Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 67(1): 450-466, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38112278

RESUMEN

With the aim of discovering small molecule inhibitors of the sporulation process in Clostridioides difficile, we prepared a series of C-7 α-(4-substituted-1H-1,2,3-triazol-1-yl)acetamide analogues of cefotetan, a known inhibitor of the C. difficile sporulation-specific protein target CdSpoVD. These analogues were evaluated using both in vitro binding assays with CdSpoVD and antisporulation assays against C. difficile. Further design concepts were aided utilizing the predicted docking scores (DS) using both AlphaFold (AF) models, and a crystal structure of the CdSpoVD protein (PDB 7RCZ). Despite being 1 order of magnitude more potent as a sporulation inhibitor than cefotetan, in vivo studies on compound 6a in a murine-model of C. difficile infection demonstrated comparable spore shedding capabilities as cefotetan. Importantly, compound 6a had no concerning broad spectrum antibacterial activities, toxicity, or hemolytic activity and thus has potential for further drug development.


Asunto(s)
Cefamicinas , Clostridioides difficile , Infecciones por Clostridium , Animales , Ratones , Cefamicinas/metabolismo , Clostridioides , Cefotetán/metabolismo , Esporas Bacterianas , Antibacterianos/química , Proteínas Bacterianas/metabolismo
2.
Int J Mol Sci ; 24(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36675151

RESUMEN

Apicomplexan infections, such as giardiasis and cryptosporidiosis, negatively impact a considerable proportion of human and commercial livestock populations. Despite this, the molecular mechanisms of disease, particularly the effect on the body beyond the gastrointestinal tract, are still poorly understood. To highlight host-parasite-microbiome biochemical interactions, we utilised integrated metabolomics-16S rRNA genomics and metabolomics-proteomics approaches in a C57BL/6J mouse model of giardiasis and compared these to Cryptosporidium and uropathogenic Escherichia coli (UPEC) infections. Comprehensive samples (faeces, blood, liver, and luminal contents from duodenum, jejunum, ileum, caecum and colon) were collected 10 days post infection and subjected to proteome and metabolome analysis by liquid and gas chromatography-mass spectrometry, respectively. Microbial populations in faeces and luminal washes were examined using 16S rRNA metagenomics. Proteome-metabolome analyses indicated that 12 and 16 key pathways were significantly altered in the gut and liver, respectively, during giardiasis with respect to other infections. Energy pathways including glycolysis and supporting pathways of glyoxylate and dicarboxylate metabolism, and the redox pathway of glutathione metabolism, were upregulated in small intestinal luminal contents and the liver during giardiasis. Metabolomics-16S rRNA genetics integration indicated that populations of three bacterial families-Autopobiaceae (Up), Desulfovibrionaceae (Up), and Akkermanasiaceae (Down)-were most significantly affected across the gut during giardiasis, causing upregulated glycolysis and short-chained fatty acid (SCFA) metabolism. In particular, the perturbed Akkermanasiaceae population seemed to cause oxidative stress responses along the gut-liver axis. Overall, the systems biology approach applied in this study highlighted that the effects of host-parasite-microbiome biochemical interactions extended beyond the gut ecosystem to the gut-liver axis. These findings form the first steps in a comprehensive comparison to ascertain the major molecular and biochemical contributors of host-parasite interactions and contribute towards the development of biomarker discovery and precision health solutions for apicomplexan infections.


Asunto(s)
Criptosporidiosis , Cryptosporidium , Microbioma Gastrointestinal , Giardiasis , Microbiota , Ratones , Animales , Humanos , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Regulación hacia Arriba , Proteoma/metabolismo , Criptosporidiosis/metabolismo , Ratones Endogámicos C57BL , Cryptosporidium/metabolismo , Metabolómica , Metaboloma , Hígado/metabolismo , Oxidación-Reducción
3.
Gut Microbes ; 14(1): 2117504, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36045589

RESUMEN

Clostridioides difficile is the most common cause of infectious antibiotic-associated diarrhea, with disease mediated by two major toxins TcdA and TcdB. In severe cases, systemic disease complications may arise, resulting in fatal disease. Systemic disease in animal models has been described, with thymic damage an observable consequence of severe disease in mice. Using a mouse model of C. difficile infection, we examined this disease phenotype, focussing on the thymus and serum markers of systemic disease. The efficacy of bezlotoxumab, a monoclonal TcdB therapeutic, to prevent toxin mediated systemic disease complications was also examined. C. difficile infection causes toxin-dependent thymic damage and CD4+CD8+ thymocyte depletion in mice. These systemic complications coincide with changes in biochemical markers of liver and kidney function, including increased serum urea and creatinine, and hypoglycemia. Administration of bezlotoxumab during C. difficile infection prevents systemic disease and thymic atrophy, without blocking gut damage, suggesting the leakage of gut contents into circulation may influence systemic disease. As the thymus has such a crucial role in T cell production and immune system development, these findings may have important implications in relapse of C. difficile disease and impaired immunity during C. difficile infection. The prevention of thymic atrophy and reduced systemic response following bezlotoxumab treatment, without altering colonic damage, highlights the importance of systemic disease in C. difficile infection, and provides new insights into the mechanism of action for this therapeutic.Abbreviations: Acute kidney injury (AKI); Alanine Transaminase (ALT); Aspartate Aminotransferase (AST); C. difficile infection (CDI); chronic kidney disease (CKD); combined repetitive oligo-peptides (CROPS); cardiovascular disease (CVD); Double positive (DP); hematoxylin and eosin (H&E); immunohistochemical (IHC); multiple organ dysfunction syndrome (MODS); phosphate buffered saline (PBS); standard error of the mean (SEM); surface layer proteins (SLP); Single positive (SP); wild-type (WT).


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Infecciones por Clostridium , Microbioma Gastrointestinal , Animales , Anticuerpos Monoclonales , Atrofia , Proteínas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Anticuerpos ampliamente neutralizantes , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/prevención & control , Enterotoxinas/metabolismo
4.
Sci Transl Med ; 14(662): eabj2381, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36103517

RESUMEN

Drug-resistant Gram-positive bacterial infections are still a substantial burden on the public health system, with two bacteria (Staphylococcus aureus and Streptococcus pneumoniae) accounting for over 1.5 million drug-resistant infections in the United States alone in 2017. In 2019, 250,000 deaths were attributed to these pathogens globally. We have developed a preclinical glycopeptide antibiotic, MCC5145, that has excellent potency (MIC90 ≤ 0.06 µg/ml) against hundreds of isolates of methicillin-resistant S. aureus (MRSA) and other Gram-positive bacteria, with a greater than 1000-fold margin over mammalian cell cytotoxicity values. The antibiotic has therapeutic in vivo efficacy when dosed subcutaneously in multiple murine models of established bacterial infections, including thigh infection with MRSA and blood septicemia with S. pneumoniae, as well as when dosed orally in an antibiotic-induced Clostridioides difficile infection model. MCC5145 exhibited reduced nephrotoxicity at microbiologically active doses in mice compared to vancomycin. MCC5145 also showed improved activity against biofilms compared to vancomycin, both in vitro and in vivo, and a low propensity to select for drug resistance. Characterization of drug action using a transposon library bioinformatic platform showed a mechanistic distinction from other glycopeptide antibiotics.


Asunto(s)
Antiinfecciosos , Infecciones por Bacterias Grampositivas , Staphylococcus aureus Resistente a Meticilina , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , Biopelículas , Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Lipoglucopéptidos/uso terapéutico , Mamíferos , Ratones , Pruebas de Sensibilidad Microbiana , Streptococcus pneumoniae , Vancomicina/farmacología , Vancomicina/uso terapéutico
5.
Antibiotics (Basel) ; 10(8)2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34438963

RESUMEN

Clostridioides (also known as Clostridium) difficile is a Gram-positive anaerobic, spore producing bacterial pathogen that causes severe gastrointestinal infection in humans. The current chemotherapeutic options are inadequate, expensive, and limited, and thus inexpensive drug treatments for C. difficile infection (CDI) with improved efficacy and specificity are urgently needed. To improve the solubility of our cationic amphiphilic 1,1'-binaphthylpeptidomimetics developed earlier that showed promise in an in vivo murine CDI model we have synthesized related compounds with an N-arytriazole or N-naphthyltriazole moiety instead of the 1,1'-biphenyl or 1,1'-binaphthyl moiety. This modification was made to increase the polarity and thus water solubility of the overall peptidomimetics, while maintaining the aromatic character. The dicationic N-naphthyltriazole derivative 40 was identified as a C. difficile-selective antibacterial with MIC values of 8 µg/mL against C. difficile strains ATCC 700057 and 132 (both ribotype 027). This compound displayed increased water solubility and reduced hemolytic activity (32 µg/mL) in an in vitro hemolysis assay and reduced cytotoxicity (CC50 32 µg/mL against HEK293 cells) relative to lead compound 2. Compound 40 exhibited mild efficacy (with 80% survival observed after 24 h compared to the DMSO control of 40%) in an in vivo murine model of C. difficile infection by reducing the severity and slowing the onset of disease.

6.
Metabolites ; 11(6)2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-34208228

RESUMEN

Cryptosporidiosis is a major human health concern globally. Despite well-established methods, misdiagnosis remains common. Our understanding of the cryptosporidiosis biochemical mechanism remains limited, compounding the difficulty of clinical diagnosis. Here, we used a systems biology approach to investigate the underlying biochemical interactions in C57BL/6J mice infected with Cryptosporidium parvum. Faecal samples were collected daily following infection. Blood, liver tissues and luminal contents were collected 10 days post infection. High-resolution liquid chromatography and low-resolution gas chromatography coupled with mass spectrometry were used to analyse the proteomes and metabolomes of these samples. Faeces and luminal contents were additionally subjected to 16S rRNA gene sequencing. Univariate and multivariate statistical analysis of the acquired data illustrated altered host and microbial energy pathways during infection. Glycolysis/citrate cycle metabolites were depleted, while short-chain fatty acids and D-amino acids accumulated. An increased abundance of bacteria associated with a stressed gut environment was seen. Host proteins involved in energy pathways and Lactobacillus glyceraldehyde-3-phosphate dehydrogenase were upregulated during cryptosporidiosis. Liver oxalate also increased during infection. Microbiome-parasite relationships were observed to be more influential than the host-parasite association in mediating major biochemical changes in the mouse gut during cryptosporidiosis. Defining this parasite-microbiome interaction is the first step towards building a comprehensive cryptosporidiosis model towards biomarker discovery, and rapid and accurate diagnostics.

7.
Gastroenterology ; 159(4): 1431-1443.e6, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32574621

RESUMEN

BACKGROUND & AIMS: The protease plasmin is an important wound healing factor, but it is not clear how it affects gastrointestinal infection-mediated damage, such as that resulting from Clostridioides difficile. We investigated the role of plasmin in C difficile-associated disease. This bacterium produces a spore form that is required for infection, so we also investigated the effects of plasmin on spores. METHODS: C57BL/6J mice expressing the precursor to plasmin, the zymogen human plasminogen (hPLG), or infused with hPLG were infected with C difficile, and disease progression was monitored. Gut tissues were collected, and cytokine production and tissue damage were analyzed by using proteomic and cytokine arrays. Antibodies that inhibit either hPLG activation or plasmin activity were developed and structurally characterized, and their effects were tested in mice. Spores were isolated from infected patients or mice and visualized using super-resolution microscopy; the functional consequences of hPLG binding to spores were determined. RESULTS: hPLG localized to the toxin-damaged gut, resulting in immune dysregulation with an increased abundance of cytokines (such as interleukin [IL] 1A, IL1B, IL3, IL10, IL12B, MCP1, MP1A, MP1B, GCSF, GMCSF, KC, TIMP-1), tissue degradation, and reduced survival. Administration of antibodies that inhibit plasminogen activation reduced disease severity in mice. C difficile spores bound specifically to hPLG and active plasmin degraded their surface, facilitating rapid germination. CONCLUSIONS: We found that hPLG is recruited to the damaged gut, exacerbating C difficile disease in mice. hPLG binds to C difficile spores, and, upon activation to plasmin, remodels the spore surface, facilitating rapid spore germination. Inhibitors of plasminogen activation might be developed for treatment of C difficile or other infection-mediated gastrointestinal diseases.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Enterocolitis Seudomembranosa/etiología , Enterocolitis Seudomembranosa/patología , Plasminógeno/farmacología , Esporas Bacterianas/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Humanos , Intestino Delgado , Ratones , Ratones Endogámicos C57BL
8.
Gut Pathog ; 12: 20, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32318117

RESUMEN

With the rise of bacterial resistance to conventional antibiotics, re-purposing of Food and Drug Administration (FDA) approved drugs currently used to treat non-bacteria related diseases as new leads for antibacterial drug discovery has become an attractive alternative. Ethoxzolamide (EZA), an FDA-approved diuretic acting as a human carbonic anhydrase inhibitor, is known to kill the gastric pathogenic bacterium Helicobacter pylori in vitro via an, as yet, unknown mechanism. To date, EZA activity and resistance have been investigated for only one H. pylori strain, P12. We have now performed a susceptibility and resistance study with H. pylori strains SS1 and 26695. Mutants resistant to EZA were isolated, characterized and their genomes sequenced. Resistance-conferring mutations were confirmed by backcrossing the mutations into the parent strain. As with P12, resistance to EZA in strains SS1 and 26695 does not develop easily, since the rate of spontaneous resistance acquisition was less than 10-8. Acquisition of resistance was associated with mutations in 3 genes in strain SS1, and in 6 different genes in strain 26695, indicating that EZA targets multiple systems. All resistant isolates had mutations affecting cell wall synthesis and control of gene expression. EZA's potential for treating duodenal ulcers has already been demonstrated. Our findings suggest that EZA may be developed into a novel anti-H. pylori drug.

9.
Proc Natl Acad Sci U S A ; 117(14): 8064-8073, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32198200

RESUMEN

Gastrointestinal infections often induce epithelial damage that must be repaired for optimal gut function. While intestinal stem cells are critical for this regeneration process [R. C. van der Wath, B. S. Gardiner, A. W. Burgess, D. W. Smith, PLoS One 8, e73204 (2013); S. Kozar et al., Cell Stem Cell 13, 626-633 (2013)], how they are impacted by enteric infections remains poorly defined. Here, we investigate infection-mediated damage to the colonic stem cell compartment and how this affects epithelial repair and recovery from infection. Using the pathogen Clostridioides difficile, we show that infection disrupts murine intestinal cellular organization and integrity deep into the epithelium, to expose the otherwise protected stem cell compartment, in a TcdB-mediated process. Exposure and susceptibility of colonic stem cells to intoxication compromises their function during infection, which diminishes their ability to repair the injured epithelium, shown by altered stem cell signaling and a reduction in the growth of colonic organoids from stem cells isolated from infected mice. We also show, using both mouse and human colonic organoids, that TcdB from epidemic ribotype 027 strains does not require Frizzled 1/2/7 binding to elicit this dysfunctional stem cell state. This stem cell dysfunction induces a significant delay in recovery and repair of the intestinal epithelium of up to 2 wk post the infection peak. Our results uncover a mechanism by which an enteric pathogen subverts repair processes by targeting stem cells during infection and preventing epithelial regeneration, which prolongs epithelial barrier impairment and creates an environment in which disease recurrence is likely.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/patología , Colon/patología , Mucosa Intestinal/patología , Células Madre/patología , Animales , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Células Cultivadas , Clostridioides difficile/metabolismo , Infecciones por Clostridium/microbiología , Colon/citología , Colon/microbiología , Modelos Animales de Enfermedad , Femenino , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Ratones , Organoides , Cultivo Primario de Células , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Madre/microbiología
10.
J Med Microbiol ; 69(2): 290-297, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32004137

RESUMEN

Introduction. Staphylococcus aureus is a recognised cause of foodborne intoxication and antibiotic-associated diarrhoea (AAD), which are both mediated by staphylococcal enterotoxins. However, unlike foodborne intoxication, AAD appears to require infection of the host. While S. aureus intoxication is widely studied, little is known about S. aureus pathogenesis in the context of gastrointestinal infection.Aim. To develop a mouse model of S. aureus gastrointestinal infection.Methodology. An established AAD mouse model was adapted for S. aureus infection, and damage observed via histopathological analysis and immunostaining of intestinal tissues.Results. Various strains colonised the mouse model, and analysis showed that although clinical signs of disease were not seen, S. aureus infection induced damage in the small intestine, disrupting host structures essential for epithelial integrity. Studies using a staphylococcal enterotoxin B mutant showed that this toxin may contribute to damage during gastrointestinal infection.Conclusion. This work presents a new mouse model of S. aureus gastrointestinal infection, while also providing insight into the pathogenesis of S. aureus in the gut.


Asunto(s)
Intestino Delgado/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Animales , Modelos Animales de Enfermedad , Enterotoxinas/genética , Enterotoxinas/metabolismo , Enterotoxinas/toxicidad , Heces/microbiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Staphylococcus aureus/genética
11.
J Antimicrob Chemother ; 75(2): 409-417, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31642901

RESUMEN

BACKGROUND: Clostridioides difficile (previously Clostridium difficile) is the leading cause of nosocomial, antibiotic-associated diarrhoea worldwide. Currently, the gold standard of treatment for C. difficile infection (CDI) is vancomycin or metronidazole, although these antibiotics also perturb the protective resident microbiota, often resulting in disease relapse. Thus, an urgent need remains for the development of new treatment strategies. Auranofin is an FDA-approved oral antirheumatic drug that was previously shown to inhibit C. difficile vegetative cell growth, toxin production and spore production in vitro. OBJECTIVES: To determine the efficacy of auranofin as a CDI therapeutic by examining the effect of treatment on toxin and spore production in vitro and in vivo, and on disease outcomes in mice. METHODS: C. difficile cultures were treated with auranofin and examined for effects on sporulation and toxin production by sporulation assay and ELISA, respectively. Mice were pretreated with auranofin prior to infection with C. difficile and monitored for physiological conditions, survival and gut damage compared with control animals. Faeces from mice were analysed to determine whether auranofin reduces sporulation and toxin production in vivo. RESULTS: Auranofin significantly reduces sporulation and toxin production under in vitro conditions and in infected mice in vivo. Mice treated with auranofin lost less weight, displayed a significant increase in survival rates and had significantly less toxin-mediated damage in their colon and caecum compared with control mice. CONCLUSIONS: Auranofin shows promise as a prospective therapeutic option for C. difficile infections.


Asunto(s)
Antibacterianos/farmacología , Auranofina/farmacología , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium , Reposicionamiento de Medicamentos , Animales , Infecciones por Clostridium/tratamiento farmacológico , Ratones , Estudios Prospectivos
12.
Nat Microbiol ; 4(12): 2237-2245, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31406331

RESUMEN

Spore-forming bacteria encompass a diverse range of genera and species, including important human and animal pathogens, and food contaminants. Clostridioides difficile is one such bacterium and is a global health threat because it is the leading cause of antibiotic-associated diarrhoea in hospitals. A crucial mediator of C. difficile disease initiation, dissemination and re-infection is the formation of spores that are resistant to current therapeutics, which do not target sporulation. Here, we show that cephamycin antibiotics inhibit C. difficile sporulation by targeting spore-specific penicillin-binding proteins. Using a mouse disease model, we show that combined treatment with the current standard-of-care antibiotic, vancomycin, and a cephamycin prevents disease recurrence. Cephamycins were found to have broad applicability as an anti-sporulation strategy, as they inhibited sporulation in other spore-forming pathogens, including the food contaminant Bacillus cereus. This study could directly and immediately affect treatment of C. difficile infection and advance drug development to control other important spore-forming bacteria that are problematic in the food industry (B. cereus), are potential bioterrorism agents (Bacillus anthracis) and cause other animal and human infections.


Asunto(s)
Antibacterianos/farmacología , Cefamicinas/farmacología , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/prevención & control , Animales , Toxinas Bacterianas/genética , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Clostridioides difficile/genética , Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/microbiología , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a las Penicilinas/efectos de los fármacos , Proteínas de Unión a las Penicilinas/genética , Esporas Bacterianas/efectos de los fármacos , Vancomicina/farmacología , Células Vero/efectos de los fármacos
13.
Eur J Med Chem ; 170: 203-224, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-30901686

RESUMEN

Clostridioides (formerly Clostridium) difficile is a Gram-positive anaerobic bacterial pathogen that causes severe gastrointestinal infection in humans. The current chemotherapeutic options are vastly inadequate, expensive and limited; this results in an exorbitant medical and financial burden. New, inexpensive chemotherapeutic treatments for C. difficile infection with improved efficacy are urgently needed. A streamlined synthetic pathway was developed to allow access to 38 novel mono- and di-cationic biaryl 1,2,3-triazolyl peptidomimetics with increased synthetic efficiency, aqueous solubility and enhanced antibacterial efficacy. The monocationic arginine derivative 28 was identified as a potent, Gram-positive selective antibacterial with MIC values of 4 µg/mL against methicillin-resistant Staphylococcus aureus and 8 µg/mL against C. difficile. Furthermore, the dicationic bis-triazole analogue 50 was found to exhibit broad-spectrum activity with substantial Gram-negative efficacy against Acinetobacter baumannii (8 µg/mL), Pseudomonas aeruginosa (8 µg/mL) and Klebsiella pneumoniae (16 µg/mL); additionally, compound 50 displayed reduced haemolytic activity (<13%) in an in vitro haemolysis assay. Membrane-disruption assays were conducted on selected derivatives to confirm the membrane-active mechanism of action inherent to the synthesized amphiphilic compounds. A comparative solubility assay was developed and utilized to optimize the aqueous solubility of the compounds for in vivo studies. The biaryl peptidomimetics 28 and 67 were found to exhibit significant efficacy in an in vivo murine model of C. difficile infection by reducing the severity and slowing the onset of disease.


Asunto(s)
Antibacterianos/química , Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/tratamiento farmacológico , Peptidomiméticos/química , Peptidomiméticos/uso terapéutico , Animales , Antibacterianos/síntesis química , Antibacterianos/farmacología , Cationes/síntesis química , Cationes/química , Cationes/farmacología , Cationes/uso terapéutico , Humanos , Masculino , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Peptidomiméticos/síntesis química , Peptidomiméticos/farmacología , Triazoles/síntesis química , Triazoles/química , Triazoles/farmacología , Triazoles/uso terapéutico
14.
Anaerobe ; 55: 117-123, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30500477

RESUMEN

Three patients with severe Clostridium difficile infection (CDI) caused by an unusual strain of C. difficile, PCR ribotype (RT) 251, were identified in New South Wales, Australia. All cases presented with severe diarrhoea, two had multiple recurrences and one died following a colectomy. C. difficile RT251 strains were isolated by toxigenic culture. Genetic characterisation was performed using techniques including toxin gene profiling, PCR ribotyping, whole genome sequencing (WGS), in-silico multi-locus-sequence-typing (MLST) and core-genome single nucleotide variant (SNV) analyses. Antimicrobial susceptibility was determined using an agar incorporation method. In vitro toxin production was confirmed by Vero cell cytotoxicity assay and pathogenicity was assessed in a murine model of CDI. All RT251 isolates contained toxin A (tcdA), toxin B (tcdB) and binary toxin (cdtA and cdtB) genes. Core-genome analyses revealed the RT251 strains were clonal, with 0-5 SNVs between isolates. WGS and MLST clustered RT251 in the same evolutionary clade (clade 2) as RT027. Despite comparatively lower levels of in vitro toxin production, in the murine model RT251 infection resembled RT027 infection. Mice showed marked weight loss, severe disease within 48 h post-infection and death. All isolates were susceptible to metronidazole and vancomycin. Our observations suggest C. difficile RT251 causes severe disease and emphasise the importance of ongoing surveillance for new and emerging strains of C. difficile with enhanced virulence.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Proteínas Bacterianas/metabolismo , Clostridioides difficile/clasificación , Clostridioides difficile/aislamiento & purificación , Infecciones por Clostridium/epidemiología , Infecciones por Clostridium/patología , Ribotipificación , Adulto , Anciano , Animales , Bioensayo , Chlorocebus aethiops , Clostridioides difficile/genética , Clostridioides difficile/metabolismo , Femenino , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Tipificación de Secuencias Multilocus , Nueva Gales del Sur , Sobrevida , Células Vero , Secuenciación Completa del Genoma , Adulto Joven
15.
Hum Vaccin Immunother ; 15(2): 508-513, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30277834

RESUMEN

Debilitating recurrent urinary tract infections (UTIs) are often associated with gastrointestinal colonisation by uropathogens, such as uropathogenic Escherichia coli (UPEC), suggesting that these populations might be a suitable target for the treatment and prevention of recurrent UTI. However, antimicrobial treatment is generally unable to prevent recurrent UTI, and often selects for multidrug resistant uropathogens in the gut, and causes dysbiosis of the gut, vaginal, and urinary microbiota. Of note, the globally-disseminated multi drug resistant UPEC lineage, ST131, is known to both persistently colonise the gut and the urinary tract, and is associated with antibiotic treatment failure, indicating the need for novel non-antibiotic therapeutics for the treatment of UTI. This study therefore presents hyperimmune bovine colostrum (HBC) as a suitable therapy for the treatment of UPEC gastrointestinal colonisation. This work demonstrates that the vaccination of pregnant cows with inactivated cells from a ST131 UPEC isolate results in a highly specific anti-UPEC HBC, and that this product is able to disrupt the gastrointestinal colonisation of ST131 UPEC in mice.


Asunto(s)
Calostro/inmunología , Infecciones por Escherichia coli/terapia , Tracto Gastrointestinal/microbiología , Inmunoterapia/métodos , Animales , Bovinos , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple , Infecciones por Escherichia coli/microbiología , Femenino , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Escherichia coli Uropatógena/efectos de los fármacos
16.
J Infect ; 77(5): 417-426, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29964142

RESUMEN

OBJECTIVES: Antibiotic-associated diarrhoea (AAD) caused by C. difficile is one of the most common nosocomial infections, however, little is known about infections related to antimicrobial use for pathogens other than C. difficile. We therefore aimed to provide insight into other bacterial causes of AAD, and how infection with these pathogens causes damage in the dysbiotic gut. METHODS: Clinical isolates from C. difficile-negative AAD patients were whole genome sequenced for in silico analysis of potential virulence factors and antimicrobial resistance determinants. A mouse model of infection was developed to assess the capacity of these isolates to cause gastrointestinal damage, which was analysed by studying specific markers in the gastrointestinal mucosa of infected mice. RESULTS: Several bacterial pathogens were isolated from patients with C. difficile-negative AAD. Each isolate showed the potential for virulence based on encoded virulence factors, as well as most showing antimicrobial resistance in vitro. Isolates of Escherichia coli, Pseudomonas aeruginosa, and Klebsiella pneumoniae were tested in the mouse model of infection, inducing damage primarily in the small intestine, affecting adherens junction integrity, cellular polarity, and cellular proliferation. CONCLUSIONS: Several pathogens of clinical importance other than C. difficile are able to cause gastrointestinal infection following antimicrobial-mediated dysbiosis. The virulence potential and multidrug resistance identified in these isolates illuminates the importance of further diagnostic screening in cases of C. difficile-negative AAD.


Asunto(s)
Antibacterianos/efectos adversos , Diarrea/microbiología , Disbiosis/etiología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/patología , Variación Genética , Animales , Antibacterianos/uso terapéutico , Diarrea/tratamiento farmacológico , Farmacorresistencia Bacteriana Múltiple , Enterotoxinas , Escherichia coli/aislamiento & purificación , Escherichia coli/patogenicidad , Heces/microbiología , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Tracto Gastrointestinal/microbiología , Humanos , Klebsiella pneumoniae/aislamiento & purificación , Klebsiella pneumoniae/patogenicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Pseudomonas aeruginosa/aislamiento & purificación , Pseudomonas aeruginosa/patogenicidad , Factores de Virulencia/genética , Secuenciación Completa del Genoma
17.
J Infect Dis ; 218(6): 979-990, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-29471349

RESUMEN

Background: Epidemiological studies point to the gut as a key reservoir of multidrug resistant Escherichia coli multilocus sequence type 131 (ST131), a globally dominant pathogenic clone causing urinary tract and bloodstream infections. Here we report a detailed investigation of its intestinal lifestyle. Methods: Clinical ST131 isolates and type 1 fimbriae null mutants were assessed for colonization of human intestinal epithelia and in mouse intestinal colonization models. Mouse gut tissue underwent histologic analysis for pathology and ST131 localization. Key findings were corroborated in mucus-producing human cell lines and intestinal biopsy specimens. Results: ST131 strains adhered to and invaded human intestinal epithelial cells more than probiotic and commensal strains. The reference ST131 strain EC958 established persistent intestinal colonization in mice, and expression of type 1 fimbriae mediated higher colonization levels. Bacterial loads were highest in the distal parts of the mouse intestine and did not cause any obvious pathology. Further analysis revealed that EC958 could bind to both mucus and underlying human intestinal epithelia. Conclusions: ST131 strains can efficiently colonize the mammalian gut and persist long term. Type 1 fimbriae enhance ST131 intestinal colonization, suggesting that mannosides, currently developed as therapeutics for bladder infections and Crohn's disease, could also be used to limit intestinal ST131 reservoirs.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple , Infecciones por Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Intestinos/microbiología , Animales , Adhesión Bacteriana , Carga Bacteriana , Células CACO-2 , Línea Celular , Células Epiteliales/citología , Células Epiteliales/microbiología , Escherichia coli/clasificación , Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Femenino , Fimbrias Bacterianas/metabolismo , Humanos , Intestinos/citología , Ratones
18.
Sci Rep ; 7(1): 3665, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28623367

RESUMEN

The increased incidence of antibiotic resistant 'superbugs' has amplified the use of broad spectrum antibiotics worldwide. An unintended consequence of antimicrobial treatment is disruption of the gastrointestinal microbiota, resulting in susceptibility to opportunistic pathogens, such as Clostridium difficile. Paradoxically, treatment of C. difficile infections (CDI) also involves antibiotic use, leaving patients susceptible to re-infection. This serious health threat has led to an urgent call for the development of new therapeutics to reduce or replace the use of antibiotics to treat bacterial infections. To address this need, we have developed colostrum-derived antibodies for the prevention and treatment of CDI. Pregnant cows were immunised to generate hyperimmune bovine colostrum (HBC) containing antibodies that target essential C. difficile virulence components, specifically, spores, vegetative cells and toxin B (TcdB). Mouse infection and relapse models were used to compare the capacity of HBC to prevent or treat primary CDI as well as prevent recurrence. Administration of TcdB-specific colostrum alone, or in combination with spore or vegetative cell-targeted colostrum, prevents and treats C. difficile disease in mice and reduces disease recurrence by 67%. C. difficile-specific colostrum should be re-considered as an immunotherapeutic for the prevention or treatment of primary or recurrent CDI.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/microbiología , Clostridioides difficile/inmunología , Infecciones por Clostridium/veterinaria , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Profilaxis Antibiótica , Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Especificidad de Anticuerpos/inmunología , Proteínas Bacterianas/inmunología , Bovinos , Enfermedades de los Bovinos/tratamiento farmacológico , Enfermedades de los Bovinos/patología , Clostridioides difficile/efectos de los fármacos , Reacciones Cruzadas/inmunología , Ratones , Pruebas de Neutralización , Recurrencia , Proteínas Represoras/inmunología
19.
Artículo en Inglés | MEDLINE | ID: mdl-28634572

RESUMEN

The human pathogen Helicobacter pylori acquires cholesterol from membrane raft domains in eukaryotic cells, commonly known as "lipid rafts." Incorporation of this cholesterol into the H. pylori cell membrane allows the bacterium to avoid clearance by the host immune system and to resist the effects of antibiotics and antimicrobial peptides. The presence of cholesterol in H. pylori bacteria suggested that this pathogen may have cholesterol-enriched domains within its membrane. Consistent with this suggestion, we identified a hypothetical H. pylori protein (HP0248) with homology to the flotillin proteins normally found in the cholesterol-enriched domains of eukaryotic cells. As shown for eukaryotic flotillin proteins, HP0248 was detected in detergent-resistant membrane fractions of H. pylori. Importantly, H. pylori HP0248 mutants contained lower levels of cholesterol than wild-type bacteria (P < 0.01). HP0248 mutant bacteria also exhibited defects in type IV secretion functions, as indicated by reduced IL-8 responses and CagA translocation in epithelial cells (P < 0.05), and were less able to establish a chronic infection in mice than wild-type bacteria (P < 0.05). Thus, we have identified an H. pylori flotillin protein and shown its importance for bacterial virulence. Taken together, the data demonstrate important roles for H. pylori flotillin in host-pathogen interactions. We propose that H. pylori flotillin may be required for the organization of virulence proteins into membrane raft-like structures in this pathogen.


Asunto(s)
Proteínas Bacterianas/metabolismo , Colesterol/metabolismo , Células Epiteliales/metabolismo , Células Eucariotas/metabolismo , Helicobacter pylori/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/genética , Línea Celular , Membrana Celular/metabolismo , Colesterol/inmunología , Citocinas , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Regulación Bacteriana de la Expresión Génica , Infecciones por Helicobacter , Helicobacter pylori/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Interleucina-8/metabolismo , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Mutación , Células RAW 264.7 , Proteínas Recombinantes , Sistemas de Secreción Tipo IV/metabolismo , Virulencia
20.
Hum Vaccin Immunother ; 13(4): 921-927, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-27835052

RESUMEN

Rotavirus gastroenteritis is a leading global cause of mortality and morbidity in young children due to diarrhea and dehydration. Over 85% of deaths occur in developing countries. In industrialised countries, 2 live oral rotavirus vaccines licensed in 2006 quickly demonstrated high effectiveness, dramatically reducing severe rotavirus gastroenteritis admissions in many settings by more than 90%. In contrast, the same vaccines reduced severe rotavirus gastroenteritis by only 30-60% in developing countries, but have been proven life-saving. Bridging this "efficacy gap" offers the possibility to save many more lives of children under the age of 5. The reduced efficacy of rotavirus vaccines in developing settings may be related to differences in transmission dynamics, as well as host luminal, mucosal and immune factors. This review will examine strategies currently under study to target the issue of reduced efficacy and effectiveness of oral rotavirus vaccines in developing settings.


Asunto(s)
Gastroenteritis/prevención & control , Infecciones por Rotavirus/prevención & control , Vacunas contra Rotavirus/inmunología , Países en Desarrollo , Gastroenteritis/epidemiología , Humanos , Infecciones por Rotavirus/epidemiología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA