Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Appl Microbiol ; 135(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38486355

RESUMEN

AIMS: The main objective of this study was to modify a recently reported multi-purpose artificial urine (MP-AU) for culture and gene expression studies of uropathogenic Escherichia coli (UPEC) strains. METHODS AND RESULTS: We used liquid chromatography mass spectrometry (LC-MS) to identify and adjust the metabolic profile of MP-AU closer to that of pooled human urine (PHU). Modification in this way facilitated growth of UPEC strains with growth rates similar to those obtained in PHU. Transcriptomic analysis of UPEC strains cultured in enhanced artificial urine (enhanced AU) and PHU showed that the gene expression profiles are similar, with <7% of genes differentially expressed between the two conditions. CONCLUSIONS: Enhancing an MP-AU with metabolites identified in PHU allows the enhanced AU to be used as a substitute for the culture and in vitro gene expression studies of UPEC strains.


Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Humanos , Escherichia coli Uropatógena/genética , Perfilación de la Expresión Génica , Expresión Génica , Proteínas de Escherichia coli/genética , Factores de Virulencia/genética
2.
PLoS Pathog ; 19(6): e1011015, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37384772

RESUMEN

Clostridioides difficile is responsible for substantial morbidity and mortality in antibiotically-treated, hospitalised, elderly patients, in which toxin production correlates with diarrhoeal disease. While the function of these toxins has been studied in detail, the contribution of other factors, including the paracrystalline surface layer (S-layer), to disease is less well understood. Here, we highlight the essentiality of the S-layer in vivo by reporting the recovery of S-layer variants, following infection with the S-layer-null strain, FM2.5. These variants carry either correction of the original point mutation, or sequence modifications which restored the reading frame, and translation of slpA. Selection of these variant clones was rapid in vivo, and independent of toxin production, with up to 90% of the recovered C. difficile population encoding modified slpA sequence within 24 h post infection. Two variants, subsequently named FM2.5varA and FM2.5varB, were selected for study in greater detail. Structural determination of SlpA from FM2.5varB indicated an alteration in the orientation of protein domains, resulting in a reorganisation of the lattice assembly, and changes in interacting interfaces, which might alter function. Interestingly, variant FM2.5varB displayed an attenuated, FM2.5-like phenotype in vivo compared to FM2.5varA, which caused disease severity more comparable to that of R20291. Comparative RNA sequencing (RNA-Seq) analysis of in vitro grown isolates revealed large changes in gene expression between R20291 and FM2.5. Downregulation of tcdA/tcdB and several genes associated with sporulation and cell wall integrity may account for the reported attenuated phenotype of FM2.5 in vivo. RNA-seq data correlated well with disease severity with the more virulent variant, FM2.5varA, showing s similar profile of gene expression to R20291 in vitro, while the attenuated FM2.5varB showed downregulation of many of the same virulence associated traits as FM2.5. Cumulatively, these data add to a growing body of evidence that the S-layer contributes to C. difficile pathogenesis and disease severity.


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Clostridioides , Clostridioides difficile/genética , Pared Celular , Células Clonales
3.
Nat Microbiol ; 8(6): 1160-1175, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37231089

RESUMEN

Clostridium perfringens is an anaerobic toxin-producing bacterium associated with intestinal diseases, particularly in neonatal humans and animals. Infant gut microbiome studies have recently indicated a link between C. perfringens and the preterm infant disease necrotizing enterocolitis (NEC), with specific NEC cases associated with overabundant C. perfringens termed C. perfringens-associated NEC (CPA-NEC). In the present study, we carried out whole-genome sequencing of 272 C. perfringens isolates from 70 infants across 5 hospitals in the United Kingdom. In this retrospective analysis, we performed in-depth genomic analyses (virulence profiling, strain tracking and plasmid analysis) and experimentally characterized pathogenic traits of 31 strains, including 4 from CPA-NEC patients. We found that the gene encoding toxin perfringolysin O, pfoA, was largely deficient in a human-derived hypovirulent lineage, as well as certain colonization factors, in contrast to typical pfoA-encoding virulent lineages. We determined that infant-associated pfoA+ strains caused significantly more cellular damage than pfoA- strains in vitro, and further confirmed this virulence trait in vivo using an oral-challenge C57BL/6 murine model. These findings suggest both the importance of pfoA+ C. perfringens as a gut pathogen in preterm infants and areas for further investigation, including potential intervention and therapeutic strategies.


Asunto(s)
Clostridium perfringens , Enfermedades del Recién Nacido , Lactante , Recién Nacido , Humanos , Animales , Ratones , Clostridium perfringens/genética , Recien Nacido Prematuro , Estudios Retrospectivos , Virulencia/genética , Genómica
4.
Proc Natl Acad Sci U S A ; 120(1): e2212175120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36574699

RESUMEN

The ability of bacterial pathogens to adapt to host niches is driven by the carriage and regulation of genes that benefit pathogenic lifestyles. Genes that encode virulence or fitness-enhancing factors must be regulated in response to changing host environments to allow rapid response to challenges presented by the host. Furthermore, this process can be controlled by preexisting transcription factors (TFs) that acquire new roles in tailoring regulatory networks, specifically in pathogens. However, the mechanisms underlying this process are poorly understood. The highly conserved Escherichia coli TF YhaJ exhibits distinct genome-binding dynamics and transcriptome control in pathotypes that occupy different host niches, such as uropathogenic E. coli (UPEC). Here, we report that this important regulator is required for UPEC systemic survival during murine bloodstream infection (BSI). This advantage is gained through the coordinated regulation of a small regulon comprised of both virulence and metabolic genes. YhaJ coordinates activation of both Type 1 and F1C fimbriae, as well as biosynthesis of the amino acid tryptophan, by both direct and indirect mechanisms. Deletion of yhaJ or the individual genes under its control leads to attenuated survival during BSI. Furthermore, all three systems are up-regulated in response to signals derived from serum or systemic host tissue, but not urine, suggesting a niche-specific regulatory trigger that enhances UPEC fitness via pleiotropic mechanisms. Collectively, our results identify YhaJ as a pathotype-specific regulatory aide, enhancing the expression of key genes that are collectively required for UPEC bloodstream pathogenesis.


Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Sepsis , Infecciones Urinarias , Escherichia coli Uropatógena , Animales , Ratones , Escherichia coli/genética , Escherichia coli/metabolismo , Infecciones Urinarias/microbiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Factores de Virulencia/genética , Escherichia coli Uropatógena/genética , Regulación Bacteriana de la Expresión Génica
5.
Nat Commun ; 13(1): 970, 2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35217634

RESUMEN

Many bacteria and archaea possess a two-dimensional protein array, or S-layer, that covers the cell surface and plays crucial roles in cell physiology. Here, we report the crystal structure of SlpA, the main S-layer protein of the bacterial pathogen Clostridioides difficile, and use electron microscopy to study S-layer organisation and assembly. The SlpA crystal lattice mimics S-layer assembly in the cell, through tiling of triangular prisms above the cell wall, interlocked by distinct ridges facing the environment. Strikingly, the array is very compact, with pores of only ~10 Å in diameter, compared to other S-layers (30-100 Å). The surface-exposed flexible ridges are partially dispensable for overall structure and assembly, although a mutant lacking this region becomes susceptible to lysozyme, an important molecule in host defence. Thus, our work gives insights into S-layer organisation and provides a basis for development of C. difficile-specific therapeutics.


Asunto(s)
Clostridioides difficile , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Clostridioides difficile/genética
6.
Front Microbiol ; 12: 670535, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721311

RESUMEN

Bacteriocins are narrow-spectrum protein antibiotics that could potentially be used to engineer the human gut microbiota. However, technologies for targeted delivery of proteins to the lower gastrointestinal (GI) tract in preclinical animal models are currently lacking. In this work, we have developed methods for the microencapsulation of Escherichia coli targeting bacteriocins, colicin E9 and Ia, in a pH responsive formulation to allow their targeted delivery and controlled release in an in vivo murine model of E. coli colonization. Membrane emulsification was used to produce a water-in-oil emulsion with the water-soluble polymer subsequently cross-linked to produce hydrogel microcapsules. The microcapsule fabrication process allowed control of the size of the drug delivery system and a near 100% yield of the encapsulated therapeutic cargo. pH-triggered release of the encapsulated colicins was achieved using a widely available pH-responsive anionic copolymer in combination with alginate biopolymers. In vivo experiments using a murine E. coli intestinal colonization model demonstrated that oral delivery of the encapsulated colicins resulted in a significant decrease in intestinal colonization and reduction in E. coli shedding in the feces of the animals. Employing controlled release drug delivery systems such as that described here is essential to enable delivery of new protein therapeutics or other biological interventions for testing within small animal models of infection. Such approaches may have considerable value for the future development of strategies to engineer the human gut microbiota, which is central to health and disease.

7.
PLoS One ; 16(5): e0245949, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33979349

RESUMEN

Clostridioides difficile is a leading cause of human antibiotic-associated diarrhoeal disease globally. Zoonotic reservoirs of infection are increasingly suspected to play a role in the emergence of this disease in the community and dogs are considered as one potential source. Here we use a canine case-control study at a referral veterinary hospital in Scotland to assess: i) the risk factors associated with carriage of C. difficile by dogs, ii) whether carriage of C. difficile is associated with clinical disease in dogs and iii) the similarity of strains isolated from dogs with local human clinical surveillance. The overall prevalence of C. difficile carriage in dogs was 18.7% (95% CI 14.8-23.2%, n = 61/327) of which 34% (n = 21/61) were toxigenic strains. We found risk factors related to prior antibiotic treatment were significantly associated with C. difficile carriage by dogs. However, the presence of toxigenic strains of C. difficile in a canine faecal sample was not associated with diarrhoeal disease in dogs. Active toxin was infrequently detected in canine faecal samples carrying toxigenic strains (2/11 samples). Both dogs in which active toxin was detected had no clinical evidence of gastrointestinal disease. Among the ten toxigenic ribotypes of C. difficile detected in dogs in this study, six of these (012, 014, 020, 026, 078, 106) were ribotypes commonly associated with human clinical disease in Scotland, while nontoxigenic isolates largely belonged to 010 and 039 ribotypes. Whilst C. difficile does not appear commonly associated with diarrhoeal disease in dogs, antibiotic treatment increases carriage of this bacteria including toxigenic strains commonly found in human clinical disease.


Asunto(s)
Antibacterianos/uso terapéutico , Clostridioides difficile/patogenicidad , Animales , Infecciones por Clostridium/epidemiología , Enfermedades de los Perros/epidemiología , Perros , Femenino , Humanos , Masculino
8.
Sci Adv ; 6(11): eaax6328, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32195337

RESUMEN

Alterations to the gut microbiome are associated with various neurological diseases, yet evidence of causality and identity of microbiome-derived compounds that mediate gut-brain axis interaction remain elusive. Here, we identify two previously unknown bacterial metabolites 3-methyl-4-(trimethylammonio)butanoate and 4-(trimethylammonio)pentanoate, structural analogs of carnitine that are present in both gut and brain of specific pathogen-free mice but absent in germ-free mice. We demonstrate that these compounds are produced by anaerobic commensal bacteria from the family Lachnospiraceae (Clostridiales) family, colocalize with carnitine in brain white matter, and inhibit carnitine-mediated fatty acid oxidation in a murine cell culture model of central nervous system white matter. This is the first description of direct molecular inter-kingdom exchange between gut prokaryotes and mammalian brain cells, leading to inhibition of brain cell function.


Asunto(s)
Carnitina , Clostridiales/metabolismo , Microbioma Gastrointestinal , Mucosa Intestinal , Sustancia Blanca/metabolismo , Animales , Carnitina/análogos & derivados , Carnitina/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones
9.
Vaccines (Basel) ; 8(1)2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-32046000

RESUMEN

Abstract: The BclA3 glycoprotein is a major component of the exosporangial layer of Clostridium difficile spores and in this study we demonstrate that this glycoprotein is a major spore surface associated antigen. Here, we confirm the role of SgtA glycosyltransferase (SgtA GT) in BclA3 glycosylation and recapitulate this process by expressing and purifying SgtA GT fused to MalE, the maltose binding protein from Escherichia coli. In vitro assays using the recombinant enzyme and BclA3 synthetic peptides demonstrated that SgtA GT was responsible for the addition of ß-O-linked GlcNAc to threonine residues of each synthetic peptide. These peptide sequences were selected from the central, collagen repeat region of the BclA3 protein. Following optimization of SgtA GT activity, we generated sufficient glycopeptide (10 mg) to allow conjugation to KLH (keyhole limpet hemocyanin) protein. Glycosylated and unglycosylated versions of these conjugates were then used as antigens to immunize rabbits and mice. Immune responses to each of the conjugates were examined by Enzyme Linked Immunosorbent Assay ELISA. Additionally, the BclA3 conjugated peptide and glycopeptide were used as antigens in an ELISA assay with serum raised against formalin-killed spores. Only the glycopeptide was recognized by anti-spore polyclonal immune serum demonstrating that the glycan moiety is a predominant spore-associated surface antigen. To determine whether antibodies to these peptides could modify persistence of spores within the gut, animals immunized intranasally with either the KLH-glycopeptide or KLH-peptide conjugate in the presence of cholera toxin, were challenged with R20291 spores. Although specific antibodies were raised to both antigens, immunization did not provide any protection against acute or recurrent disease.

10.
Gut Microbes ; 11(3): 481-496, 2020 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-31793403

RESUMEN

Susceptibility of patients to antibiotic-associated C. difficile disease is intimately associated with specific changes to gut microbiome composition. In particular, loss of microbes that modify bile salt acids (BSA) play a central role; primary bile acids stimulate spore germination whilst secondary bile acids limit C. difficile vegetative growth. To determine the relative contribution of bile salt (BS) metabolism on C. difficile disease severity, we treated mice with three combinations of antibiotics prior to infection. Mice given clindamycin alone became colonized but displayed no tissue pathology while severe disease, exemplified by weight loss and inflammatory tissue damage occurred in animals given a combination of five antibiotics and clindamycin. Animals given only the five antibiotic cocktails showed only transient colonization and no disease. C. difficile colonization was associated with a reduction in bacterial diversity, an inability to amplify bile salt hydrolase (BSH) sequences from fecal DNA and a relative increase in primary bile acids (pBA) in cecal lavages from infected mice. Further, the link between BSA modification and the microbiome was confirmed by the isolation of strains of Lactobacillus murinus that modified primary bile acids in vitro, thus preventing C. difficile germination. Interestingly, BSH activity did not correlate with disease severity which appeared linked to alternations in mucin, which may indirectly lead to increased exposure of the epithelial surface to inflammatory signals. These data confirm the role of microbial metabolic activity in protection of the gut and highlights the need for greater understanding the function of bacterial communities in disease prevention.


Asunto(s)
Antibacterianos/efectos adversos , Ácidos y Sales Biliares/metabolismo , Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/microbiología , Microbioma Gastrointestinal , Amidohidrolasas/genética , Animales , Biodiversidad , Clostridioides difficile/patogenicidad , ADN Bacteriano , Modelos Animales de Enfermedad , Heces/microbiología , Inflamación/microbiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Índice de Severidad de la Enfermedad , Esporas Bacterianas/crecimiento & desarrollo
11.
Nat Commun ; 9(1): 5148, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30498235

RESUMEN

The original version of this Article contained an error in the spelling of the author David Ruano-Gallego, which was incorrectly given as David R. Gallego. This has now been corrected in both the PDF and HTML versions of the Article.

12.
Nat Commun ; 9(1): 4187, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305622

RESUMEN

Niche-adaptation of a bacterial pathogen hinges on the ability to recognize the complexity of signals from the environment and integrate that information with the regulation of genes critical for infection. Here we report the transcriptome of the attaching and effacing pathogen Citrobacter rodentium during infection of its natural murine host. Pathogen gene expression in vivo was heavily biased towards the virulence factor repertoire and was found to be co-ordinated uniquely in response to the host. Concordantly, we identified the host-specific induction of a metabolic pathway that overlapped with the regulation of virulence. The essential type 3 secretion system and an associated suite of distinct effectors were found to be modulated co-ordinately through a unique mechanism involving metabolism of microbiota-derived 1,2-propanediol, which dictated the ability to colonize the host effectively. This study provides novel insights into how host-specific metabolic adaptation acts as a cue to fine-tune virulence.


Asunto(s)
Sistemas de Secreción Bacterianos , Citrobacter rodentium/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Interacciones Huésped-Patógeno , Animales , Adhesión Bacteriana , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidad , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/patología , Regulación Bacteriana de la Expresión Génica , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Metabolómica , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Propilenglicol/metabolismo , Análisis de Secuencia de ARN , Transcriptoma/genética , Virulencia/genética , Factores de Virulencia/metabolismo
13.
Sci Transl Med ; 9(406)2017 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878013

RESUMEN

There is a medical need for antibacterial agents that do not damage the resident gut microbiota or promote the spread of antibiotic resistance. We recently described a prototypic precision bactericidal agent, Av-CD291.2, which selectively kills specific Clostridium difficile strains and prevents them from colonizing mice. We have since selected two Av-CD291.2-resistant mutants that have a surface (S)-layer-null phenotype due to distinct point mutations in the slpA gene. Using newly identified bacteriophage receptor binding proteins for targeting, we constructed a panel of Avidocin-CDs that kills diverse C. difficile isolates in an S-layer sequence-dependent manner. In addition to bacteriophage receptor recognition, characterization of the mutants also uncovered important roles for S-layer protein A (SlpA) in sporulation, resistance to innate immunity effectors, and toxin production. Surprisingly, S-layer-null mutants were found to persist in the hamster gut despite a complete attenuation of virulence. These findings suggest antimicrobials targeting virulence factors dispensable for fitness in the host force pathogens to trade virulence for viability and would have clear clinical advantages should resistance emerge. Given their exquisite specificity for the pathogen, Avidocin-CDs have substantial therapeutic potential for the treatment and prevention of C. difficile infections.


Asunto(s)
Antiinfecciosos/farmacología , Clostridioides difficile/patogenicidad , Glicoproteínas de Membrana/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Bacteriocinas/farmacología , Clostridioides difficile/efectos de los fármacos , Recuento de Colonia Microbiana , Cricetinae , Farmacorresistencia Bacteriana/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Inmunidad Innata/efectos de los fármacos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Viabilidad Microbiana/efectos de los fármacos , Mutación/genética , Fenotipo , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/fisiología , Toxinas Biológicas/metabolismo , Virulencia/efectos de los fármacos
14.
Front Microbiol ; 7: 1930, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27965652

RESUMEN

Infections caused by Shiga toxin (Stx)-producing E. coli strains constitute a health problem, as they are problematic to treat. Stx production is a key virulence factor associated with the pathogenicity of enterohaemorrhagic E. coli (EHEC) and can result in the development of haemolytic uremic syndrome in infected patients. The genes encoding Stx are located on temperate lysogenic phages integrated into the bacterial chromosome and expression of the toxin is generally coupled to phage induction through the SOS response. We aimed to find new compounds capable of blocking expression of Stx type 2 (Stx2) as this subtype of Stx is more strongly associated with human disease. High-throughput screening of a small-molecule library identified a lead compound that reduced Stx2 expression in a dose-dependent manner. We show that the optimized compound interferes with the SOS response by directly affecting the activity and oligomerization of RecA, thus limiting phage activation and Stx2 expression. Our work suggests that RecA is highly susceptible to inhibition and that targeting this protein is a viable approach to limiting production of Stx2 by EHEC. This type of approach has the potential to limit production and transfer of other phage induced and transduced determinants.

15.
Sci Rep ; 6: 30201, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27444885

RESUMEN

Protein antibiotics, known as bacteriocins, are widely produced by bacteria for intraspecies competition. The potency and targeted action of bacteriocins suggests that they could be developed into clinically useful antibiotics against highly drug resistant Gram-negative pathogens for which there are few therapeutic options. Here we show that Pseudomonas aeruginosa specific bacteriocins, known as pyocins, show strong efficacy in a murine model of P. aeruginosa lung infection, with the concentration of pyocin S5 required to afford protection from a lethal infection at least 100-fold lower than the most commonly used inhaled antibiotic tobramycin. Additionally, pyocins are stable in the lung, poorly immunogenic at high concentrations and efficacy is maintained in the presence of pyocin specific antibodies after repeated pyocin administration. Bacteriocin encoding genes are frequently found in microbial genomes and could therefore offer a ready supply of highly targeted and potent antibiotics active against problematic Gram-negative pathogens.


Asunto(s)
Antibacterianos/farmacología , Enfermedades Pulmonares/tratamiento farmacológico , Pulmón/microbiología , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Bacteriocinas/farmacología , Modelos Animales de Enfermedad , Femenino , Enfermedades Pulmonares/microbiología , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/microbiología , Piocinas/farmacología , Especificidad de la Especie , Tobramicina/farmacología
16.
Sci Rep ; 6: 23463, 2016 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-26996606

RESUMEN

The uses of fluorescent reporters derived from green fluorescent protein have proved invaluable for the visualisation of biological processes in bacteria grown under aerobic conditions. However, their requirement for oxygen has limited their application in obligate anaerobes such as Clostridium difficile. Fluorescent proteins derived from Light, Oxygen or Voltage sensing (LOV) domains have been shown to bridge this limitation, but their utility as translational fusions to monitor protein expression and localisation in a strict anaerobic bacterium has not been reported. Here we demonstrate the utility of phiLOV in three species of Clostridium and its application as a marker of real-time protein translation and dynamics through genetic fusion with the cell division protein, FtsZ. Time lapse microscopy of dividing cells suggests that Z ring assembly arises through the extension of the FtsZ arc starting from one point on the circumference. Furthermore, through incorporation of phiLOV into the flagella subunit, FliC, we show the potential of bacterial LOV-based fusion proteins to be successfully exported to the extracellular environment.


Asunto(s)
Técnicas Biosensibles/métodos , Clostridioides difficile/genética , Imagen Molecular/métodos , Imagen Óptica/métodos , Biosíntesis de Proteínas , Proteínas Bacterianas/biosíntesis , Clostridioides difficile/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Transporte de Proteínas
17.
Antimicrob Agents Chemother ; 60(2): 968-81, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26643348

RESUMEN

The microbiome dysbiosis caused by antibiotic treatment has been associated with both susceptibility to and relapse of Clostridium difficile infection (CDI). Bacteriophage (phage) therapy offers target specificity and dose amplification in situ, but few studies have focused on its use in CDI treatment. This mainly reflects the lack of strictly virulent phages that target this pathogen. While it is widely accepted that temperate phages are unsuitable for therapeutic purposes due to their transduction potential, analysis of seven C. difficile phages confirmed that this impact could be curtailed by the application of multiple phage types. Here, host range analysis of six myoviruses and one siphovirus was conducted on 80 strains representing 21 major epidemic and clinically severe ribotypes. The phages had complementary coverage, lysing 18 and 62 of the ribotypes and strains tested, respectively. Single-phage treatments of ribotype 076, 014/020, and 027 strains showed an initial reduction in the bacterial load followed by the emergence of phage-resistant colonies. However, these colonies remained susceptible to infection with an unrelated phage. In contrast, specific phage combinations caused the complete lysis of C. difficile in vitro and prevented the appearance of resistant/lysogenic clones. Using a hamster model, the oral delivery of optimized phage combinations resulted in reduced C. difficile colonization at 36 h postinfection. Interestingly, free phages were recovered from the bowel at this time. In a challenge model of the disease, phage treatment delayed the onset of symptoms by 33 h compared to the time of onset of symptoms in untreated animals. These data demonstrate the therapeutic potential of phage combinations to treat CDI.


Asunto(s)
Bacteriófagos/fisiología , Clostridioides difficile/patogenicidad , Clostridioides difficile/virología , Animales , Toxinas Bacterianas/metabolismo , Bacteriófagos/clasificación , Bacteriófagos/genética , Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/virología , Modelos Animales de Enfermedad , Femenino , Especificidad del Huésped , Mesocricetus , Filogenia , Ribotipificación , Esporas Bacterianas/virología
18.
Curr Opin Chem Biol ; 27: 39-45, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26087123

RESUMEN

Chromophore-binding domains from plant and bacterial photoreceptor proteins have recently gathered increasing attention as new sources of genetically encoded fluorescent proteins (FPs). In particular, FPs based on the flavin-binding LOV (light, oxygen, or voltage sensing) domain offer advantages over green fluorescent protein (GFP) owing to their smaller size, pH and thermal stability, utility under anaerobic conditions and their ability to generate reactive oxygen species. This review focuses on the potential applications of this emerging class of fluorescent reporters, discusses the advantages and limitations of LOV-based FPs, whilst offering insights regarding the further development of this technology for bioimaging and photodynamic therapy.


Asunto(s)
Proteínas Bacterianas/química , Técnicas Biosensibles/métodos , Dinitrocresoles/química , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/química , Imagen Molecular/métodos , Proteínas Bacterianas/genética , Proteínas Fluorescentes Verdes/genética , Modelos Moleculares , Oxígeno/química , Unión Proteica , Estabilidad Proteica , Rayos Ultravioleta
19.
Eur J Immunol ; 44(12): 3658-68, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25236797

RESUMEN

Signal regulatory protein alpha (SIRPα/CD172a) is a conserved transmembrane protein thought to play an inhibitory role in immune function by binding the ubiquitous ligand CD47. SIRPα expression has been used to identify dendritic cell subsets across species and here we examined its expression and function on intestinal DCs in mice. Normal mucosa contains four subsets of DCs based on their expression of CD103 and CD11b and three of these express SIRPα. However, loss of SIRPα signaling in mice leads to a selective reduction in the CD103(+) CD11b(+) subset of DCs in the small intestine, colon, and among migratory DCs in the mesenteric lymph node. In parallel, these mice have reduced numbers of TH 17 cells in steady-state intestinal mucosa, and a defective TH 17 response to Citrobacter infection. Identical results were obtained in CD47KO mice. DC precursors from SIRPα mutant mice had an enhanced ability to generate CD103(+) CD11b(+) DCs in vivo, but CD103(+) CD11b(+) DCs from mutant mice were more prone to die by apoptosis. These data show a previously unappreciated and crucial role for SIRPα in the homeostasis of CD103(+) CD11b(+) DCs in the intestine, as well as providing further evidence that this subset of DCs is critical for the development of mucosal TH 17 responses.


Asunto(s)
Antígenos CD/inmunología , Antígeno CD11b/inmunología , Células Dendríticas/inmunología , Homeostasis/fisiología , Inmunidad Mucosa/fisiología , Cadenas alfa de Integrinas/inmunología , Mucosa Intestinal/inmunología , Receptores Inmunológicos/inmunología , Animales , Antígenos CD/genética , Apoptosis/genética , Apoptosis/inmunología , Antígeno CD11b/genética , Citrobacter/genética , Citrobacter/inmunología , Células Dendríticas/citología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Cadenas alfa de Integrinas/genética , Mucosa Intestinal/citología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Mesenterio/citología , Mesenterio/inmunología , Ratones , Ratones Mutantes , Receptores Inmunológicos/genética , Células Th17/citología , Células Th17/inmunología
20.
Mol Microbiol ; 94(2): 272-89, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25135277

RESUMEN

Clostridium difficile is a prominent nosocomial pathogen, proliferating and causing enteric disease in individuals with a compromised gut microflora. We characterized the post-translational modification of flagellin in C. difficile 630. The structure of the modification was solved by nuclear magnetic resonance and shown to contain an N-acetylglucosamine substituted with a phosphorylated N-methyl-l-threonine. A reverse genetics approach investigated the function of the putative four-gene modification locus. All mutants were found to have truncated glycan structures by LC-MS/MS, taking into account bioinformatic analysis, we propose that the open reading frame CD0241 encodes a kinase involved in the transfer of the phosphate to the threonine, the CD0242 protein catalyses the addition of the phosphothreonine to the N-acetylglucosamine moiety and CD0243 transfers the methyl group to the threonine. Some mutations affected motility and caused cells to aggregate to each other and abiotic surfaces. Altering the structure of the flagellin modification impacted on colonization and disease recurrence in a murine model of infection, showing that alterations in the surface architecture of C. difficile vegetative cells can play a significant role in disease. We show that motility is not a requirement for colonization, but that colonization was compromised when the glycan structure was incomplete.


Asunto(s)
Clostridioides difficile/química , Clostridioides difficile/fisiología , Flagelina/química , Flagelina/metabolismo , Locomoción , Procesamiento Proteico-Postraduccional , Propiedades de Superficie , Animales , Cromatografía Liquida , Clostridioides difficile/metabolismo , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/patología , Modelos Animales de Enfermedad , Espectroscopía de Resonancia Magnética , Ratones , Recurrencia , Espectrometría de Masas en Tándem , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...