoting cytokine IL-12. The expression of MHC-II in IMs was rapidly upregulated following vaccination and then maintained at an elevated level in immune mice, suggesting IMs may have a role in sustained antigen stimulation. Our findings present a longitudinal analysis of CD4+ T cell development post-vaccination with an intracellular bacterial LAV, and highlight the benefit of inflammation in the development of Th1 immunity. Future studies focusing on the induction of IMs may reveal key strategies for improving vaccine-induced T cell immunity.


Asunto(s)
Linfocitos T CD4-Positivos , Infecciones por Salmonella , Ratones , Animales , Monocitos , Vacunas Atenuadas , Inflamación
2.
PLoS Pathog ; 17(10): e1010004, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34695149

RESUMEN

While Salmonella enterica is seen as an archetypal facultative intracellular bacterial pathogen where protection is mediated by CD4+ T cells, identifying circulating protective cells has proved very difficult, inhibiting steps to identify key antigen specificities. Exploiting a mouse model of vaccination, we show that the spleens of C57BL/6 mice vaccinated with live-attenuated Salmonella serovar Typhimurium (S. Typhimurium) strains carried a pool of IFN-γ+ CD4+ T cells that could adoptively transfer protection, but only transiently. Circulating Salmonella-reactive CD4+ T cells expressed the liver-homing chemokine receptor CXCR6, accumulated over time in the liver and assumed phenotypic characteristics associated with tissue-associated T cells. Liver memory CD4+ T cells showed TCR selection bias and their accumulation in the liver could be inhibited by blocking CXCL16. These data showed that the circulation of CD4+ T cells mediating immunity to Salmonella is limited to a brief window after which Salmonella-specific CD4+ T cells migrate to peripheral tissues. Our observations highlight the importance of triggering tissue-specific immunity against systemic infections.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica/inmunología , Hígado/inmunología , Salmonelosis Animal/inmunología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/inmunología
3.
Front Microbiol ; 12: 651488, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815340

RESUMEN

Enterotoxigenic E. coli (ETEC) is a common cause of diarrhea in children in low- and middle-income countries, and in travelers to these countries. ETEC is also an important cause of morbidity and premature mortality in piglets, calves, goat kids and lambs. The major virulence determinants of ETEC are enterotoxins and colonization factors, which enable the pathogen to colonize the small intestine and deliver enterotoxins, such as the heat-stable enterotoxins, STp and STh, to epithelial cells. Because most ETEC strains are host-specific, there are few convenient animal models to investigate the pathogenesis of ETEC infections or to evaluate specific anti-ETEC interventions, such as drugs and vaccines. An exception is ETEC strains bearing F41 pili, which mediate intestinal colonization of various young animals, including neonatal mice, to cause disease and in some cases death. In this study, we used the archetypal F41-producing bovine ETEC strain, B41 (O101:NM; K99, F41, STp) to validate and further explore the contribution of F41 and STp to bacterial virulence. By using targeted gene deletion and trans-complementation studies, augmented by whole genome sequencing, and in vitro and animal studies of virulence, we established that F41 mediates colonization of the mouse intestine and is essential for bacterial virulence. In addition, we showed for the first time that STp is as important as F41 for virulence. Together, these findings validate the use of neonatal mice to study the pathogenesis of F41-bearing ETEC and to investigate possible specific anti-ETEC interventions including vaccines that target heat-stable enterotoxins.

4.
Risk Manag Healthc Policy ; 14: 247-252, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33519249

RESUMEN

This work is part of a project on the development of a smart prefabricated sanitising chamber (SPSC) to provide extra measures against the transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Stabilised hypochlorous acid (HOCl) is an approved disinfectant against SARS-CoV-2 by the Environmental Protection Association US in its liquid form on non-porous surfaces. This review is extended to cover its viricidal/bactericidal efficacy in aerosolised or sprayed form which showed an effective dose of as low as 20 ppm and the exposure duration of at least 60 s. The aerosolised application was also recommended with particle size of less than 200 µm to increase the contact with pathogens. The review also includes the safety and toxicity of HOCl with different concentrations. The review calls for more investigations into the effect of HOCl in mist and fog form on the respiratory system when transitioning through the proposed SPSC.

5.
J Am Chem Soc ; 143(1): 286-293, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33373526

RESUMEN

Microbes employ a remarkably intricate electron transport system to extract energy from the environment. The respiratory cascade of bacteria culminates in the terminal transfer of electrons onto higher redox potential acceptors in the extracellular space. This general and inducible mechanism of electron efflux during normal bacterial proliferation leads to a characteristic fall in bulk redox potential (Eh), the degree of which is dependent on growth phase, the microbial taxa, and their physiology. Here, we show that the general reducing power of bacteria can be subverted to induce the abiotic production of a carbon-centered radical species for targeted bioorthogonal molecular synthesis. Using two species, Escherichia coli and Salmonella enterica serovar Typhimurium as model microbes, a common redox active aryldiazonium salt is employed to intervene in the terminal respiratory electron flow, affording radical production that is mediated by native redox-active molecular shuttles and active bacterial metabolism. The aryl radicals are harnessed to initiate and sustain a bioorthogonal controlled radical polymerization via reversible addition-fragmentation chain transfer (BacRAFT), yielding a synthetic extracellular matrix of "living" vinyl polymers with predetermined molecular weight and low dispersity. The ability to interface the ubiquitous reducing power of bacteria into synthetic materials design offers a new means for creating engineered living materials with promising adaptive and self-regenerative capabilities.


Asunto(s)
Transporte de Electrón/fisiología , Escherichia coli/metabolismo , Radicales Libres/metabolismo , Ácidos Polimetacrílicos/metabolismo , Salmonella typhimurium/metabolismo , Compuestos Azo/química , Compuestos Azo/metabolismo , Radicales Libres/química , Metacrilatos/química , Metacrilatos/metabolismo , Oxidación-Reducción , Polimerizacion
6.
Sci Rep ; 10(1): 15009, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32929135

RESUMEN

A neural reflex mediated by the splanchnic sympathetic nerves regulates systemic inflammation in negative feedback fashion, but its consequences for host responses to live infection are unknown. To test this, conscious instrumented sheep were infected intravenously with live E. coli bacteria and followed for 48 h. A month previously, animals had undergone either bilateral splanchnic nerve section or a sham operation. As established for rodents, sheep with cut splanchnic nerves mounted a stronger systemic inflammatory response: higher blood levels of tumor necrosis factor alpha and interleukin-6 but lower levels of the anti-inflammatory cytokine interleukin-10, compared with sham-operated animals. Sequential blood cultures revealed that most sham-operated sheep maintained high circulating levels of live E. coli throughout the 48-h study period, while all sheep without splanchnic nerves rapidly cleared their bacteraemia and recovered clinically. The sympathetic inflammatory reflex evidently has a profound influence on the clearance of systemic bacterial infection.


Asunto(s)
Bacteriemia/fisiopatología , Nervios Esplácnicos/fisiología , Sistema Nervioso Simpático , Animales , Presión Arterial , Bacteriemia/sangre , Bacteriemia/microbiología , Carga Bacteriana , Catecolaminas/sangre , Citocinas/sangre , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/fisiopatología , Femenino , Reflejo/fisiología , Ovinos , Nervios Esplácnicos/cirugía , Sistema Nervioso Simpático/microbiología , Sistema Nervioso Simpático/fisiología
7.
Biochem J ; 476(22): 3435-3453, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31675053

RESUMEN

Key physiological differences between bacterial and mammalian metabolism provide opportunities for the development of novel antimicrobials. We examined the role of the multifunctional enzyme S-adenosylhomocysteine/Methylthioadenosine (SAH/MTA) nucleosidase (Pfs) in the virulence of S. enterica var Typhimurium (S. Typhimurium) in mice, using a defined Pfs deletion mutant (i.e. Δpfs). Pfs was essential for growth of S. Typhimurium in M9 minimal medium, in tissue cultured cells, and in mice. Studies to resolve which of the three known functions of Pfs were key to murine virulence suggested that downstream production of autoinducer-2, spermidine and methylthioribose were non-essential for Salmonella virulence in a highly sensitive murine model. Mass spectrometry revealed the accumulation of SAH in S. Typhimurium Δpfs and complementation of the Pfs mutant with the specific SAH hydrolase from Legionella pneumophila reduced SAH levels, fully restored growth ex vivo and the virulence of S. Typhimurium Δpfs for mice. The data suggest that Pfs may be a legitimate target for antimicrobial development, and that the key role of Pfs in bacterial virulence may be in reducing the toxic accumulation of SAH which, in turn, suppresses an undefined methyltransferase.


Asunto(s)
Proteínas Bacterianas/metabolismo , N-Glicosil Hidrolasas/metabolismo , Purina-Nucleósido Fosforilasa/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/enzimología , Salmonella typhimurium/patogenicidad , Animales , Proteínas Bacterianas/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/metabolismo , N-Glicosil Hidrolasas/genética , Purina-Nucleósido Fosforilasa/genética , S-Adenosilhomocisteína/metabolismo , Salmonella typhimurium/genética , Virulencia
8.
Mol Med ; 25(1): 12, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30943912

RESUMEN

BACKGROUND: The polymeric immunoglobulin receptor (pIgR) maintains the integrity of epithelial barriers by transporting polymeric antibodies and antigens through the epithelial mucosa into the lumen. In this study, we examined the role of pIgR in maintaining gut barrier integrity, which is important for the normal development in mice. METHODS: Cohorts of pIgR-/- mice and their wildtype controls were housed under Specific Pathogen Free (SPF) conditions and monitored for weight gain as an indicator of development over time. The general physiology of the gastrointestinal tract was analysed using immunohistochemistry in young (8-12 weeks of age) and aged mice (up to 18 months of age), and the observed immunopathology in pIgR-/- mice was further characterised using flow cytometry. Urinary metabolites were analysed using gas chromatography-mass spectrometry (GC-MS), which revealed changes in metabolites that correlated with age-related increase in gut permeability in pIgR-/- mice. RESULTS: We observed that pIgR-/- mice exhibited delayed growth, and this phenomenon is associated with low-grade gut inflammation that increased with ageing. The gross intraepithelial lymphocytic (IEL) infiltration characteristic of pIgR-/- mice was redefined as CD8α+αß+ T cells, the majority of which expressed high levels of CD103 and CD69 consistent with tissue resident memory T cells (TRM). Comparison of the urinary metabolome between pIgR-/- and wild-type mice revealed key changes in urinary biomarkers fucose, glycine and Vitamin B5, suggestive of altered mucosal permeability. A significant increase in gut permeability was confirmed by analysing the site-specific uptake of sugar probes in different parts of the intestine. CONCLUSION: Our data show that loss of the secretory antibody system in mice results in enhanced accumulation of inflammatory IELs in the gut, which likely reflects ongoing inflammation in reaction to gut microbiota or food antigens, leading to delayed growth in pIgR-/- mice. We demonstrate that this leads to the presence of a unique urinary metabolome profile, which may provide a biomarker for altered gut permeability.


Asunto(s)
Tracto Gastrointestinal/inmunología , Linfocitos Intraepiteliales/inmunología , Metaboloma , Receptores de Inmunoglobulina Polimérica/genética , Orina/química , Animales , Anticuerpos/genética , Citocinas/sangre , Femenino , Tracto Gastrointestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
9.
J Biol Chem ; 293(24): 9506-9519, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29720401

RESUMEN

Methionine (Met) is an amino acid essential for many important cellular and biosynthetic functions, including the initiation of protein synthesis and S-adenosylmethionine-mediated methylation of proteins, RNA, and DNA. The de novo biosynthetic pathway of Met is well conserved across prokaryotes but absent from vertebrates, making it a plausible antimicrobial target. Using a systematic approach, we examined the essentiality of de novo methionine biosynthesis in Salmonella enterica serovar Typhimurium, a bacterial pathogen causing significant gastrointestinal and systemic diseases in humans and agricultural animals. Our data demonstrate that Met biosynthesis is essential for S. Typhimurium to grow in synthetic medium and within cultured epithelial cells where Met is depleted in the environment. During systemic infection of mice, the virulence of S. Typhimurium was not affected when either de novo Met biosynthesis or high-affinity Met transport was disrupted alone, but combined disruption in both led to severe in vivo growth attenuation, demonstrating a functional redundancy between de novo biosynthesis and acquisition as a mechanism of sourcing Met to support growth and virulence for S. Typhimurium during infection. In addition, our LC-MS analysis revealed global changes in the metabolome of S. Typhimurium mutants lacking Met biosynthesis and also uncovered unexpected interactions between Met and peptidoglycan biosynthesis. Together, this study highlights the complexity of the interactions between a single amino acid, Met, and other bacterial processes leading to virulence in the host and indicates that disrupting the de novo biosynthetic pathway alone is likely to be ineffective as an antimicrobial therapy against S. Typhimurium.


Asunto(s)
Metionina/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Animales , Transporte Biológico , Vías Biosintéticas , Femenino , Células HeLa , Humanos , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/metabolismo , Virulencia
10.
Front Microbiol ; 8: 1525, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28848532

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) is the most common bacterial cause of diarrhea in children in developing countries, as well as in travelers to these countries. To cause disease, ETEC needs to produce a series of virulence proteins including enterotoxins, colonization factors and secretion pathways, which enable this pathogen to colonize the human small intestine and deliver enterotoxins to epithelial cells. Previously, a number of studies have demonstrated that CfaD, an AraC-like transcriptional regulator, plays a key role in virulence gene expression by ETEC. In this study, we carried out a transcriptomic analysis of ETEC strain, H10407, grown under different conditions, and determined the complete set of genes that are regulated by CfaD. In this way, we identified a number of new target genes, including rnr-1, rnr-2, etpBAC, agn43, flu, traM and ETEC_3214, whose expression is strongly activated by CfaD. Using promoter-lacZ reporters, primer extension and electrophoretic mobility shift assays, we characterized the CfaD-mediated activation of several selected target promoters. We also showed that the gut-associated environmental signal, sodium bicarbonate, stimulates CfaD-mediated upregulation of its virulence target operons. Finally, we screened a commercial small molecule library and identified a compound (CH-1) that specifically inhibited the regulatory function of CfaD, and by 2-D analoging, we identified a second inhibitor (CH-2) with greater potency.

11.
Artículo en Inglés | MEDLINE | ID: mdl-27917373

RESUMEN

The empirical and pragmatic nature of diagnostic microbiology has given rise to several different schemes to subtype E.coli, including biotyping, serotyping, and pathotyping. These schemes have proved invaluable in identifying and tracking outbreaks, and for prognostication in individual cases of infection, but they are imprecise and potentially misleading due to the malleability and continuous evolution of E. coli. Whole genome sequencing can be used to accurately determine E. coli subtypes that are based on allelic variation or differences in gene content, such as serotyping and pathotyping. Whole genome sequencing also provides information about single nucleotide polymorphisms in the core genome of E. coli, which form the basis of sequence typing, and is more reliable than other systems for tracking the evolution and spread of individual strains. A typing scheme for E. coli based on genome sequences that includes elements of both the core and accessory genomes, should reduce typing anomalies and promote understanding of how different varieties of E. coli spread and cause disease. Such a scheme could also define pathotypes more precisely than current methods.


Asunto(s)
Proteínas de Escherichia coli/economía , Escherichia coli/clasificación , Escherichia coli/genética , Escherichia coli/patogenicidad , Proteínas de Unión al GTP/economía , Genoma Bacteriano , Tipificación Molecular/métodos , Proteínas de Unión al ARN/economía , Adhesinas Bacterianas/genética , Técnicas de Tipificación Bacteriana/métodos , Secuencia de Bases , ADN Bacteriano/genética , Diarrea/microbiología , Brotes de Enfermedades , Escherichia coli Enterohemorrágica/clasificación , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/aislamiento & purificación , Escherichia coli Enteropatógena/clasificación , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/aislamiento & purificación , Escherichia coli Enterotoxigénica/genética , Escherichia coli/aislamiento & purificación , Infecciones por Escherichia coli/microbiología , Evolución Molecular , Genes Bacterianos , Humanos , Filogenia , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Serotipificación/métodos , Virulencia/genética
12.
Nat Microbiol ; 1: 15010, 2016 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-27571974

RESUMEN

Atypical enteropathogenic Escherichia coli (aEPEC) is an umbrella term given to E. coli that possess a type III secretion system encoded in the locus of enterocyte effacement (LEE), but lack the virulence factors (stx, bfpA) that characterize enterohaemorrhagic E. coli and typical EPEC, respectively. The burden of disease caused by aEPEC has recently increased in industrialized and developing nations, yet the population structure and virulence profile of this emerging pathogen are poorly understood. Here, we generated whole-genome sequences of 185 aEPEC isolates collected during the Global Enteric Multicenter Study from seven study sites in Asia and Africa, and compared them with publicly available E. coli genomes. Phylogenomic analysis revealed ten distinct widely distributed aEPEC clones. Analysis of genetic variation in the LEE pathogenicity island identified 30 distinct LEE subtypes divided into three major lineages. Each LEE lineage demonstrated a preferred chromosomal insertion site and different complements of non-LEE encoded effector genes, indicating distinct patterns of evolution of these lineages. This study provides the first detailed genomic framework for aEPEC in the context of the EPEC pathotype and will facilitate further studies into the epidemiology and pathogenicity of EPEC by enabling the detection and tracking of specific clones and LEE variants.


Asunto(s)
Escherichia coli Enteropatógena/clasificación , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Evolución Molecular , Islas Genómicas , Genotipo , Fosfoproteínas/genética , África/epidemiología , Asia/epidemiología , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/microbiología , Variación Genética , Genoma Bacteriano , Filogenia , Análisis de Secuencia de ADN
13.
J Bacteriol ; 197(9): 1659-67, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25733612

RESUMEN

UNLABELLED: Klebsiella pneumoniae is an important cause of nosocomial infections, primarily through the formation of surface-associated biofilms to promote microbial colonization on host tissues. Expression of type 3 fimbriae by K. pneumoniae facilitates surface adherence, a process strongly activated by the cyclic di-GMP (c-di-GMP)-dependent transcriptional activator MrkH. In this study, we demonstrated the critical importance of MrkH in facilitating K. pneumoniae attachment on a variety of medically relevant materials and demonstrated the mechanism by which bacteria activate expression of type 3 fimbriae to colonize these materials. Sequence analysis revealed a putative MrkH recognition DNA sequence ("MrkH box"; TATCAA) located in the regulatory region of the mrkHI operon. Mutational analysis, electrophoretic mobility shift assay, and quantitative PCR experiments demonstrated that MrkH binds to the cognate DNA sequence to autoregulate mrkHI expression in a c-di-GMP-dependent manner. A half-turn deletion, but not a full-turn deletion, between the MrkH box and the -35 promoter element rendered MrkH ineffective in activating mrkHI expression, implying that a direct interaction between MrkH and RNA polymerase exists. In vivo analyses showed that residues L260, R265, N268, C269, E273, and I275 in the C-terminal domain of the RNA polymerase α subunit are involved in the positive control of mrkHI expression by MrkH and revealed the regions of MrkH required for DNA binding and transcriptional activation. Taken together, the data suggest a model whereby c-di-GMP-dependent MrkH recruits RNA polymerase to the mrkHI promoter to autoactivate mrkH expression. Increased MrkH production subsequently drives mrkABCDF expression when activated by c-di-GMP, leading to biosynthesis of type 3 fimbriae and biofilm formation. IMPORTANCE: Bacterial biofilms can cause persistent infections that are refractory to antimicrobial treatments. This study investigated how a commonly encountered hospital-acquired pathogen, Klebsiella pneumoniae, controls the expression of MrkH, the principal regulator of type 3 fimbriae and biofilm formation. We discovered a regulatory circuit whereby MrkH acts as a c-di-GMP-dependent transcriptional activator of both the gene cluster of type 3 fimbriae and the mrkHI operon. In this positive-feedback loop, whereby MrkH activates its own production, K. pneumoniae has evolved a mechanism to ensure rapid MrkH production, expression of type 3 fimbriae, and subsequent biofilm formation under favorable conditions. Deciphering the molecular mechanisms of biofilm formation by bacterial pathogens is important for the development of innovative treatment strategies for biofilm infections.


Asunto(s)
Adhesión Bacteriana , GMP Cíclico/análogos & derivados , Regulación Bacteriana de la Expresión Génica , Homeostasis , Klebsiella pneumoniae/fisiología , Factores de Transcripción/metabolismo , Sitios de Unión , Biopelículas/crecimiento & desarrollo , GMP Cíclico/metabolismo , Análisis Mutacional de ADN , ADN Bacteriano/química , ADN Bacteriano/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Fimbrias Bacterianas/fisiología , Perfilación de la Expresión Génica , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Operón , Regiones Promotoras Genéticas , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Factores de Transcripción/genética
14.
Infect Immun ; 83(1): 346-53, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25368119

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 causes bloody diarrhea and hemolytic-uremic syndrome (HUS) and is the most prevalent E. coli serotype associated with food-borne illness worldwide. This pathogen is transmitted via the fecal-oral route and has a low infectious dose that has been estimated to be between 10 and 100 cells. We and others have previously identified three prophage-encoded AraC-like transcriptional regulators, PatE, PsrA, and PsrB in the EHEC O157:H7 EDL933 strain. Our analysis showed that PatE plays an important role in facilitating survival of EHEC under a number of acidic conditions, but the contribution of PsrA and PsrB to acid resistance (AR) was unknown. Here, we investigated the involvement of PsrA and PsrB in the survival of E. coli O157:H7 in acid. Our results showed that PsrB, but not PsrA, enhanced the survival of strain EDL933 under various acidic conditions. Transcriptional analysis using promoter-lacZ reporters and electrophoretic mobility shift assays demonstrated that PsrB activates transcription of the hdeA operon, which encodes a major acid stress chaperone, by interacting with its promoter region. Furthermore, using a mouse model, we showed that expression of PsrB significantly enhanced the ability of strain EDL933 to overcome the acidic barrier of the mouse stomach. Taken together, our results indicate that EDL933 acquired enhanced acid tolerance via horizontally acquired regulatory genes encoding transcriptional regulators that activate its AR machinery.


Asunto(s)
Ácidos/toxicidad , Escherichia coli O157/efectos de los fármacos , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Profagos/genética , Estrés Fisiológico , Factores de Transcripción/metabolismo , Animales , Fusión Artificial Génica , ADN Bacteriano/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Escherichia coli O157/genética , Escherichia coli O157/fisiología , Escherichia coli O157/virología , Proteínas de Escherichia coli/genética , Perfilación de la Expresión Génica , Genes Reporteros , Ratones Endogámicos C57BL , Viabilidad Microbiana/efectos de los fármacos , Unión Proteica , Factores de Transcripción/genética , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
15.
Mater Sci Eng C Mater Biol Appl ; 47: 63-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25492173

RESUMEN

Bacterial infection associated with medical devices remains a challenge to modern medicine as more patients are being implanted with medical devices that provide surfaces and environment for bacteria colonization. In particular, bacteria are commonly found to adhere more preferably to hydrophobic materials and many of which are used to make medical devices. Bacteria are also becoming increasingly resistant to common antibiotic treatments as a result of misuse and abuse of antibiotics. There is an urgent need to find alternatives to antibiotics in the prevention and treatment of device-associated infections world-wide. Silver nanoparticles have emerged as a promising non-drug antimicrobial agent which has shown effectiveness against a wide range of both Gram-negative and Gram-positive pathogen. However, for silver nanoparticles to be clinically useful, they must be properly incorporated into medical device materials whose wetting properties could be detrimental to not only the incorporation of the hydrophilic Ag nanoparticles but also the release of active Ag ions. This study aimed at impregnating the hydrophobic polycaprolactone (PCL) polymer, which is a FDA-approved polymeric medical device material, with hydrophilic silver nanoparticles. Furthermore, a novel approach was employed to uniformly, incorporate silver nanoparticles into the PCL matrix in situ and to improve the release of Ag ions from the matrix so as to enhance antimicrobial efficacy.


Asunto(s)
Antiinfecciosos/administración & dosificación , Equipos y Suministros , Nanopartículas del Metal , Poliésteres/química , Plata/química , Animales , Antiinfecciosos/farmacología , Biopelículas , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica de Rastreo , Espectroscopía de Fotoelectrones , Pseudomonas aeruginosa/efectos de los fármacos , Espectroscopía Infrarroja por Transformada de Fourier , Staphylococcus aureus/efectos de los fármacos
16.
PLoS One ; 8(11): e79038, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244411

RESUMEN

The Gram-negative bacterial pathogen Klebsiella pneumoniae forms biofilms to facilitate colonization of biotic and abiotic surfaces. The formation of biofilms by K. pneumoniae requires the expression of type 3 fimbriae: elongate proteinaceous filaments extruded by a chaperone-usher system in the bacterial outer membrane. The expression of the mrkABCDF cluster that encodes this fimbrial system is strongly positively regulated by MrkH, a transcriptional activator that responds to the second messenger, c-di-GMP. In this study, we analyzed the mechanism by which the MrkH protein activates transcriptional initiation from the mrkA promoter. A mutational analysis supported by electrophoretic mobility shift assays demonstrated that a 12-bp palindromic sequence (the MrkH box) centered at -78.5 is the binding site of MrkH. Deletion of half a turn, but not a full turn, of DNA located between the MrkH box and the mrkA promoter destroyed the ability of MrkH to activate mrkA transcription. In addition, a 10-bp AT-rich sequence (the UP element) centered at -63.5 contributed significantly to MrkH-dependent mrkA transcription. In vivo analysis of rpoA mutants showed that the R265 and E273 determinants in the C-terminal domain of RNA polymerase α subunit are needed for MrkH-mediated activation of mrkA transcription. Furthermore, results from mutagenesis of the mrkH gene suggest that the N-terminal region of the protein is involved in transcriptional activation. Taken together, our results suggest that MrkH activates mrkA expression by interacting directly with RNA polymerase, to overcome the inefficient transcriptional initiation caused by the presence of defective core promoter elements.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fimbrias Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Klebsiella pneumoniae/metabolismo , Operón/fisiología , Regiones Promotoras Genéticas/fisiología , Proteínas Bacterianas/genética , Fimbrias Bacterianas/genética , Klebsiella pneumoniae/genética , Estructura Terciaria de Proteína , Transcripción Genética/fisiología
17.
J Biol Chem ; 288(43): 31115-26, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24019519

RESUMEN

The misuse of antibiotics during past decades has led to pervasive antibiotic resistance in bacteria. Hence, there is an urgent need for the development of new and alternative approaches to combat bacterial infections. In most bacterial pathogens the expression of virulence is tightly regulated at the transcriptional level. Therefore, targeting pathogens with drugs that interfere with virulence gene expression offers an effective alternative to conventional antimicrobial chemotherapy. Many Gram-negative intestinal pathogens produce AraC-like proteins that control the expression of genes required for infection. In this study we investigated the prototypical AraC-like virulence regulator, RegA, from the mouse attaching and effacing pathogen, Citrobacter rodentium, as a potential drug target. By screening a small molecule chemical library and chemical optimization, we identified two compounds that specifically inhibited the ability of RegA to activate its target promoters and thus reduced expression of a number of proteins required for virulence. Biophysical, biochemical, genetic, and computational analyses indicated that the more potent of these two compounds, which we named regacin, disrupts the DNA binding capacity of RegA by interacting with amino acid residues within a conserved region of the DNA binding domain. Oral administration of regacin to mice, commencing 15 min before or 12 h after oral inoculation with C. rodentium, caused highly significant attenuation of intestinal colonization by the mouse pathogen comparable to that of an isogenic regA-deletion mutant. These findings demonstrate that chemical inhibition of the DNA binding domains of transcriptional regulators is a viable strategy for the development of antimicrobial agents that target bacterial pathogens.


Asunto(s)
Antibacterianos/farmacología , Factor de Transcripción de AraC/antagonistas & inhibidores , Citrobacter rodentium/metabolismo , Citrobacter rodentium/patogenicidad , Infecciones por Enterobacteriaceae/metabolismo , Factores de Virulencia/antagonistas & inhibidores , Animales , Antibacterianos/química , Factor de Transcripción de AraC/genética , Factor de Transcripción de AraC/metabolismo , Citrobacter rodentium/genética , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/patología , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Intestinos/microbiología , Intestinos/patología , Ratones , Estructura Terciaria de Proteína , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
18.
Infect Immun ; 81(11): 4232-43, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24002063

RESUMEN

Atypical enteropathogenic Escherichia coli (aEPEC) causes endemic diarrhea, diarrheal outbreaks, and persistent diarrhea in humans, but the mechanism by which aEPEC causes disease is incompletely understood. Virulence regulators and their associated regulons, which often include adhesins, play key roles in the expression of virulence factors in enteric pathogenic bacteria. In this study we identified a transcriptional regulator, RalR, in the rabbit-specific aEPEC strain, E22 (O103:H2) and examined its involvement in the regulation of virulence. Microarray analysis and quantitative real-time reverse transcription-PCR demonstrated that RalR enhances the expression of a number of genes encoding virulence-associated factors, including the Ral fimbria, the Aap dispersin, and its associated transport system, and downregulates several housekeeping genes, including fliC. These observations were confirmed by proteomic analysis of secreted and heat-extracted surface-associated proteins and by adherence and motility assays. To investigate the mechanism of RalR-mediated activation, we focused on its most highly upregulated target operons, ralCDEFGHI and aap. By using primer extension, electrophoretic mobility shift assay, and mutational analysis, we identified the promoter and operator sequences for these two operons. By employing promoter-lacZ reporter systems, we demonstrated that RalR activates the expression of its target genes by binding to one or more 8-bp palindromic sequences (with the consensus of TGTGCACA) located immediately upstream of the promoter core regions. Importantly, we also demonstrated that RalR is essential for virulence since infection of rabbits with E22 carrying a knockout mutation in the ralR gene completely abolished its ability to cause disease.


Asunto(s)
Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Factores de Transcripción/metabolismo , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/genética , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Análisis por Micromatrices , Regiones Promotoras Genéticas , Proteoma/análisis , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/genética , Virulencia
19.
Infect Immun ; 81(4): 1078-89, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23340312

RESUMEN

AraC-like regulators play a key role in the expression of virulence factors in enteric pathogens, such as enteropathogenic Escherichia coli (EPEC), enterotoxigenic E. coli, enteroaggregative E. coli, and Citrobacter rodentium. Bioinformatic analysis of the genome of rabbit-specific EPEC (REPEC) strain E22 (O103:H2) revealed the presence of a gene encoding an AraC-like regulatory protein, RegR, which shares 71% identity to the global virulence regulator, RegA, of C. rodentium. Microarray analysis demonstrated that RegR exerts 25- to 400-fold activation on transcription of several genes encoding putative virulence-associated factors, including a fimbrial operon (SEF14), a serine protease, and an autotransporter adhesin. These observations were confirmed by proteomic analysis of secreted and heat-extracted surface-associated proteins. The mechanism of RegR-mediated activation was investigated by using its most highly upregulated gene target, sefA. Transcriptional analyses and electrophoretic mobility shift assays showed that RegR activates the expression of sefA by binding to a region upstream of the sefA promoter, thereby relieving gene silencing by the global regulatory protein H-NS. Moreover, RegR was found to contribute significantly to virulence in a rabbit infection experiment. Taken together, our findings indicate that RegR controls the expression of a series of accessory adhesins that significantly enhance the virulence of REPEC strain E22.


Asunto(s)
Proteínas Bacterianas/metabolismo , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Regulación Bacteriana de la Expresión Génica , Regulón , Factores de Transcripción/metabolismo , Factores de Virulencia/biosíntesis , Animales , Proteínas Bacterianas/genética , Biología Computacional , ADN Bacteriano/metabolismo , Modelos Animales de Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Proteínas de Escherichia coli/análisis , Proteínas Fimbrias/biosíntesis , Proteínas Fimbrias/genética , Perfilación de la Expresión Génica , Análisis por Micromatrices , Regiones Promotoras Genéticas , Unión Proteica , Proteoma/análisis , Conejos , Factores de Transcripción/genética , Transcripción Genética , Factores de Virulencia/genética
20.
Infect Immun ; 80(6): 2042-52, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22451516

RESUMEN

Enteropathogenic Escherichia coli (EPEC) is a major cause of diarrhea in infants in developing countries. We have identified a functional type II secretion system (T2SS) in EPEC that is homologous to the pathway responsible for the secretion of heat-labile enterotoxin by enterotoxigenic E. coli. The wild-type EPEC T2SS was able to secrete a heat-labile enterotoxin reporter, but an isogenic T2SS mutant could not. We showed that the major substrate of the T2SS in EPEC is SslE, an outer membrane lipoprotein (formerly known as YghJ), and that a functional T2SS is essential for biofilm formation by EPEC. T2SS and SslE mutants were arrested at the microcolony stage of biofilm formation, suggesting that the T2SS is involved in the development of mature biofilms and that SslE is a dominant effector of biofilm development. Moreover, the T2SS was required for virulence, as infection of rabbits with a rabbit-specific EPEC strain carrying a mutation in either the T2SS or SslE resulted in significantly reduced intestinal colonization and milder disease.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Escherichia coli Enteropatógena/fisiología , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/metabolismo , Factores de Virulencia/metabolismo , Animales , Membrana Celular , Escherichia coli Enteropatógena/citología , Escherichia coli Enteropatógena/genética , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Mutación , Conejos , Especificidad por Sustrato , Virulencia , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...