Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Appl Microbiol ; 134(6)2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37296327

RESUMO

AIMS: The care of patients undergoing long-term urethral catheterization is frequently complicated by Proteus mirabilis infection. This organism forms dense, crystalline biofilms, which block catheters leading to serious clinical conditions. However, there are currently no truly effective approaches to control this problem. Here, we describe the development of a novel theranostic catheter coating, to simultaneously provide early warning of blockage, and actively delay crystalline biofilm formation. METHODS AND RESULTS: The coating comprises of a pH sensitive upper polymer layer (poly(methyl methacrylate-co-methacrylic acid); Eudragit S 100®) and a hydrogel base layer of poly(vinyl alcohol), which is loaded with therapeutic agents (acetohydroxamic acid or ciprofloxacin hydrochloride) and a fluorescent dye, 5(6)-carboxyfluorescein (CF). The elevation of urinary pH due to P. mirabilis urease activity results in the dissolution of the upper layer and release of cargo agents contained in the base layer. Experiments using in vitro models, which were representative of P. mirabilis catheter-associated urinary tract infections, demonstrated that these coatings significantly delay time taken for catheters to block. Coatings containing both CF dye and ciprofloxacin HCl were able to provide an average of ca. 79 h advanced warning of blockage and extend catheter lifespan ca. 3.40-fold. CONCLUSIONS: This study has demonstrated the potential for theranostic, infection-responsive coatings to form a promising approach to combat catheter encrustation and actively delay blockage.


Assuntos
Infecções por Proteus , Infecções Urinárias , Humanos , Cateteres Urinários , Cateterismo Urinário/efeitos adversos , Infecções por Proteus/prevenção & controle , Infecções por Proteus/etiologia , Proteus mirabilis , Infecções Urinárias/prevenção & controle , Biofilmes
2.
Appl Microbiol Biotechnol ; 105(3): 1063-1078, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33427933

RESUMO

Members of the human gut microbiota use glycoside hydrolase (GH) enzymes, such as ß-galactosidases, to forage on host mucin glycans and dietary fibres. A human faecal metagenomic fosmid library was constructed and functionally screened to identify novel ß-galactosidases. Out of the 16,000 clones screened, 30 ß-galactosidase-positive clones were identified. The ß-galactosidase gene found in the majority of the clones was BAD_1582 from Bifidobacterium adolescentis, subsequently named bgaC. This gene was cloned with a hexahistidine tag, expressed in Escherichia coli and His-tagged-BgaC was purified using Ni2+-NTA affinity chromatography and size filtration. The enzyme had optimal activity at pH 7.0 and 37 °C, with a wide range of pH (4-10) and temperature (0-40 °C) stability. It required a divalent metal ion co-factor; maximum activity was detected with Mg2+, while Cu2+ and Mn2+ were inhibitory. Kinetic parameters were determined using ortho-nitrophenyl-ß-D-galactopyranoside (ONPG) and lactose substrates. BgaC had a Vmax of 107 µmol/min/mg and a Km of 2.5 mM for ONPG and a Vmax of 22 µmol/min/mg and a Km of 3.7 mM for lactose. It exhibited low product inhibition by galactose with a Ki of 116 mM and high tolerance for glucose (66% activity retained in presence of 700 mM glucose). In addition, BgaC possessed transglycosylation activity to produce galactooligosaccharides (GOS) from lactose, as determined by TLC and HPLC analysis. The enzymatic characteristics of B. adolescentis BgaC make it an ideal candidate for dairy industry applications and prebiotic manufacture.Key points• Bifidobacterium adolescentis BgaC ß-galactosidase was selected from human faecal metagenome.• BgaC possesses sought-after properties for biotechnology, e.g. low product inhibition.• BgaC has transglycosylation activity producing prebiotic oligosaccharides. Graphical Abstract.


Assuntos
Bifidobacterium adolescentis , Galactose , Humanos , Concentração de Íons de Hidrogênio , Lactose , Metagenoma , Oligossacarídeos , Temperatura , beta-Galactosidase/genética
3.
Antimicrob Agents Chemother ; 60(3): 1530-6, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26711744

RESUMO

Proteus mirabilis forms dense crystalline biofilms on catheter surfaces that occlude urine flow, leading to serious clinical complications in long-term catheterized patients, but there are presently no truly effective approaches to control catheter blockage by this organism. This study evaluated the potential for bacteriophage therapy to control P. mirabilis infection and prevent catheter blockage. Representative in vitro models of the catheterized urinary tract, simulating a complete closed drainage system as used in clinical practice, were employed to evaluate the performance of phage therapy in preventing blockage. Models mimicking either an established infection or early colonization of the catheterized urinary tract were treated with a single dose of a 3-phage cocktail, and the impact on time taken for catheters to block, as well as levels of crystalline biofilm formation, was measured. In models of established infection, phage treatment significantly increased time taken for catheters to block (∼ 3-fold) compared to untreated controls. However, in models simulating early-stage infection, phage treatment eradicated P. mirabilis and prevented blockage entirely. Analysis of catheters from models of established infection 10 h after phage application demonstrated that phage significantly reduced crystalline biofilm formation but did not significantly reduce the level of planktonic cells in the residual bladder urine. Taken together, these results show that bacteriophage constitute a promising strategy for the prevention of catheter blockage but that methods to deliver phage in sufficient numbers and within a key therapeutic window (early infection) will also be important to the successful application of phage to this problem.


Assuntos
Bacteriófagos/patogenicidade , Terapia por Fagos/métodos , Infecções por Proteus/terapia , Proteus mirabilis/virologia , Cateterismo Urinário/efeitos adversos , Cateteres Urinários/microbiologia , Bacteriófagos/isolamento & purificação , Biofilmes/crescimento & desenvolvimento , Cateteres de Demora/microbiologia , Drenagem , Humanos , Microscopia Eletrônica de Transmissão , Modelos Biológicos
4.
J Antimicrob Chemother ; 69(8): 2215-23, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24710024

RESUMO

OBJECTIVES: Selective digestive decontamination (SDD) is an infection prevention measure for critically ill patients in intensive care units (ICUs) that aims to eradicate opportunistic pathogens from the oropharynx and intestines, while sparing the anaerobic flora, by the application of non-absorbable antibiotics. Selection for antibiotic-resistant bacteria is still a major concern for SDD. We therefore studied the impact of SDD on the reservoir of antibiotic resistance genes (i.e. the resistome) by culture-independent approaches. METHODS: We evaluated the impact of SDD on the gut microbiota and resistome in a single ICU patient during and after an ICU stay by several metagenomic approaches. We also determined by quantitative PCR the relative abundance of two common aminoglycoside resistance genes in longitudinally collected samples from 12 additional ICU patients who received SDD. RESULTS: The patient microbiota was highly dynamic during the hospital stay. The abundance of antibiotic resistance genes more than doubled during SDD use, mainly due to a 6.7-fold increase in aminoglycoside resistance genes, in particular aph(2″)-Ib and an aadE-like gene. We show that aph(2″)-Ib is harboured by anaerobic gut commensals and is associated with mobile genetic elements. In longitudinal samples of 12 ICU patients, the dynamics of these two genes ranged from a ∼10(4) fold increase to a ∼10(-10) fold decrease in relative abundance during SDD. CONCLUSIONS: ICU hospitalization and the simultaneous application of SDD has large, but highly individualized, effects on the gut resistome of ICU patients. Selection for transferable antibiotic resistance genes in anaerobic commensal bacteria could impact the risk of transfer of antibiotic resistance genes to opportunistic pathogens.


Assuntos
Antibacterianos/uso terapêutico , Descontaminação/métodos , Farmacorresistência Bacteriana/genética , Intestinos/microbiologia , Orofaringe/microbiologia , Antibacterianos/administração & dosagem , Técnicas de Tipagem Bacteriana , Sequência de Bases , Clostridium/efeitos dos fármacos , Clostridium/isolamento & purificação , Cuidados Críticos , DNA Bacteriano/genética , Fezes/microbiologia , Humanos , Masculino , Microbiota/efeitos dos fármacos , Microbiota/genética , Dados de Sequência Molecular , Fosfotransferases (Aceptor do Grupo Álcool)/genética , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Simbiose
5.
Biotechnol Bioeng ; 110(10): 2573-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23568752

RESUMO

Proteus mirabilis is a prominent cause of catheter-associated urinary tract infections (CAUTIs) among patients undergoing long-term bladder catheterization. There are currently no effective means of preventing P. mirabilis infections, and strategies for prophylaxis and rapid early diagnosis are urgently required. Aptamers offer significant potential for development of countermeasures against P. mirabilis CAUTI and are an ideal class of molecules for the development of diagnostics and therapeutics. Here we demonstrate the application of Cell-SELEX to identify DNA aptamers that show high affinity for P. mirabilis. While the aptamers identified displayed high affinity for P. mirabilis cells in dot blotting assays, they also bound to other uropathogenic bacteria. To improve aptamer specificity for P. mirabilis, an in silico maturation (ISM) approach was employed. Two cycles of ISM allowed the identification of an aptamer showing 36% higher specificity, evaluated as a ratio of binding signal for P. mirabilis to that for Escherichia coli (also a cause of CAUTI and the most common urinary tract pathogen). Aptamers that specifically recognize P. mirabilis would have diagnostic and therapeutic values and constitute useful tools for studying membrane-associated proteins in this organism.


Assuntos
Aptâmeros de Nucleotídeos/química , Simulação por Computador , Proteus mirabilis , Técnica de Seleção de Aptâmeros/métodos , Aptâmeros de Nucleotídeos/metabolismo , Escherichia coli , Proteus mirabilis/isolamento & purificação , Proteus mirabilis/metabolismo , Sensibilidade e Especificidade
6.
Biosens Bioelectron ; 203: 114027, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35114463

RESUMO

Therapeutic monoclonal antibodies (mAbs) are successful biomedicines; however, evaluation of their pharmacokinetics and pharmacodynamics demands highly specific discrimination from human immunoglobulin G naturally present in the blood. Here, we developed a novel anti-idiotype aptamer (termed A14#1) with extraordinary specificity against the anti-vascular endothelial growth factor therapeutic mAb, bevacizumab. Structural analysis of the antibody-aptamer complex showed that several bases of A14#1 recognized only the complementarity determining region (CDR) of bevacizumab, thereby contributing to its extraordinary specificity. As the CDR of bevacizumab is predicted to be highly positively charged under mildly acidic conditions and that DNA is negatively charged, the affinity of A14#1 to bevacizumab markedly increased at pH 4.7 (KD = 44 pM) than at pH 7.4 (KD = 12 nM). A14#1-based electrochemical detection method capable of detecting 31 pM of bevacizumab at pH 4.7 was thus developed. A14#1 could be potentially useful for therapeutic drug measurement as a novel ligand of bevacizumab.


Assuntos
Aptâmeros de Nucleotídeos , Técnicas Biossensoriais , Anticorpos Monoclonais , Afinidade de Anticorpos , Aptâmeros de Nucleotídeos/química , Regiões Determinantes de Complementaridade/química , Regiões Determinantes de Complementaridade/genética , Humanos , Concentração de Íons de Hidrogênio
7.
Proc Natl Acad Sci U S A ; 105(36): 13580-5, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18757757

RESUMO

Bile salt hydrolases (BSHs) catalyze the "gateway" reaction in a wider pathway of bile acid modification by the gut microbiota. Because bile acids function as signaling molecules regulating their own biosynthesis, lipid absorption, cholesterol homeostasis, and local mucosal defenses in the intestine, microbial BSH activity has the potential to greatly influence host physiology. However, the function, distribution, and abundance of BSH enzymes in the gut community are unknown. Here, we show that BSH activity is a conserved microbial adaptation to the human gut environment with a high level of redundancy in this ecosystem. Through metagenomic analyses we identified functional BSH in all major bacterial divisions and archaeal species in the gut and demonstrate that BSH is enriched in the human gut microbiome. Phylogenetic analysis illustrates that selective pressure in the form of conjugated bile acid has driven the evolution of members of the Ntn_CGH-like family of proteins toward BSH activity in gut-associated species. Furthermore, we demonstrate that BSH mediates bile tolerance in vitro and enhances survival in the murine gut in vivo. Overall, we demonstrate the use of function-driven metagenomics to identify functional anchors in complex microbial communities, and dissect the gut microbiome according to activities relevant to survival in the mammalian gastrointestinal tract.


Assuntos
Amidoidrolases/genética , Amidoidrolases/metabolismo , Trato Gastrointestinal/enzimologia , Metagenoma/genética , Sequência de Aminoácidos , Evolução Molecular , Trato Gastrointestinal/microbiologia , Humanos , Methanobrevibacter/enzimologia , Methanobrevibacter/genética , Filogenia
8.
Front Microbiol ; 12: 730071, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34803947

RESUMO

Phage bacteria interactions can affect structure, dynamics, and function of microbial communities. In the context of biological wastewater treatment (BWT), the presence of phages can alter the efficiency of the treatment process and influence the quality of the treated effluent. The active role of phages in BWT has been demonstrated, but many questions remain unanswered regarding the diversity of phages in these engineered environments, the dynamics of infection, the determination of bacterial hosts, and the impact of their activity in full-scale processes. A deeper understanding of the phage ecology in BWT can lead the improvement of process monitoring and control, promote higher influent quality, and potentiate the use of phages as biocontrol agents. In this review, we highlight suitable methods for studying phages in wastewater adapted from other research fields, provide a critical overview on the current state of knowledge on the effect of phages on structure and function of BWT bacterial communities, and highlight gaps, opportunities, and priority questions to be addressed in future research.

9.
ACS Infect Dis ; 7(5): 1283-1296, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33843198

RESUMO

The contribution of the gut microbiome to human health has long been established, with normal gut microbiota conferring protection against invasive pathogens. Antibiotics can disrupt the microbial balance of the gut, resulting in disease and the development of antimicrobial resistance. The effect of antibiotic administration route on gut dysbiosis remains under-studied to date, with conflicting evidence on the differential effects of oral and parenteral delivery. We have profiled the rat gut microbiome following treatment with commonly prescribed antibiotics (amoxicillin and levofloxacin), via either oral or intravenous administration. Fecal pellets were collected over a 13-day period and bacterial populations were analyzed by 16S rRNA gene sequencing. Significant dysbiosis was observed in all treatment groups, regardless of administration route. More profound dysbiotic effects were observed following amoxicillin treatment than those with levofloxacin, with population richness and diversity significantly reduced, regardless of delivery route. The effect on specific taxonomic groups was assessed, revealing significant disruption following treatment with both antibiotics. Enrichment of a number of groups containing known gut pathogens was observed, in particular, with amoxicillin, such as the family Enterobacteriaceae. Depletion of other commensal groups was also observed. The degree of dysbiosis was significantly reduced toward the end of the sampling period, as bacterial populations began to return to pretreatment composition. Richness and diversity levels appeared to return to pretreatment levels more quickly in intravenous groups, suggesting convenient parenteral delivery systems may have a role to play in reducing longer term gut dysbiosis in the treatment of infection.


Assuntos
Microbioma Gastrointestinal , Animais , Antibacterianos , Disbiose/induzido quimicamente , Enterobacteriaceae , RNA Ribossômico 16S/genética , Ratos
10.
BMC Genomics ; 11: 46, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20085629

RESUMO

BACKGROUND: Little is known regarding the pool of mobile genetic elements associated with the human gut microbiome. In this study we employed the culture independent TRACA system to isolate novel plasmids from the human gut microbiota, and a comparative metagenomic analysis to investigate the distribution and relative abundance of functions encoded by these plasmids in the human gut microbiome. RESULTS: Novel plasmids were acquired from the human gut microbiome, and homologous nucleotide sequences with high identity (>90%) to two plasmids (pTRACA10 and pTRACA22) were identified in the multiple human gut microbiomes analysed here. However, no homologous nucleotide sequences to these plasmids were identified in the murine gut or environmental metagenomes. Functions encoded by the plasmids pTRACA10 and pTRACA22 were found to be more prevalent in the human gut microbiome when compared to microbial communities from other environments. Among the most prevalent functions identified was a putative RelBE toxin-antitoxin (TA) addiction module, and subsequent analysis revealed that this was most closely related to putative TA modules from gut associated bacteria belonging to the Firmicutes. A broad phylogenetic distribution of RelE toxin genes was observed in gut associated bacterial species (Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria), but no RelE homologues were identified in gut associated archaeal species. We also provide indirect evidence for the horizontal transfer of these genes between bacterial species belonging to disparate phylogenetic divisions, namely Gram negative Proteobacteria and Gram positive species from the Firmicutes division. CONCLUSIONS: The application of a culture independent system to capture novel plasmids from the human gut mobile metagenome, coupled with subsequent comparative metagenomic analysis, highlighted the unexpected prevalence of plasmid encoded functions in the gut microbial ecosystem. In particular the increased relative abundance and broad phylogenetic distribution was identified for a putative RelBE toxin/antitoxin addiction module, a putative phosphohydrolase/phosphoesterase, and an ORF of unknown function. Our analysis also indicates that some plasmids or plasmid families are present in the gut microbiomes of geographically isolated human hosts with a broad global distribution (America, Japan and Europe), and are potentially unique to the human gut microbiome. Further investigation of the plasmid population associated with the human gut is likely to provide important insights into the development, functioning and evolution of the human gut microbiota.


Assuntos
Trato Gastrointestinal/microbiologia , Genoma Bacteriano , Metagenoma/genética , Metagenômica/métodos , Plasmídeos/genética , Adulto , Sequência de Aminoácidos , Bactérias/genética , Toxinas Bacterianas/genética , Hibridização Genômica Comparativa , DNA Bacteriano/genética , Humanos , Masculino , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Alinhamento de Sequência , Análise de Sequência de DNA
11.
Viruses ; 12(6)2020 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-32486377

RESUMO

Burkholderia species have environmental, industrial and medical significance, and are important opportunistic pathogens in individuals with cystic fibrosis (CF). Using a combination of existing and newly determined genome sequences, this study investigated prophage carriage across the species B. vietnamiensis, and also isolated spontaneously inducible prophages from a reference strain, G4. Eighty-one B. vietnamiensis genomes were bioinformatically screened for prophages using PHASTER (Phage Search Tool Enhanced Release) and prophage regions were found to comprise up to 3.4% of total genetic material. Overall, 115 intact prophages were identified and there was evidence of polylysogeny in 32 strains. A novel, inducible Mu-like phage (vB_BvM-G4P1) was isolated from B. vietnamiensis G4 that had lytic activity against strains of five Burkholderia species prevalent in CF infections, including the Boston epidemic B. dolosa strain SLC6. The cognate prophage to vB_BvM-G4P1 was identified in the lysogen genome and was almost identical (>93.5% tblastx identity) to prophages found in 13 other B. vietnamiensis strains (17% of the strain collection). Phylogenomic analysis determined that the G4P1-like prophages were widely distributed across the population structure of B. vietnamiensis. This study highlights how genomic characterization of Burkholderia prophages can lead to the discovery of novel bacteriophages with potential therapeutic or biotechnological applications.


Assuntos
Burkholderia/virologia , Lisogenia , Prófagos/patogenicidade , Burkholderia/genética , Infecções por Burkholderia/microbiologia , Cromossomos Bacterianos/genética , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Genoma Bacteriano/genética , Genoma Viral/genética , Humanos , Lisogenia/fisiologia , Microscopia Eletrônica de Transmissão , Filogenia , Prófagos/genética , Prófagos/fisiologia , Ativação Viral
12.
Methods Mol Biol ; 2021: 139-158, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31309503

RESUMO

Urethral catheters are among the most widely used medical devices, applied to manage a wide range of conditions in hospital, community, and care home settings. In long-term catheterized individuals, infection with Proteus mirabilis frequently complicates the care of patients owing to formation of extensive crystalline biofilms. Here we describe the use of an in vitro bladder model of the catheterized urinary tract and associated analyses to study P. mirabilis crystalline biofilm formation. The model originally described by Stickler et al. (1999, 310:494-501, Methods Enzymol) replicates a complete sterile closed drainage system as used in clinical practice, and permits formation of biofilms directly on catheters under conditions representative of those encountered in vivo. Models may be used to replicate either established infection or early stage colonization, and we describe a range of associated methods for quantification and visualization of biofilms formed on catheters. These methods are also easily adapted to study catheter-associated biofilm formation by other urinary tract pathogens.


Assuntos
Infecções Relacionadas a Cateter/diagnóstico , Infecções por Proteus/diagnóstico , Proteus mirabilis/fisiologia , Infecções Urinárias/microbiologia , Técnicas Bacteriológicas , Biofilmes , Humanos , Técnicas In Vitro , Modelos Biológicos , Proteus mirabilis/isolamento & purificação , Cateteres Urinários/microbiologia
13.
Proc Inst Mech Eng H ; 233(1): 68-83, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29807465

RESUMO

Urinary catheters have been used on an intermittent or indwelling basis for centuries, in order to relieve urinary retention and incontinence. Nevertheless, the use of urinary catheters in the clinical setting is fraught with complication, the most common of which is the development of nosocomial urinary tract infections, known as catheter-associated urinary tract infections. Infections of this nature are not only significant owing to their high incidence rate and subsequent economic burden but also to the severe medical consecutions that result. A range of techniques have been employed in recent years, utilising various technologies in attempts to counteract the perilous medical cascade following catheter blockage. This review will focus on the current advancement (within the last 10 years) in prevention of encrustation and blockage of long-term indwelling catheters both from engineering and medical perspectives, with particular emphasis on the importance of stimuli-responsive systems.


Assuntos
Cateteres de Demora , Engenharia/métodos , Cateteres Urinários , Antibacterianos/farmacologia , Cateteres de Demora/efeitos adversos , Falha de Equipamento , Humanos
14.
Front Microbiol ; 10: 1783, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31447809

RESUMO

Proteus mirabilis often complicates the care of catheterized patients through the formation of crystalline biofilms which block urine flow. Bacteriophage therapy has been highlighted as a promising approach to control this problem, but relatively few phages infecting P. mirabilis have been characterized. Here we characterize five phages capable of infecting P. mirabilis, including those shown to reduce biofilm formation, and provide insights regarding the wider ecological and evolutionary relationships of these phages. Transmission electron microscopy (TEM) imaging of phages vB_PmiP_RS1pmA, vB_PmiP_RS1pmB, vB_PmiP_RS3pmA, and vB_PmiP_RS8pmA showed that all share morphologies characteristic of the Podoviridae family. The genome sequences of vB_PmiP_RS1pmA, vB_PmiP_RS1pmB, and vB_PmiP_RS3pmA showed these are species of the same phage differing only by point mutations, and are closely related to vB_PmiP_RS8pmA. Podophages characterized in this study were also found to share similarity in genome architecture and composition to other previously described P. mirabilis podophages (PM16 and PM75). In contrast, vB_PimP_RS51pmB showed morphology characteristic of the Myoviridae family, with no notable similarity to other phage genomes examined. Ecogenomic profiling of all phages revealed no association with human urinary tract viromes, but sequences similar to vB_PimP_RS51pmB were found within human gut, and human oral microbiomes. Investigation of wider host-phage evolutionary relationships through tetranucleotide profiling of phage genomes and bacterial chromosomes, indicated vB_PimP_RS51pmB has a relatively recent association with Morganella morganii and other non-Proteus members of the Morganellaceae family. Subsequent host range assays confirmed vB_PimP_RS51pmB can infect M. morganii.

15.
J Chromatogr A ; 1600: 127-136, 2019 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-31047664

RESUMO

Faeces are comprised of a wide array of metabolites arising from the circulatory system as well as the human microbiome. A global metabolite analysis (metabolomics) of faecal extracts offers the potential to uncover new compounds which may be indicative of the onset of bowel diseases such as colorectal cancer (CRC). To date, faecal metabolomics is still in its infancy and the compounds of low abundance present in faecal extracts poorly characterised. In this study, extracts of faeces from healthy subjects were profiled using a sensitive nanoflow-nanospray LC-MS platform which resulted in highly repeatable peak retention times (<2% CV) and intensities (<15% CV). Analysis of the extracts revealed wide coverage of the faecal metabolome including detection of low abundant signalling compounds such as sex steroids and eicosanoids, alongside highly abundant pharmaceuticals and tetrapyrrole metabolites. A small pilot study investigating differences in metabolomics profiles of faecal samples obtained from 7 CRC, 25 adenomatous polyp and 26 healthy groups revealed that secondary bile acids, conjugated androgens, eicosanoids, phospholipids and an unidentified haem metabolite were potential classes of metabolites that discriminated between the CRC and control sample groups. However, much larger follow up studies are needed to confirm which components of the faecal metabolome are associated with actual CRC disease rather than dietary influences. This study reveals the potential of nanospray-nanoflow LC-MS profiling of faecal samples from large scale cohort studies for uncovering the role of the faecal metabolome in colorectal disease formation.


Assuntos
Cromatografia Líquida , Fezes/química , Metaboloma , Espectrometria de Massas por Ionização por Electrospray , Ácidos e Sais Biliares/análise , Eicosanoides/análise , Feminino , Voluntários Saudáveis , Humanos , Masculino , Metabolômica , Fosfolipídeos/análise , Projetos Piloto
16.
Environ Microbiol ; 10(7): 1888-902, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18430018

RESUMO

Samples of the sponge Haliclona simulans were collected from Irish waters and subjected to a culture-independent analysis to determine the microbial, polyketide synthase (PKS) and non-ribosomal peptide synthase (NRPS) diversity. 16S rRNA gene libraries were prepared from total sponge, bacterial enriched sponge and seawater samples. Eight phyla from the Bacteria were detected in the sponge by phylogenetic analyses of the 16S rRNA gene libraries. The most abundant phylum in the total sponge library was the Proteobacteria (86%), with the majority of these clones being from the gamma-Proteobacteria (77%); two groups of clones were dominant and together made up 69% of the total. Both of these groups were related to other sponge-derived microbes and comprised novel genera. Within the other bacterial phyla groups of clones representing novel candidate genera within the phyla Verrucomicrobia and Lentisphaerae were also found. Selective enrichment of the bacterial component of the sponge prior to 16S rRNA gene analysis resulted in a 16S rRNA gene library dominated by a novel genus of delta-Proteobacteria, most closely related to the Bdellovibrio. The potential for the sponge microbiota to produce secondary metabolites was also analysed by polymerase chain reaction amplification of PKS and NRPS genes. While no NRPS sequences were isolated seven ketosynthase (KS) sequences were obtained from the sponge metagenome. Analyses of these clones revealed a diverse collection of PKS sequences which were most closely affiliated with PKS from members of the Cyanobacteria, Myxobacteria and Dinoflagellata.


Assuntos
Biodiversidade , Gammaproteobacteria/enzimologia , Gammaproteobacteria/genética , Haliclona/microbiologia , Policetídeo Sintases/genética , Animais , DNA Bacteriano/análise , DNA Ribossômico/análise , DNA Ribossômico/química , DNA Ribossômico/genética , Gammaproteobacteria/crescimento & desenvolvimento , Biblioteca Genômica , Dados de Sequência Molecular , Peptídeo Sintases/metabolismo , Policetídeo Sintases/metabolismo , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Água do Mar
18.
ISME J ; 12(4): 942-958, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29259289

RESUMO

Just as the expansion in genome sequencing has revealed and permitted the exploitation of phylogenetic signals embedded in bacterial genomes, the application of metagenomics has begun to provide similar insights at the ecosystem level for microbial communities. However, little is known regarding this aspect of bacteriophage associated with microbial ecosystems, and if phage encode discernible habitat-associated signals diagnostic of underlying microbiomes. Here we demonstrate that individual phage can encode clear habitat-related 'ecogenomic signatures', based on relative representation of phage-encoded gene homologues in metagenomic data sets. Furthermore, we show the ecogenomic signature encoded by the gut-associated ɸB124-14 can be used to segregate metagenomes according to environmental origin, and distinguish 'contaminated' environmental metagenomes (subject to simulated in silico human faecal pollution) from uncontaminated data sets. This indicates phage-encoded ecological signals likely possess sufficient discriminatory power for use in biotechnological applications, such as development of microbial source tracking tools for monitoring water quality.


Assuntos
Bacteriófagos/genética , Genoma Viral , Metagenoma , Ecossistema , Monitoramento Ambiental , Fezes/virologia , Trato Gastrointestinal/virologia , Humanos , Metagenômica , Microbiota
19.
Artigo em Inglês | MEDLINE | ID: mdl-29963501

RESUMO

Biofilm formation in wounds is considered a major barrier to successful treatment, and has been associated with the transition of wounds to a chronic non-healing state. Here, we present a novel laboratory model of wound biofilm formation using ex-vivo porcine skin and a custom burn wound array device. The model supports high-throughput studies of biofilm formation and is compatible with a range of established methods for monitoring bacterial growth, biofilm formation, and gene expression. We demonstrate the use of this model by evaluating the potential for bacteriophage to control biofilm formation by Staphylococcus aureus, and for population density dependant expression of S. aureus virulence factors (regulated by the Accessory Gene Regulator, agr) to signal clinically relevant wound infection. Enumeration of colony forming units and metabolic activity using the XTT assay, confirmed growth of bacteria in wounds and showed a significant reduction in viable cells after phage treatment. Confocal laser scanning microscopy confirmed the growth of biofilms in wounds, and showed phage treatment could significantly reduce the formation of these communities. Evaluation of agr activity by qRT-PCR showed an increase in activity during growth in wound models for most strains. Activation of a prototype infection-responsive dressing designed to provide a visual signal of wound infection, was related to increased agr activity. In all assays, excellent reproducibility was observed between replicates using this model.


Assuntos
Biofilmes/crescimento & desenvolvimento , Queimaduras/microbiologia , Pele/lesões , Staphylococcus aureus/crescimento & desenvolvimento , Infecção dos Ferimentos/prevenção & controle , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Queimaduras/patologia , Queimaduras/veterinária , Humanos , Terapia por Fagos/veterinária , Reprodutibilidade dos Testes , Pele/patologia , Infecções Estafilocócicas/patologia , Infecções Estafilocócicas/terapia , Infecções Estafilocócicas/veterinária , Infecções Estafilocócicas/virologia , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/fisiologia , Staphylococcus aureus/virologia , Suínos , Transativadores/genética , Transativadores/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/fisiologia , Infecção dos Ferimentos/terapia , Infecção dos Ferimentos/veterinária , Infecção dos Ferimentos/virologia
20.
Mol Biosyst ; 3(11): 749-58, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17940657

RESUMO

This article outlines current and possible future strategies to access the mobile metagenome of bacterial ecosystems. Evidence for the role of this genetic resource in development and maintenance of core community functions of the human gut microbiota is reviewed.


Assuntos
Bactérias/genética , Trato Gastrointestinal/microbiologia , Genoma Bacteriano/genética , Genômica/métodos , Regulação Bacteriana da Expressão Gênica , Humanos , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa