Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 855
Filter
1.
Int J Mol Sci ; 25(5)2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38474038

ABSTRACT

Biofilm-associated bacterial infections are the major reason for treatment failure in many diseases including burn trauma infections. Uncontrolled inflammation induced by bacteria leads to materiality, tissue damage, and chronic diseases. Specialized proresolving mediators (SPMs), including maresin-like lipid mediators (MarLs), are enzymatically biosynthesized from omega-3 essential long-chain polyunsaturated fatty acids, especially docosahexaenoic acid (DHA), by macrophages and other leukocytes. SPMs exhibit strong inflammation-resolving activities, especially inflammation provoked by bacterial infection. In this study, we explored the potential direct inhibitory activities of three MarLs on Gram-positive (Staphylococcus aureus) and Gram-negative (Pseudomonas aeruginosa and Escherichia coli) bacteria in their biofilms that are leading bacteria in burn trauma-related infections. We also examined the effects of MarLs on the bactericidal activities of a typical broad-spectrum antibiotic, carbenicillin (carb), on these bacteria in their preformed biofilms. The results revealed that MarLs combined with carbenicillin can inhibit the survival of Gram-positive and Gram-negative bacteria in their biofilms although MarLs alone did not exhibit bactericidal activity. Thus, our findings suggest that the combination of MarLs and carbenicillin can lower the antibiotic requirements to kill the bacteria in preformed biofilms.


Subject(s)
Burns , Communicable Diseases , Staphylococcal Infections , Wound Infection , Humans , Anti-Bacterial Agents/pharmacology , Carbenicillin/pharmacology , Gram-Negative Bacteria , Gram-Positive Bacteria , Biofilms , Bacteria , Escherichia coli , Inflammation , Microbial Sensitivity Tests
2.
Molecules ; 26(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203552

ABSTRACT

Here, we investigated general porin regulation in Yersinia pseudotuberculosis 488, the causative agent of Far Eastern scarlet-like fever, in response to sublethal concentrations of antibiotics. We chose four antibiotics of different classes and measured gene expression using qRT-PCR and GFP reporter systems. Our data showed temporal regulation of the general porin genes ompF and ompC caused by antibiotic stress. The porin transcription initially decreased, providing early defensive response of the bacterium, while it returned to that of the untreated cells on prolonged antibiotic exposure. Unlike the major porin genes, the transcription of the alternative porin genes ompX and lamB was increased. Moreover, a short-term ompR- and marA-mediated porin regulation was observed. The main finding was a phenotypic heterogeneity of Y. pseudotuberculosis population manifested in variable porin gene expression under carbenicillin exposure. This may offer adaptive fitness advantages for a particular bacterial subpopulation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Carbenicillin/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Porins/biosynthesis , Stress, Physiological/drug effects , Yersinia pseudotuberculosis/metabolism
3.
Science ; 371(6531)2021 02 19.
Article in English | MEDLINE | ID: mdl-33602825

ABSTRACT

Although metabolism plays an active role in antibiotic lethality, antibiotic resistance is generally associated with drug target modification, enzymatic inactivation, and/or transport rather than metabolic processes. Evolution experiments of Escherichia coli rely on growth-dependent selection, which may provide a limited view of the antibiotic resistance landscape. We sequenced and analyzed E. coli adapted to representative antibiotics at increasingly heightened metabolic states. This revealed various underappreciated noncanonical genes, such as those related to central carbon and energy metabolism, which are implicated in antibiotic resistance. These metabolic alterations lead to lower basal respiration, which prevents antibiotic-mediated induction of tricarboxylic acid cycle activity, thus avoiding metabolic toxicity and minimizing drug lethality. Several of the identified metabolism-specific mutations are overrepresented in the genomes of >3500 clinical E. coli pathogens, indicating clinical relevance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Genes, Bacterial , Mutation , Adaptation, Physiological , Carbenicillin/pharmacology , Ciprofloxacin/pharmacology , Citric Acid Cycle/genetics , Directed Molecular Evolution , Energy Metabolism/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Gene Knockdown Techniques , Genome, Bacterial , Ketoglutarate Dehydrogenase Complex/genetics , Microbial Sensitivity Tests , Sequence Analysis, DNA , Streptomycin/pharmacology
4.
Microbiology (Reading) ; 166(11): 1095-1106, 2020 11.
Article in English | MEDLINE | ID: mdl-32909933

ABSTRACT

Antibiotic resistance in Pseudomonas aeruginosa is a serious concern in healthcare systems. Among the determinants of antibiotic resistance in P. aeruginosa, efflux pumps belonging to the resistance-nodulation-division (RND) family confer resistance to a broad range of antibacterial compounds. The MexXY efflux system is widely overexpressed in P. aeruginosa isolates from cystic fibrosis (CF) patients. MexXY can form functional complexes with two different outer membrane factors (OMFs), OprA and OprM. In this study, using state-of-the-art genetic tools, the substrate specificities of MexXY-OprA and MexXY-OprM complexes were determined. Our results show, for the first time, that the substrate profile of the MexXY system from P. aeruginosa PA7 can vary depending on which OM factor (OprM or OprA) it complexes with. While both MexXY-OprA and MexXY-OprM complexes are capable of effluxing aminoglycosides, the bi-anionic ß-lactam molecules carbenicillin and sulbenicillin were found to only be the substrate of MexXY-OprA. Our study therefore shows that by partnering with different OMF proteins MexY can expand its substrate profile.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Carbenicillin/metabolism , Drug Resistance, Multiple, Bacterial , Membrane Transport Proteins/metabolism , Pseudomonas aeruginosa/physiology , Sulbenicillin/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Carbenicillin/pharmacology , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Multiprotein Complexes , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Substrate Specificity , Sulbenicillin/pharmacology , beta-Lactams/metabolism , beta-Lactams/pharmacology
5.
Microb Genom ; 6(9)2020 09.
Article in English | MEDLINE | ID: mdl-32213259

ABSTRACT

Acinetobacter baumannii (Aba) is an emerging opportunistic pathogen associated to nosocomial infections. The rapid increase in multidrug resistance (MDR) among Aba strains underscores the urgency of understanding how this pathogen evolves in the clinical environment. We conducted here a whole-genome sequence comparative analysis of three phylogenetically and epidemiologically related MDR Aba strains from Argentinean hospitals, assigned to the CC104O/CC15P clonal complex. While the Ab244 strain was carbapenem-susceptible, Ab242 and Ab825, isolated after the introduction of carbapenem therapy, displayed resistance to these last resource ß-lactams. We found a high chromosomal synteny among the three strains, but significant differences at their accessory genomes. Most importantly, carbapenem resistance in Ab242 and Ab825 was attributed to the acquisition of a Rep_3 family plasmid carrying a blaOXA-58 gene. Other differences involved a genomic island carrying resistance to toxic compounds and a Tn10 element exclusive to Ab244 and Ab825, respectively. Also remarkably, 44 insertion sequences (ISs) were uncovered in Ab825, in contrast with the 14 and 11 detected in Ab242 and Ab244, respectively. Moreover, Ab825 showed a higher killing capacity as compared to the other two strains in the Galleria mellonella infection model. A search for virulence and persistence determinants indicated the loss or IS-mediated interruption of genes encoding many surface-exposed macromolecules in Ab825, suggesting that these events are responsible for its higher relative virulence. The comparative genomic analyses of the CC104O/CC15P strains conducted here revealed the contribution of acquired mobile genetic elements such as ISs and plasmids to the adaptation of A. baumannii to the clinical setting.


Subject(s)
Acinetobacter Infections/microbiology , Acinetobacter baumannii/classification , Drug Resistance, Bacterial , Plasmids/genetics , Whole Genome Sequencing/methods , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Adaptation, Physiological , Aminoglycosides/pharmacology , Animals , Argentina , Base Composition , Carbenicillin/pharmacology , DNA Transposable Elements , Disease Models, Animal , Genomics , Humans , Phylogeny , Synteny
6.
Chembiochem ; 21(6): 825-835, 2020 03 16.
Article in English | MEDLINE | ID: mdl-31553819

ABSTRACT

Antibiotics are known to promote bacterial formation of enhanced biofilms, the mechanism of which is not well understood. Here, using biolayer interferometry, we have shown that bacterial cultures containing antibiotics that target cell walls cause biomass deposition on surfaces over time with a linear profile rather than the Langmuir-like profiles exhibited by bacterial adherence in the absence of antibiotics. We observed about three times the initial rate and 12 times the final biomass deposition on surfaces for cultures containing carbenicillin than without. Unexpectedly, in the presence of antibiotics, the rate of biomass deposition inversely correlated with bacterial densities from different stages of a culture. Detailed studies revealed that carbenicillin caused faster growth of filaments that were seeded on surfaces from young bacteria (from lag phase) than those from high-density fast-growing bacteria, with rates of filament elongation of about 0.58 and 0.13 µm min-1 , respectively. With surfaces that do not support bacterial adherence, few filaments were observed even in solution. These filaments aggregated in solution and formed increased amounts of biofilms on surfaces. These results reveal the lifestyle of antibiotic-induced filamentous bacteria, as well as one way in which the antibiotics promote biofilm formation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Carbenicillin/pharmacology , Cell Wall/drug effects , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Bacterial Adhesion/drug effects , Escherichia coli/cytology , Microbial Sensitivity Tests , Pseudomonas aeruginosa/cytology , Surface Properties
7.
Commun Biol ; 2: 259, 2019.
Article in English | MEDLINE | ID: mdl-31312728

ABSTRACT

Antibiotic killing does not occur at a single, precise time for all cells within a population. Variability in time to death can be caused by stochastic expression of genes, resulting in differences in endogenous stress-resistance levels between individual cells in a population. Here we investigate whether single-cell differences in gene expression prior to antibiotic exposure are related to cell survival times after antibiotic exposure for a range of genes of diverse function. We quantified the time to death of single cells under antibiotic exposure in combination with expression of reporters. For some reporters, including genes involved in stress response and cellular processes like metabolism, the time to cell death had a strong relationship with the initial expression level of the genes. Our results highlight the single-cell level non-uniformity of antibiotic killing and also provide examples of key genes where cell-to-cell variation in expression is strongly linked to extended durations of antibiotic survival.


Subject(s)
Anti-Bacterial Agents/pharmacology , Computational Biology , Escherichia coli Infections/drug therapy , Systems Biology , AraC Transcription Factor/metabolism , Carbenicillin/pharmacology , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Image Processing, Computer-Assisted , Promoter Regions, Genetic , Stochastic Processes
8.
Microbiol Res ; 226: 55-64, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31284945

ABSTRACT

Functional association between genomic loci and specific biological traits remains lacking in many fungi, including the African tree pathogen Ceratocystis albifundus. This is mainly because of the absence of suitable transformation systems for allowing genetic manipulation of this and other fungi. Here, we present an optimized protocol for Agrobacterium tumefaciens-mediated transformation of C. albifundus. Strain AGL-1 of A. tumefaciens and four binary T-DNA vectors (conferring hygromycin B or geneticin resistance and/or expressing the green fluorescent protein [GFP]) were used for transforming germinated conidia of three isolates of C. albifundus. Stable expression of these T-DNA-encoded traits was confirmed through sequential sub-culturing of fungal transformants on selective and non-selective media and by using PCR and sequence analysis. Single-copy integration of the respective T-DNAs into the genomes of these fungi was confirmed using Southern hybridization analysis. The range of experimental parameters determined and optimised included: (i) concentrations of hygromycin B and geneticin required for inhibiting growth of the wild type fungus and (ii) the dependence of transformation on acetosyringone for inducing the bacterium's virulence genes, as well as (iii) the duration of fungus-bacterium co-cultivation periods and (iv) the concentrations of fungal conidia and bacterial cells used for the latter. The system developed in this study is stable with a high-efficiency, yielding up to 400 transformants per 106 conidia. This is the first report of a transformation protocol for C. albifundus and its availability will be invaluable for functional studies in this important fungus.


Subject(s)
Agrobacterium tumefaciens/genetics , Ascomycota/genetics , Transformation, Genetic , Ascomycota/cytology , Ascomycota/drug effects , Ascomycota/growth & development , Blotting, Southern , Carbenicillin/pharmacology , Coculture Techniques , DNA, Bacterial , Gene Expression Regulation, Fungal , Gentamicins/pharmacology , Green Fluorescent Proteins/genetics , Hygromycin B/pharmacology , Kanamycin/pharmacology , Polymerase Chain Reaction , Sequence Analysis , Virulence/genetics
9.
Mar Drugs ; 17(5)2019 May 24.
Article in English | MEDLINE | ID: mdl-31137680

ABSTRACT

Pseudomonas aeruginosa biofilms are typically associated with the chronic lung infection of cystic fibrosis (CF) patients and represent a major challenge for treatment. This opportunistic bacterial pathogen secretes alginate, a polysaccharide that is one of the main components of its biofilm. Targeting this major biofilm component has emerged as a tempting therapeutic strategy for tackling biofilm-associated bacterial infections. The enormous potential in genetic diversity of the marine microbial community make it a valuable resource for mining activities responsible for a broad range of metabolic processes, including the alginolytic activity responsible for degrading alginate. A collection of 36 bacterial isolates were purified from marine water based on their alginolytic activity. These isolates were identified based on their 16S rRNA gene sequences. Pseudoalteromonas sp. 1400 showed the highest alginolytic activity and was further confirmed to produce the enzyme alginate lyase. The purified alginate lyase (AlyP1400) produced by Pseudoalteromonas sp. 1400 showed a band of 23 KDa on a protein electrophoresis gel and exhibited a bifunctional lyase activity for both poly-mannuronic acid and poly-glucuronic acid degradation. A tryptic digestion of this gel band analyzed by liquid chromatography-tandem mass spectrometry confirmed high similarity to the alginate lyases in polysaccharide lyase family 18. The purified alginate lyase showed a maximum relative activity at 30 °C at a slightly acidic condition. It decreased the sodium alginate viscosity by over 90% and reduced the P. aeruginosa (strain PA14) biofilms by 69% after 24 h of incubation. The combined activity of AlyP1400 with carbenicillin or ciprofloxacin reduced the P. aeruginosa biofilm thickness, biovolume and surface area in a flow cell system. The present data revealed that AlyP1400 combined with conventional antibiotics helped to disrupt the biofilms produced by P. aeruginosa and can be used as a promising combinational therapeutic strategy.


Subject(s)
Biofilms/drug effects , Polysaccharide-Lyases/pharmacology , Pseudoalteromonas/enzymology , Pseudomonas aeruginosa/drug effects , Alginates/metabolism , Anti-Bacterial Agents/pharmacology , Aquatic Organisms/enzymology , Aquatic Organisms/genetics , Carbenicillin/pharmacology , Ciprofloxacin/pharmacology , Polysaccharide-Lyases/genetics , Polysaccharide-Lyases/metabolism , Pseudoalteromonas/genetics , Pseudomonas aeruginosa/physiology , RNA, Ribosomal, 16S/genetics
10.
Appl Environ Microbiol ; 85(14)2019 07 15.
Article in English | MEDLINE | ID: mdl-31076438

ABSTRACT

Pseudomonas aeruginosa is a bacterial pathogen that causes severe chronic infections in immunocompromised individuals. This bacterium is highly adaptable to its environments, which frequently select for traits that promote bacterial persistence. A clinically significant temporal adaptation is the formation of surface- or cell-adhered bacterial biofilms that are associated with increased resistance to immune and antibiotic clearance. Extensive research has shown that bacterial flagellar motility promotes formation of such biofilms, whereupon the bacteria subsequently become nonmotile. However, recent evidence shows that antibiotic-tolerant nonattached bacterial aggregates, distinct from surface-adhered biofilms, can form, and these have been reported in the context of lung infections, otitis media, nonhealing wounds, and soft tissue fillers. It is unclear whether the same bacterial traits are required for aggregate formation as for biofilm formation. In this report, using isogenic mutants, we demonstrate that P. aeruginosa aggregates in liquid cultures are spontaneously formed independent of bacterial flagellar motility and independent of an exogenous scaffold. This contrasts with the role of the flagellum to initiate surface-adhered biofilms. Similarly to surface-attached biofilms, these aggregates exhibit increased antibiotic tolerance compared to planktonic cultures. These findings provide key insights into the requirements for aggregate formation that contrast with those for biofilm formation and that may have relevance for the persistence and dissemination of nonmotile bacteria found within chronic clinical infections.IMPORTANCE In this work, we have investigated the role of bacterial motility with regard to antibiotic-tolerant bacterial aggregate formation. Previous work has convincingly demonstrated that P. aeruginosa flagellar motility promotes the formation of surface-adhered biofilms in many systems. In contrast, aggregate formation by P. aeruginosa was observed for nonmotile but not for motile cells in the presence of an exogenous scaffold. Here, we demonstrate that both wild-type P. aeruginosa and mutants that genetically lack motility spontaneously form antibiotic-tolerant aggregates in the absence of an exogenously added scaffold. Additionally, we also demonstrate that wild-type (WT) and nonmotile P. aeruginosa bacteria can coaggregate, shedding light on potential physiological interactions and heterogeneity of aggregates.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbenicillin/pharmacology , Drug Resistance, Bacterial/physiology , Gentamicins/pharmacology , Pseudomonas aeruginosa/physiology , Biofilms , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects
11.
Curr Genet ; 65(1): 213-222, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29951698

ABSTRACT

The Pseudomonas aeruginosa RsaL is a negative regulator of the quorum sensing signal synthesis gene lasI. The expression of RsaL is directly activated by the LasI cognate regulator LasR. Thus, RsaL and LasI-LasR (LasI/R) form a regulatory loop. Further studies revealed that RsaL is a global regulator which controls the expression of numerous genes through quorum sensing system dependent and independent pathways. However, whether RsaL is involved in antibiotic tolerance remains elusive. In this study, we found that the mutation of rsaL increased bacterial tolerance to ciprofloxacin and carbenicillin. Through motif search, gene expression analyses and electrophoretic mobility shift assays, we found that RsaL directly represses the expression of the narK1K2GHJI operon, which is involved in the tolerance to ciprofloxacin. We further demonstrated that the narK1K2GHJI operon is directly regulated by LasR. In combination, our study revealed a novel operon under the control of the RsaL, LasI/R regulatory loop.


Subject(s)
Bacterial Proteins/genetics , Carbenicillin/pharmacology , Ciprofloxacin/pharmacology , Drug Tolerance/genetics , Pseudomonas aeruginosa/drug effects , Repressor Proteins/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Mutation , Operon/genetics , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Quorum Sensing/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
12.
J Biomater Appl ; 33(4): 599-605, 2018 10.
Article in English | MEDLINE | ID: mdl-30326805

ABSTRACT

Biofilm formation has been implicated as a cause of post-tympanostomy tube otorrhea in patients suffering from otitis media with effusion, and biofilms have been found to adhere to all available types of tympanostomy tubes (TT) made from silicone. In this study, we present a novel stent designed with a reduced surface area and a titanium dioxide (TiO2) coating to prevent biofilm formation. Using a radio frequency power supply, tympanostomy stents (TS) made from Nitinol (Nikel-titanium) were coated with TiO2 to form an oxide layer on the metallic target. We successfully reproduced biofilms with carbenicillin-resistant Pseudomonas aeruginosa strain, PAO1-GFP (green fluorescent protein) on the tubes in vitro. We then compared the levels of biofilm formation by this strain on the two types of implants using several methods, including bacterial quantification, electron microscopy, and confocal laser fluorescent microscopy. Our results provide definitive evidence that the combination of the TiO2 coating and minimized surface area of the Nitinol stent inhibited the P. aeruginosa biofilm formation. The ability of the TS to prevent viable bacteria colonization (over 10 folds, compared to silicone TT) was verified by anti-biofilm test. Future studies will reveal more useful in reducing otorrhea and plugging complications as a novel tympanostomy tube.


Subject(s)
Alloys/chemistry , Biofilms , Coated Materials, Biocompatible/chemistry , Middle Ear Ventilation/instrumentation , Pseudomonas aeruginosa/physiology , Stents/microbiology , Titanium/chemistry , Bacterial Adhesion , Carbenicillin/pharmacology , Drug Resistance, Bacterial , Equipment Design , Humans , Silicones/chemistry
13.
Plant Cell Physiol ; 59(10): 2086-2098, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-29986082

ABSTRACT

The presence of antibiotics in soils could be due to natural production by soil microorganisms or to the effect of anthropogenic activities. However, the impact of these compounds on plant physiology has not been thoroughly investigated. To evaluate the effect of ß-lactam antibiotics (carbenicillin and penicillin) on the growth and development of Arabidopsis thaliana roots, plants were grown in the presence of different amounts and we found a reduction in root size, an increase in the size of root hairs as well as an abnormal position closer to the tip of the roots. Those phenomena were dependent on the accumulation of both antibiotics inside root tissues and also correlated with a decrease in size of the root apical meristem not related to an alteration in cell division but to a decrease in cell expansion. Using an RNA sequencing analysis, we detected an increase in the expression of genes related to the response to oxidative stress, which would explain the increase in the levels of endogenous reactive oxygen species found in the presence of those antibiotics. Moreover, some auxin-responsive genes were misregulated, especially an induction of CYP79B3, possibly explaining the increase in auxin levels in the presence of carbenicillin and the decrease in the amount of indole glucosinolates, involved in the control of fungal infections. Accordingly, penicillin-treated plants were hypersensitive to the endophyte fungus Colletotrichum tofieldiae. These results underscore the risks for plant growth of ß-lactam antibiotics in agricultural soils, and suggest a possible function for these compounds as fungus-produced signaling molecules to modify plant behavior.


Subject(s)
Anti-Bacterial Agents/pharmacology , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Glucosinolates/metabolism , Arabidopsis/drug effects , Carbenicillin/pharmacology , Gene Expression Regulation, Plant/physiology , Penicillins/pharmacology
14.
Proc Natl Acad Sci U S A ; 115(16): 4069-4074, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610312

ABSTRACT

It is widely acknowledged that faster-growing bacteria are killed faster by ß-lactam antibiotics. This notion serves as the foundation for the concept of bacterial persistence: dormant bacterial cells that do not grow are phenotypically tolerant against ß-lactam treatment. Such correlation has often been invoked in the mathematical modeling of bacterial responses to antibiotics. Due to the lack of thorough quantification, however, it is unclear whether and to what extent the bacterial growth rate can predict the lysis rate upon ß-lactam treatment under diverse conditions. Enabled by experimental automation, here we measured >1,000 growth/killing curves for eight combinations of antibiotics and bacterial species and strains, including clinical isolates of bacterial pathogens. We found that the lysis rate of a bacterial population linearly depends on the instantaneous growth rate of the population, regardless of how the latter is modulated. We further demonstrate that this predictive power at the population level can be explained by accounting for bacterial responses to the antibiotic treatment by single cells. This linear dependence of the lysis rate on the growth rate represents a dynamic signature associated with each bacterium-antibiotic pair and serves as the quantitative foundation for designing combination antibiotic therapy and predicting the population-structure change in a population with mixed phenotypes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriolysis/drug effects , Carbenicillin/pharmacology , Escherichia coli/drug effects , Bacterial Load , Biomass , Culture Media/pharmacology , Escherichia coli/growth & development , High-Throughput Screening Assays/instrumentation , Kinetics , Nephelometry and Turbidimetry , Robotics , Temperature
15.
Biochem Biophys Res Commun ; 494(1-2): 365-371, 2017 12 09.
Article in English | MEDLINE | ID: mdl-28988110

ABSTRACT

Isolated Agrobacterium tumefaciens was exposed to different extremely low frequencies of square amplitude modulated waves (QAMW) from two generators to determine the resonance frequency that causes growth inhibition. The carrier was 10 MHz sine wave with amplitude ±10 Vpp which was modulated by a second wave generator with a modulation depth of ± 2Vpp and constant field strength of 200 V/m at 28 °C. The exposure of A. tumefaciens to 1.0 Hz QAMW for 90 min inhibited the bacterial growth by 49.2%. In addition, the tested antibiotics became more effective against A. tumefaciens after the exposure. Furthermore, results of DNA, dielectric relaxation and TEM showed highly significant molecular and morphological changes due to the exposure to 1.0 Hz QAMW for 90 min. An in-vivo study has been carried out on healthy tomato plants to test the pathogenicity of A. tumefaciens before and after the exposure to QAMW at the inhibiting frequency. Symptoms of crown gall and all pathological symptoms were more aggressive in tomato plants treated with non-exposed bacteria, comparing with those treated with exposed bacteria. We concluded that, the exposure of A. tumefaciens to 1.0 Hz QAMW for 90 min modified its cellular activity and DNA structure, which inhibited the growth and affected the microbe pathogenicity.


Subject(s)
Agrobacterium tumefaciens/radiation effects , Anti-Bacterial Agents/pharmacology , DNA, Bacterial/radiation effects , Electromagnetic Radiation , Agrobacterium tumefaciens/drug effects , Agrobacterium tumefaciens/genetics , Agrobacterium tumefaciens/growth & development , Amikacin/pharmacology , Carbenicillin/pharmacology , Cefaclor/pharmacology , Chloramphenicol/pharmacology , Ciprofloxacin/pharmacology , DNA, Bacterial/drug effects , Fluoroquinolones/pharmacology , Gatifloxacin , Gentamicins/pharmacology , Solanum lycopersicum/microbiology , Plant Tumors/microbiology , Rifampin/pharmacology
16.
PLoS One ; 12(7): e0180925, 2017.
Article in English | MEDLINE | ID: mdl-28749949

ABSTRACT

There is an urgent need to develop new drug treatment strategies to control the global spread of drug-sensitive and multidrug-resistant Mycobacterium tuberculosis (M. tuberculosis). The ß-lactam class of antibiotics is among the safest and most widely prescribed antibiotics, but they are not effective against M. tuberculosis due to intrinsic resistance. This study shows that 2-aminoimidazole (2-AI)-based small molecules potentiate ß-lactam antibiotics against M. tuberculosis. Active 2-AI compounds significantly reduced the minimal inhibitory and bactericidal concentrations of ß-lactams by increasing M. tuberculosis cell envelope permeability and decreasing protein secretion including ß-lactamase. Metabolic labeling and transcriptional profiling experiments revealed that 2-AI compounds impair mycolic acid biosynthesis, export and linkage to the mycobacterial envelope, counteracting an important defense mechanism reducing permeability to external agents. Additionally, other important constituents of the M. tuberculosis outer membrane including sulfolipid-1 and polyacyltrehalose were also less abundant in 2-AI treated bacilli. As a consequence of 2-AI treatment, M. tuberculosis displayed increased sensitivity to SDS, increased permeability to nucleic acid staining dyes, and rapid binding of cell wall targeting antibiotics. Transcriptional profiling analysis further confirmed that 2-AI induces transcriptional regulators associated with cell envelope stress. 2-AI based small molecules potentiate the antimicrobial activity of ß-lactams by a mechanism that is distinct from specific inhibitors of ß-lactamase activity and therefore may have value as an adjunctive anti-TB treatment.


Subject(s)
Anti-Infective Agents/pharmacology , Cell Membrane Permeability/drug effects , Imidazoles/pharmacology , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/enzymology , beta-Lactamases/metabolism , beta-Lactams/pharmacology , Carbenicillin/pharmacology , Coloring Agents/chemistry , Lipids/analysis , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Nucleic Acids/metabolism , Penicillin V/pharmacology , Sodium Dodecyl Sulfate/pharmacology , Staining and Labeling , Transcription, Genetic/drug effects , Vancomycin/pharmacology
17.
BMC Microbiol ; 16: 82, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27159970

ABSTRACT

BACKGROUND: The emergence of antibiotic resistant pathogenic bacteria has reduced our ability to combat infectious diseases. At the same time the numbers of new antibiotics reaching the market have decreased. This situation has created an urgent need to discover novel antibiotic scaffolds. Recently, the application of pattern recognition techniques to identify molecular fingerprints in 'omics' studies, has emerged as an important tool in biomedical research and laboratory medicine to identify pathogens, to monitor therapeutic treatments or to develop drugs with improved metabolic stability, toxicological profile and efficacy. Here, we hypothesize that a combination of metabolic intracellular fingerprints and extracellular footprints would provide a more comprehensive picture about the mechanism of action of novel antibiotics in drug discovery programs. RESULTS: In an attempt to integrate the metabolomics approach as a classification tool in the drug discovery processes, we have used quantitative (1)H NMR spectroscopy to study the metabolic response of Escherichia coli cultures to different antibiotics. Within the frame of our study the effects of five different and well-known antibiotic classes on the bacterial metabolome were investigated both by intracellular fingerprint and extracellular footprint analysis. The metabolic fingerprints and footprints of bacterial cultures were affected in a distinct manner and provided complementary information regarding intracellular and extracellular targets such as protein synthesis, DNA and cell wall. While cell cultures affected by antibiotics that act on intracellular targets showed class-specific fingerprints, the metabolic footprints differed significantly only when antibiotics that target the cell wall were applied. In addition, using a training set of E. coli fingerprints extracted after treatment with different antibiotic classes, the mode of action of streptomycin, tetracycline and carbenicillin could be correctly predicted. CONCLUSION: The metabolic profiles of E. coli treated with antibiotics with intracellular and extracellular targets could be separated in fingerprint and footprint analysis, respectively and provided complementary information. Based on the specific fingerprints obtained for different classes of antibiotics, the mode of action of several antibiotics could be predicted. The same classification approach should be applicable to studies of other pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Metabolomics/methods , Proton Magnetic Resonance Spectroscopy/methods , Carbenicillin/pharmacology , Drug Discovery , Escherichia coli/classification , Microbial Sensitivity Tests , Multivariate Analysis , Pilot Projects , Streptomycin/pharmacology , Tetracycline/pharmacology
18.
FEMS Microbiol Lett ; 363(11)2016 06.
Article in English | MEDLINE | ID: mdl-27190289

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen, known to develop robust biofilms. Its biofilm development increases when antibiotics are presented at subminimal inhibitory concentrations (MICs) for reasons that remain unclear. In order to identify genes that affect biofilm development under such a sublethal antibiotic stress condition, we screened a transposon (Tn) mutant library of PAO1, a prototype P. aeruginosa strain. Among ∼5000 mutants, a fiuA gene mutant was verified to form very defective biofilms in the presence of sub-MIC carbenicillin. The fiuA gene encodes ferrichrome receptor A, involved in the iron acquisition process. Of note, biofilm formation was not decreased in the ΔpchΔpvd mutant defective in the production of pyochelin and pyoverdine, two well-characterized P. aeruginosa siderophore molecules. Moreover, ΔfiuA, a non-polar fiuA deletion mutant, produced a significantly decreased level of elastase, a major virulence determinant. Mouse airway infection experiments revealed that the mutant expressed significantly less pathogenicity. Our results suggest that the fiuA gene has pleiotropic functions that affect P. aeruginosa biofilm development and virulence. The targeting of FiuA could enable the attenuation of P. aeruginosa virulence and may be suitable for the development of a drug that specifically controls the virulence of this important pathogen.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Ferrichrome/metabolism , Pseudomonas aeruginosa/pathogenicity , Virulence Factors/genetics , Animals , Bacterial Outer Membrane Proteins/metabolism , Biofilms/drug effects , Biofilms/growth & development , Carbenicillin/pharmacology , DNA Transposable Elements , Gene Library , Iron/metabolism , Lung/microbiology , Mice , Microbial Sensitivity Tests , Oligopeptides/biosynthesis , Pancreatic Elastase/biosynthesis , Phenols/metabolism , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Sequence Deletion , Thiazoles/metabolism , Virulence Factors/metabolism
19.
Proc Natl Acad Sci U S A ; 113(6): 1648-53, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26831117

ABSTRACT

ß-Lactams disrupt bacterial cell wall synthesis, and these agents are the most widely used antibiotics. One of the principle mechanisms by which bacteria resist the action of ß-lactams is by producing ß-lactamases, enzymes that degrade ß-lactams. In Gram-negative bacteria, production of ß-lactamases is often induced in response to the antibiotic-associated damage to the cell wall. Here, we have identified a previously unidentified mechanism that governs ß-lactamase production. In the Gram-negative enteric pathogen Vibrio parahaemolyticus, we found a histidine kinase/response regulator pair (VbrK/VbrR) that controls expression of a ß-lactamase. Mutants lacking either VbrK or VbrR do not produce the ß-lactamase and are no longer resistant to ß-lactam antibiotics. Notably, VbrK autophosphorylation is activated by ß-lactam antibiotics, but not by other lactams. However, single amino acid substitutions in the putative periplasmic binding pocket of VbrK leads its phosphorylation in response to both ß-lactam and other lactams, suggesting that this kinase is a ß-lactam receptor that can directly detect ß-lactam antibiotics instead of detecting the damage to cell wall resulting from ß-lactams. In strong support of this idea, we found that purified periplasmic sensor domain of VbrK binds penicillin, and that such binding is critical for VbrK autophosphorylation and ß-lactamase production. Direct recognition of ß-lactam antibiotics by a histidine kinase receptor may represent an evolutionarily favorable mechanism to defend against ß-lactam antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial/drug effects , Protein Kinases/metabolism , beta-Lactams/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carbenicillin/pharmacology , Conserved Sequence , Gene Expression Regulation, Bacterial/drug effects , Histidine Kinase , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Sequence Data , Mutation/genetics , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary , Sequence Analysis, RNA , Substrate Specificity/drug effects , Transcription, Genetic/drug effects , Vibrio parahaemolyticus/drug effects , Vibrio parahaemolyticus/genetics , beta-Lactamases/metabolism
20.
Sci Rep ; 6: 19538, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26758525

ABSTRACT

Transient resistance can allow microorganisms to temporarily survive lethal concentrations of antibiotics. This can be accomplished through stochastic mechanisms, where individual cells within a population display diverse phenotypes to hedge against the appearance of an antibiotic. To date, research on transient stochastic resistance has focused primarily on mechanisms where a subpopulation of cells enters a dormant, drug-tolerant state. However, a fundamental question is whether stochastic gene expression can also generate variable resistance levels among growing cells in a population. We hypothesized that stochastic expression of antibiotic-inducible resistance mechanisms might play such a role. To investigate this, we focused on a prototypical example of such a system: the multiple antibiotic resistance activator MarA. Previous studies have shown that induction of MarA can lead to a multidrug resistant phenotype at the population level. We asked whether MarA expression also has a stochastic component, even when uninduced. Time lapse microscopy showed that isogenic cells express heterogeneous, dynamic levels of MarA, which were correlated with transient antibiotic survival. This finding has important clinical implications, as stochastic expression of resistance genes may be widespread, allowing populations to hedge against the sudden appearance of an antibiotic.


Subject(s)
DNA-Binding Proteins/genetics , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Anti-Bacterial Agents/pharmacology , Carbenicillin/pharmacology , Escherichia coli/growth & development , Gene Expression , Microbial Sensitivity Tests , Microbial Viability/drug effects , Microbial Viability/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...