Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Appl Microbiol Biotechnol ; 107(11): 3621-3636, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37133800

ABSTRACT

Citrobacter koseri is an emerging Gram-negative bacterial pathogen, which causes urinary tract infections. We isolated and characterized a novel S16-like myovirus CKP1 (vB_CkoM_CkP1), infecting C. koseri. CkP1 has a host range covering the whole C. koseri species, i.e., all strains that were tested, but does not infect other species. Its linear 168,463-bp genome contains 291 coding sequences, sharing sequence similarity with the Salmonella phage S16. Based on surface plasmon resonance and recombinant green florescence protein fusions, the tail fiber (gp267) was shown to decorate C. koseri cells, binding with a nanomolar affinity, without the need of accessory proteins. Both phage and the tail fiber specifically bind to bacterial cells by the lipopolysaccharide polymer. We further demonstrate that CkP1 is highly stable towards different environmental conditions of pH and temperatures and is able to control C. koseri cells in urine samples. Altogether, CkP1 features optimal in vitro characteristics to be used both as a control and detection agent towards drug-resistant C. koseri infections. KEY POINTS: • CkP1 infects all C. koseri strains tested • CkP1 recognizes C. koseri lipopolysaccharide through its long tail fiber • Both phage CkP1 and its tail fiber can be used to treat or detect C. koseri pathogens.


Subject(s)
Bacteriophages , Citrobacter koseri , Bacteriophages/genetics , Citrobacter koseri/genetics , Lipopolysaccharides , Host Specificity
2.
BMC Oral Health ; 21(1): 112, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33706748

ABSTRACT

BACKGROUND: Implants are a predictable and well-established treatment method in dentistry. Nevertheless, looking at possible failures of dental implants, early and late loss have to be distinguished. The intent of the study was to report microbiological findings on the surface of implants with severe peri-implantitis, which had to be explanted. METHODS: 53 specimens of implants from 48 patients without severe general illnesses have been examined. The groups investigated were implants that had to be removed in the period of osseointegration (early loss, 13 patients with 14 implants) or after the healing period (late loss, 14 patients with 17 implants). The implant losses were compared with two control groups (implants with no bone loss directly after completed osseointegration, two to four months after implant placement (17 patients with 17 implants) and implants with no bone loss and prosthetic restoration for more than three years (5 patients with 5 implants)). Data about the bacteria located in the peri-implant sulcus was collected using amplification and high throughput sequencing of the 16S rRNA gene. RESULTS: The biofilm composition differed substantially between individuals. Both in early and late implant loss, Fusobacterium nucleatum and Porphyromonas gingivalis were found to be abundant. Late lost implants showed higher bacterial diversity and in addition higher abundances of Treponema, Fretibacterium, Pseudoramibacter and Desulfobulbus, while microbial communities of early loss implants were very heterogeneous and showed no significantly more abundant bacterial taxa. CONCLUSIONS: Specific peri-implant pathogens were found around implants that were lost after a primarily uneventful osseointegration. P. gingivalis and F. nucleatum frequently colonized the implant in early and late losses and could therefore be characteristic for implant loss in general. In general, early lost implants showed also lower microbial diversity than late losses. However, the microbial results were not indicative of the causes of early and late losses.


Subject(s)
Alveolar Bone Loss , Dental Implants , Peri-Implantitis , Humans , Osseointegration , Porphyromonas gingivalis , RNA, Ribosomal, 16S
3.
Food Microbiol ; 92: 103552, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32950146

ABSTRACT

The fermentation of vegetables is a traditional preservation method, that experiences a renaissance even in domestic households. Table salt is added to the fermentation batches to favor the growth of lactic acid bacteria usually. On an industrial scale, the fermentation brine is typically prepared with non-iodized table salt. In our study, we investigated the microbiota of cucumber fermentations using culture-dependent and -independent methods. We could show that the fermentation process of cucumbers and the involved microbiota is influenced by the concentration of table salt and not by the use of iodized table salt. Therefore, we conclude that the use of iodized table salt does not negatively affect the fermentation process. We could verify that iodine permeates the cucumbers by diffusion, leading to satisfactory iodine concentrations in the final food product. The industrial use of iodized table salt in food fermentations could contribute to maintain a constant iodine supply to the general public.


Subject(s)
Cucumis sativus/microbiology , Fermented Foods/microbiology , Iodine/pharmacology , Microbiota/drug effects , Sodium Chloride, Dietary/pharmacology , Sodium Chloride/pharmacology , Cucumis sativus/chemistry , Cucumis sativus/metabolism , Fermentation , Fermented Foods/analysis , Food Microbiology , Iodine/analysis , Salts/analysis , Salts/pharmacology , Sodium Chloride/analysis , Sodium Chloride, Dietary/analysis
4.
J Clin Periodontol ; 46(4): 481-490, 2019 04.
Article in English | MEDLINE | ID: mdl-30941800

ABSTRACT

AIM: Aim of this study was to investigate the influence of an anti-inflammatory diet on different parameters in patients with gingivitis. MATERIALS AND METHODS: Thirty patients were randomly allocated to an experimental and a control group stratified by their plaque values. The experimental group had to change to a diet low in processed carbohydrates and animal proteins, and rich in omega-3 fatty acids, vitamin C, vitamin D, antioxidants, plant nitrates and fibres for 4 weeks. The control group did not change their diet. Both groups suspended interdental cleaning. Periodontal parameters were assessed by a blinded dentist. Serological and subgingival plaque samples were taken at baseline and end. RESULTS: While there were no differences regarding the plaque values, the experimental group showed a significant reduction in gingival bleeding (GI Baseline: 1.04 ± 0.21, GI End: 0.61 ± 0.29, p < 0.05), a significant increase in Vitamin D values and a significant weight loss. There were no inter-group differences regarding the inflammatory serological parameters, the serological omega fatty acids, nor the subgingival microbiome composition. CONCLUSION: The evaluated diet could significantly reduce gingivitis in a clinically relevant range, while serological inflammatory parameters and the subgingival microbiome seem to be unaffected in this study duration. (German Clinical Trials Register; DRKS00009888).


Subject(s)
Gingivitis , Anti-Inflammatory Agents , Dental Plaque Index , Diet , Humans , Periodontal Index
5.
Mol Microbiol ; 103(1): 55-66, 2017 01.
Article in English | MEDLINE | ID: mdl-27671713

ABSTRACT

Propionate is an abundant carboxylic acid in nature. Microorganisms metabolize propionate aerobically via the 2-methylcitrate pathway. This pathway depends on a series of three reactions in the citric acid cycle that leads to the conversion of succinate to oxaloacetate. Interestingly, the γ-proteobacterium Escherichia coli can use propionate as a carbon and electron source under oxic but not under anoxic conditions. RT-PCR and transcriptomic analysis revealed a posttranscriptional regulation of the prpBCDE-gene cluster encoding the necessary enzymes for propionate metabolism. The polycistronic mRNA seems to be hydrolyzed in the 3'-5' direction under anoxic conditions. This regulatory strategy is highly constructive because the last gene of the operon encodes the first enzyme of the propionate metabolism. Further analysis revealed that RNase R is involved in the hydrolysis of the prp transcripts. Consequently, an rnr-deletion strain could metabolize propionate under anoxic conditions. To the best of our knowledge, this is the first study describing the influence of RNase R on the anaerobic metabolism of E. coli.


Subject(s)
Escherichia coli K12/metabolism , Escherichia coli Proteins/metabolism , Exoribonucleases/metabolism , Propionates/metabolism , Anaerobiosis/physiology , Citrates/metabolism , Citric Acid Cycle/physiology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli K12/genetics , Gene Expression Regulation, Bacterial/genetics , Multigene Family/genetics , Operon/genetics
6.
Environ Microbiol ; 19(12): 5060-5077, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29076652

ABSTRACT

Bacteriophages are ubiquitous in nature and represent a vast repository of genetic diversity, which is driven by the endless coevolution cycle with a diversified group of bacterial hosts. Studying phage-host interactions is important to gain novel insights into their dynamic adaptation. In this study, we isolated 12 phages infecting species of the Acinetobacter baumannii-Acinetobacter calcoaceticus complex which exhibited a narrow host range and similar morphological features (podoviruses with short tails of 9-12 nm and isometric heads of 50-60 nm). Notably, the alignment of the newly sequenced phage genomes (40-41 kb of DNA length) and all Acinetobacter podoviruses deposited in Genbank has shown high synteny, regardless of the date and source of isolation that spans from America to Europe and Asia. Interestingly, the C-terminal pectate lyase domain of these phage tail fibres is often the only difference found among these viral genomes, demonstrating a very specific genomic variation during the course of their evolution. We proved that the pectate lyase domain is responsible for phage depolymerase activity and binding to specific Acinetobacter bacterial capsules. We discuss how this mechanism of phage-host co-evolution impacts the tail specificity apparatus of Acinetobacter podoviruses.


Subject(s)
Acinetobacter baumannii/virology , Acinetobacter calcoaceticus/virology , Host Specificity/physiology , Podoviridae/metabolism , Polygalacturonase/metabolism , Polysaccharide-Lyases/metabolism , Asia , Base Sequence , Europe , Genome, Viral/genetics , Podoviridae/classification , Podoviridae/genetics , Protein Domains/physiology , Virion/genetics
7.
PLoS Pathog ; 11(3): e1004744, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25780925

ABSTRACT

Sigma factors are essential global regulators of transcription initiation in bacteria which confer promoter recognition specificity to the RNA polymerase core enzyme. They provide effective mechanisms for simultaneously regulating expression of large numbers of genes in response to challenging conditions, and their presence has been linked to bacterial virulence and pathogenicity. In this study, we constructed nine his-tagged sigma factor expressing and/or deletion mutant strains in the opportunistic pathogen Pseudomonas aeruginosa. To uncover the direct and indirect sigma factor regulons, we performed mRNA profiling, as well as chromatin immunoprecipitation coupled to high-throughput sequencing. We furthermore elucidated the de novo binding motif of each sigma factor, and validated the RNA- and ChIP-seq results by global motif searches in the proximity of transcriptional start sites (TSS). Our integrated approach revealed a highly modular network architecture which is composed of insulated functional sigma factor modules. Analysis of the interconnectivity of the various sigma factor networks uncovered a limited, but highly function-specific, crosstalk which orchestrates complex cellular processes. Our data indicate that the modular structure of sigma factor networks enables P. aeruginosa to function adequately in its environment and at the same time is exploited to build up higher-level functions by specific interconnections that are dominated by a participation of RpoN.


Subject(s)
Host-Parasite Interactions/physiology , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/physiology , Sigma Factor/genetics , Signal Transduction/physiology , Chromatin Immunoprecipitation , Cluster Analysis , High-Throughput Nucleotide Sequencing , Pseudomonas Infections/metabolism , Receptor Cross-Talk/physiology , Sequence Analysis, RNA , Sigma Factor/metabolism , Transcriptome
8.
Antimicrob Agents Chemother ; 60(8): 4722-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27216077

ABSTRACT

Emerging resistance to antimicrobials and the lack of new antibiotic drug candidates underscore the need for optimization of current diagnostics and therapies to diminish the evolution and spread of multidrug resistance. As the antibiotic resistance status of a bacterial pathogen is defined by its genome, resistance profiling by applying next-generation sequencing (NGS) technologies may in the future accomplish pathogen identification, prompt initiation of targeted individualized treatment, and the implementation of optimized infection control measures. In this study, qualitative RNA sequencing was used to identify key genetic determinants of antibiotic resistance in 135 clinical Pseudomonas aeruginosa isolates from diverse geographic and infection site origins. By applying transcriptome-wide association studies, adaptive variations associated with resistance to the antibiotic classes fluoroquinolones, aminoglycosides, and ß-lactams were identified. Besides potential novel biomarkers with a direct correlation to resistance, global patterns of phenotype-associated gene expression and sequence variations were identified by predictive machine learning approaches. Our research serves to establish genotype-based molecular diagnostic tools for the identification of the current resistance profiles of bacterial pathogens and paves the way for faster diagnostics for more efficient, targeted treatment strategies to also mitigate the future potential for resistance evolution.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Transcriptome/genetics , Aminoglycosides/pharmacology , Fluoroquinolones/pharmacology , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Microbial Sensitivity Tests/methods , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , beta-Lactams/pharmacology
10.
Clin Oral Implants Res ; 27(12): e161-e166, 2016 Dec.
Article in English | MEDLINE | ID: mdl-25808842

ABSTRACT

BACKGROUND: Cementing dental restorations on implants poses the risk of undetected excess cement. Such cement remnants may favor the development of inflammation in the peri-implant tissue. The effect of excess cement on the bacterial community is not yet known. The aim of this study was to analyze the effect of two different dental cements on the composition of the microbial peri-implant community. METHODS: In a cohort of 38 patients, samples of the peri-implant tissue were taken with paper points from one implant per patient. In 15 patients, the suprastructure had been cemented with a zinc oxide-eugenol cement (Temp Bond, TB) and in 23 patients with a methacrylate cement (Premier Implant Cement, PIC). The excess cement found as well as suppuration was documented. Subgingival samples of all patients were analyzed for taxonomic composition by means of 16S amplicon sequencing. RESULTS: None of the TB-cemented implants had excess cement or suppuration. In 14 (61%) of the PIC, excess cement was found. Suppuration was detected in 33% of the PIC implants without excess cement and in 100% of the PIC implants with excess cement. The taxonomic analysis of the microbial samples revealed an accumulation of oral pathogens in the PIC patients independent of the presence of excess cement. Significantly fewer oral pathogens occurred in patients with TB compared to patients with PIC. CONCLUSION: Compared with TB, PIC favors the development of suppuration and the growth of periodontal pathogens.


Subject(s)
Dental Cements/chemistry , Dental Prosthesis, Implant-Supported , Peri-Implantitis/microbiology , Adult , Aged , Female , Humans , Male , Methacrylates , Middle Aged , Young Adult , Zinc Oxide-Eugenol Cement
11.
J Bacteriol ; 197(6): 1026-39, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25384477

ABSTRACT

The PA4203 gene encodes a LysR regulator and lies between the ppgL gene (PA4204), which encodes a periplasmic gluconolactonase, and, in the opposite orientation, the PA4202 (nmoA) gene, coding for a nitronate monooxygenase, and ddlA (PA4201), encoding a d-alanine alanine ligase. The intergenic regions between PA4203 and ppgL and between PA4203 and nmoA are very short (79 and 107 nucleotides, respectively). Here we show that PA4203 (nmoR) represses its own transcription and the expression of nmoA. A chromatin immunoprecipitation analysis showed the presence of a single NmoR binding site between nmoA and nmoR, which was confirmed by electrophoretic mobility shift assays (EMSAs) with the purified NmoR protein. Despite this observation, a transcriptome analysis revealed more genes to be affected in an nmoR mutant, including genes known to be part of the MexT LysR activator regulon. The PA1225 gene, encoding a quinone oxidoreductase, was the most highly upregulated gene in the nmoR deletion mutant, independently of MexT. Finally, deletion of the nmoA gene resulted in an increased sensitivity of the cells to 3-nitropropionic acid (3-NPA), confirming the role of the nitronate monooxygenase protein in the detoxification of nitronate.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Mixed Function Oxygenases/metabolism , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/genetics , Computer Simulation , DNA, Intergenic/genetics , Gene Deletion , Genome, Bacterial , Mixed Function Oxygenases/genetics , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pseudomonas aeruginosa/genetics
12.
PLoS Pathog ; 8(6): e1002760, 2012.
Article in English | MEDLINE | ID: mdl-22719254

ABSTRACT

The genetic adaptation of pathogens in host tissue plays a key role in the establishment of chronic infections. While whole genome sequencing has opened up the analysis of genetic changes occurring during long-term infections, the identification and characterization of adaptive traits is often obscured by a lack of knowledge of the underlying molecular processes. Our research addresses the role of Pseudomonas aeruginosa small colony variant (SCV) morphotypes in long-term infections. In the lungs of cystic fibrosis patients, the appearance of SCVs correlates with a prolonged persistence of infection and poor lung function. Formation of P. aeruginosa SCVs is linked to increased levels of the second messenger c-di-GMP. Our previous work identified the YfiBNR system as a key regulator of the SCV phenotype. The effector of this tripartite signaling module is the membrane bound diguanylate cyclase YfiN. Through a combination of genetic and biochemical analyses we first outline the mechanistic principles of YfiN regulation in detail. In particular, we identify a number of activating mutations in all three components of the Yfi regulatory system. YfiBNR is shown to function via tightly controlled competition between allosteric binding sites on the three Yfi proteins; a novel regulatory mechanism that is apparently widespread among periplasmic signaling systems in bacteria. We then show that during long-term lung infections of CF patients, activating mutations invade the population, driving SCV formation in vivo. The identification of mutational "scars" in the yfi genes of clinical isolates suggests that Yfi activity is both under positive and negative selection in vivo and that continuous adaptation of the c-di-GMP network contributes to the in vivo fitness of P. aeruginosa during chronic lung infections. These experiments uncover an important new principle of in vivo persistence, and identify the c-di-GMP network as a valid target for novel anti-infectives directed against chronic infections.


Subject(s)
Adaptation, Physiological/physiology , Bacterial Proteins/metabolism , Cystic Fibrosis/microbiology , Membrane Proteins/metabolism , Pseudomonas Infections/genetics , Pseudomonas aeruginosa , Signal Transduction/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cystic Fibrosis/complications , Humans , Immunoblotting , Immunoprecipitation , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutagenesis, Site-Directed , Mutation , Polymerase Chain Reaction , Protein Conformation , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/genetics , Respiratory Tract Infections/genetics , Respiratory Tract Infections/microbiology
13.
Appl Environ Microbiol ; 80(16): 4911-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24907323

ABSTRACT

Pseudomonas putida is a Gram-negative soil bacterium which is well-known for its versatile lifestyle, controlled by a large repertoire of transcriptional regulators. Besides one- and two-component regulatory systems, the genome of P. putida reveals 19 extracytoplasmic function (ECF) sigma factors involved in the adaptation to changing environmental conditions. In this study, we demonstrate that knockout of extracytoplasmic function sigma factor ECF-10, encoded by open reading frame PP4553, resulted in 2- to 4-fold increased antibiotic resistance to quinolone, ß-lactam, sulfonamide, and chloramphenicol antibiotics. In addition, the ECF-10 mutant exhibited enhanced formation of biofilms after 24 h of incubation. Transcriptome analysis using Illumina sequencing technology resulted in the detection of 12 genes differentially expressed (>2-fold) in the ECF-10 knockout mutant strain compared to their levels of expression in wild-type cells. Among the upregulated genes were ttgA, ttgB, and ttgC, which code for the major multidrug efflux pump TtgABC in P. putida KT2440. Investigation of an ECF-10 and ttgA double-knockout strain and a ttgABC-overexpressing strain demonstrated the involvement of efflux pump TtgABC in the stress resistance and biofilm formation phenotypes of the ECF-10 mutant strain, indicating a new role for this efflux pump beyond simple antibiotic resistance in P. putida KT2440.


Subject(s)
Bacterial Proteins/genetics , Biofilms , Pseudomonas putida/genetics , Sigma Factor/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial , Gene Knockout Techniques , Pseudomonas putida/drug effects , Pseudomonas putida/physiology , Sigma Factor/metabolism , Stress, Physiological
14.
Nucleic Acids Res ; 40(10): 4320-33, 2012 May.
Article in English | MEDLINE | ID: mdl-22275523

ABSTRACT

Most bacteria control oxidative stress through the H(2)O(2)-responsive transactivator OxyR, a member of the LTTR family (LysR Type Transcriptional Regulators), which activates the expression of defensive genes such as those encoding catalases, alkyl hydroperoxide reductases and superoxide dismutases. In the human opportunistic pathogen Pseudomonas aeruginosa, OxyR positively regulates expression of the oxidative stress response genes katA, katB, ahpB and ahpCF. To identify additional targets of OxyR in P. aeruginosa PAO1, we performed chromatin immunoprecipitation in combination with whole genome tiling array analyses (ChIP-chip). We detected 56 genes including all the previously identified defensive genes and a battery of novel direct targets of OxyR. Electrophoretic mobility shift assays (EMSAs) for selected newly identified targets indicated that ∼70% of those were bound by purified oxidized OxyR and their regulation was confirmed by quantitative real-time polymerase chain reaction. Furthermore, a thioredoxin system was identified to enzymatically reduce OxyR under oxidative stress. Functional classification analysis showed that OxyR controls a core regulon of oxidative stress defensive genes, and other genes involved in regulation of iron homeostasis (pvdS), quorum-sensing (rsaL), protein synthesis (rpsL) and oxidative phosphorylation (cyoA and snr1). Collectively, our results indicate that OxyR is involved in oxidative stress defense and regulates other aspects of cellular metabolism as well.


Subject(s)
Gene Expression Regulation, Bacterial , Oxidative Stress/genetics , Pseudomonas aeruginosa/genetics , Trans-Activators/metabolism , Binding Sites , Chromatin Immunoprecipitation , Genome, Bacterial , Oligonucleotide Array Sequence Analysis , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/metabolism , Regulon , Thioredoxins/metabolism
15.
Antimicrob Agents Chemother ; 57(3): 1361-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23274661

ABSTRACT

Quinolone antibiotics constitute a clinically successful and widely used class of broad-spectrum antibiotics; however, the emergence and spread of resistance increasingly limits the use of fluoroquinolones in the treatment and management of microbial disease. In this study, we evaluated the quantitative contributions of quinolone target alteration and efflux pump expression to fluoroquinolone resistance in Pseudomonas aeruginosa. We generated isogenic mutations in hot spots of the quinolone resistance-determining regions (QRDRs) of gyrA, gyrB, and parC and inactivated the efflux regulator genes so as to overexpress the corresponding multidrug resistance (MDR) efflux pumps. We then introduced the respective mutations into the reference strain PA14 singly and in various combinations. Whereas the combined inactivation of two efflux regulator-encoding genes did not lead to resistance levels higher than those obtained by inactivation of only one efflux regulator-encoding gene, the combination of mutations leading to increased efflux and target alteration clearly exhibited an additive effect. This combination of target alteration and overexpression of efflux pumps was commonly observed in clinical P. aeruginosa isolates; however, these two mechanisms were frequently found not to be sufficient to explain the level of fluoroquinolone resistance. Our results suggest that there are additional mechanisms, independent of the expression of the MexAB-OprM, MexCD-OprJ, MexEF-OprN, and/or MexXY-OprM efflux pump, that increase ciprofloxacin resistance in isolates with mutations in the QRDRs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Fluoroquinolones/pharmacology , Multidrug Resistance-Associated Proteins/deficiency , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Biological Transport , DNA Gyrase/genetics , DNA Gyrase/metabolism , DNA Topoisomerase IV/genetics , DNA Topoisomerase IV/metabolism , Drug Resistance, Bacterial/genetics , Gene Expression/drug effects , Microbial Sensitivity Tests , Multidrug Resistance-Associated Proteins/genetics , Mutation , Pseudomonas aeruginosa/metabolism
16.
Environ Microbiol ; 15(2): 597-609, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23278968

ABSTRACT

Pseudomonas aeruginosa pathogenicity and its capability to adapt to multiple environments are dependent on the production of diverse virulence factors, controlled by the sophisticated quorum sensing (QS) network of P. aeruginosa. To better understand the molecular mechanisms that underlie this adaptation we searched for novel key regulators of virulence factor production by screening a PA14 transposon mutant library for potential candidates acting downstream of the unique 2-alkyl-4-quinolone (AQ) QS system of P. aeruginosa. We focused the work on a protein named HemK with high homology to PrmC of Escherichia coli displaying a similar enzymatic activity (therefore also referred to as PrmC). In this study, we demonstrate that PrmC is an S-adenosyl-l-methionine (AdoMet)-dependent methyltransferase of peptide chain release factors (RFs) essential for the expression of several virulence factors, such as pyocyanin, rhamnolipids and the type III-secreted toxin ExoT. Furthermore, the PA14_prmC mutant strain is unable to grow under anoxic conditions and has a significantly reduced pathogenicity in the infection model Galleria mellonella. Along with transcriptomic and proteomic analyses, the presented data indicate that the methylation of RFs in P. aeruginosa seems to have a global effect on cellular processes related to the virulence of this nosocomial pathogen.


Subject(s)
Adaptation, Physiological/genetics , Methyltransferases/metabolism , Pseudomonas aeruginosa/enzymology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Larva/microbiology , Methyltransferases/genetics , Moths/microbiology , Mutation , Proteomics , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/pathogenicity , Quorum Sensing/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
17.
Carbohydr Polym ; 308: 120642, 2023 May 15.
Article in English | MEDLINE | ID: mdl-36813335

ABSTRACT

Many of the proposed health-related properties of pectins are based on their fermentability in the large intestine, but detailed structure-related studies on pectin fermentation have not been reported so far. Here, pectin fermentation kinetics were studied with a focus on structurally different pectic polymers. Therefore, six commercial pectins from citrus, apple, and sugar beet were chemically characterized and fermented in in vitro fermentation assays with human fecal samples over different periods of time (0 h, 4 h, 24 h, 48 h). Structure elucidation of intermediate cleavage products showed differences in fermentation speed and/or fermentation rate among the pectins, but the order in which specific structural pectic elements were fermented was comparable across all pectins. Neutral side chains of rhamnogalacturonan type I were fermented first (between 0 and 4 h), followed by homogalacturonan units (between 0 and 24 h) and, at last, the rhamnogalacturonan type I backbone (between 4 and 48 h). This indicates that fermentation of different pectic structural units might take place in different sections of the colon, potentially affecting their nutritional properties. For the formation of different short-chain fatty acids, mainly acetate, propionate, and butyrate, and the influence on microbiota, there was no time-dependent correlation regarding the pectic subunits. However, an increase of members of the bacterial genera Faecalibacterium, Lachnoclostridium, and Lachnospira was observed for all pectins.


Subject(s)
Pectins , Rhamnogalacturonans , Humans , Fermentation , Pectins/chemistry , Feces/microbiology , Bacteria/metabolism
18.
Sci Rep ; 13(1): 19340, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37935741

ABSTRACT

Purified diets (PD) increase standardization and repeatability in rodent studies but lead to differences in the phenotype of animals compared to grain-based "chow" diets. PD contain less fiber and are often devoid of soluble fiber, which can impact gut health. Thus, the aim of the present study was to modify the PD AIN93G by addition of soluble fiber, to promote more natural gut development as seen with chow diets. One hundred twenty male C57BL/6J mice were fed over 12 weeks either a chow diet, AIN93G or one of three modified AIN93G with increased fiber content and different ratios of soluble fiber to cellulose. Gut health was assessed through histological and immunohistochemical parameters and gut barrier gene expression. Gut microbiota composition was analyzed and its activity characterized through short chain fatty acid (SCFA) quantification. Feeding AIN93G led to tissue atrophy, a less diverse microbiota and a lower production of SCFA compared to chow diet. The addition of soluble fiber mitigated these effects, leading to intermediate colon and caecum crypt lengths and microbiota composition compared to both control diets. In conclusion, the addition of soluble fibers in PDs seems essential for gut morphology as well as a diverse and functional gut microbiome.


Subject(s)
Colon , Dietary Fiber , Mice , Male , Animals , Dietary Fiber/metabolism , Mice, Inbred C57BL , Colon/metabolism , Cecum/metabolism , Diet , Fatty Acids, Volatile/metabolism
19.
JMIR Res Protoc ; 12: e42529, 2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37027187

ABSTRACT

BACKGROUND: On the national level, nutritional monitoring requires the assessment of reliable representative dietary intake data. To achieve this, standardized tools need to be developed, validated, and kept up-to-date with recent developments in food products and the nutritional behavior of the population. Recently, the human intestinal microbiome has been identified as an essential mediator between nutrition and host health. Despite growing interest in this connection, only a few associations between the microbiome, nutrition, and health have been clearly established. Available studies paint an inconsistent picture, partly due to a lack of standardization. OBJECTIVE: First, we aim to verify if food consumption, as well as energy and nutrient intake of the German population, can be recorded validly by means of the dietary recall software GloboDiet, which will be applied in the German National Nutrition Monitoring. Second, we aim to obtain high-quality data using standard methods on the microbiome, combined with dietary intake data and additional fecal sample material, and to also assess the functional activity of the microbiome by measuring microbial metabolites. METHODS: Healthy female and male participants aged between 18 and 79 years were recruited. Anthropometric measurements included body height and weight, BMI, and bioelectrical impedance analysis. For validation of the GloboDiet software, current food consumption was assessed with a 24-hour recall. Nitrogen and potassium concentrations were measured from 24-hour urine collections to enable comparison with the intake of protein and potassium estimated by the GloboDiet software. Physical activity was measured over at least 24 hours using a wearable accelerometer to validate the estimated energy intake. Stool samples were collected in duplicate for a single time point and used for DNA isolation and subsequent amplification and sequencing of the 16S rRNA gene to determine microbiome composition. For the identification of associations between nutrition and the microbiome, the habitual diet was determined using a food frequency questionnaire covering 30 days. RESULTS: In total, 117 participants met the inclusion criteria. The study population was equally distributed between the sexes and 3 age groups (18-39, 40-59, and 60-79 years). Stool samples accompanying habitual diet data (30-day food frequency questionnaire) are available for 106 participants. Current diet data and 24-hour urine samples for the validation of GloboDiet are available for 109 participants, of which 82 cases also include physical activity data. CONCLUSIONS: We completed the recruitment and sample collection of the ErNst study with a high degree of standardization. Samples and data will be used to validate the GloboDiet software for the German National Nutrition Monitoring and to compare microbiome composition and nutritional patterns. TRIAL REGISTRATION: German Register of Clinical Studies DRKS00015216; https://drks.de/search/de/trial/DRKS00015216. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID): DERR1-10.2196/42529.

20.
Chembiochem ; 13(16): 2339-45, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23011873

ABSTRACT

The natural myxobacterial product argyrin is a cyclic peptide exhibiting immunosuppressive activity as well as antibacterial activity directed against the highly intrinsically resistant opportunistic pathogen Pseudomonas aeruginosa. In this study, we used whole-genome sequencing technology as a powerful tool to determine the mode of action of argyrin. Sequencing of argyrin-resistant P. aeruginosa isolates selected in vitro uncovered six point mutations that distinguished the resistant mutants from their susceptible parental strain. All six mutations were localized within one gene: fusA1, which encodes for the elongation factor EF-G. After the reintroduction of selected mutations into the susceptible wild type, the strain became resistant to argyrin. Surface plasmon resonance experiments confirmed the interaction of argyrin A with FusA1. Interestingly, EF-G has been previously shown to be the target of the anti-Staphylococcus antibiotic fusidic acid. Mapping of the mutations onto a structural model of EF-G revealed that the mutations conveying resistance against argyrin were clustered within domain III on the side opposite to that involved in fusidic acid binding, thus indicating that argyrin exhibits a new mode of protein synthesis inhibition. Although no mutations causing argyrin resistance have been found in other genes of P. aeruginosa, analysis of the sequence identity in EF-G and its correlation with argyrin resistance in different bacteria imply that additional factors such as uptake of argyrin play a role in the argyrin resistance of other organisms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Peptide Elongation Factor G/genetics , Peptides, Cyclic/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Mutation , Peptides, Cyclic/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL