Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Appl Microbiol ; 135(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39066496

ABSTRACT

AIMS: Staphylococcus aureus is an opportunistic pathogen whose treatment is further complicated by its ability to form biofilms. In this study, we examine the impact of growing S. aureus biofilms on different polymerizing surfaces, specifically agar and agarose, on the pathogen's tolerance to fluoroquinolones. METHODS AND RESULTS: Biofilms of two methicillin-resistant strains of S. aureus were grown on agar or agarose in the presence of the same added nutrients, and their antibiotic susceptibility to two fluoroquinolones, moxifloxacin (MXF) and delafloxacin (DLX), were measured. We also compared the metabolism and extracellular polymeric substances (EPS) production of biofilms that were grown on agar and agarose. CONCLUSIONS: Biofilms that were grown on agarose were consistently more susceptible to antibiotics than those grown on agar. We found that in biofilms that were grown on agar, extracellular protein composition was higher, and adding EPS to agarose-grown biofilms increased their tolerance to DLX to levels that were comparable to agar-grown biofilms.


Subject(s)
Agar , Anti-Bacterial Agents , Biofilms , Fluoroquinolones , Microbial Sensitivity Tests , Sepharose , Staphylococcus aureus , Biofilms/drug effects , Biofilms/growth & development , Fluoroquinolones/pharmacology , Anti-Bacterial Agents/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Staphylococcus aureus/growth & development , Culture Media/chemistry , Moxifloxacin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology
2.
Microbiol Spectr ; 12(1): e0356723, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38078717

ABSTRACT

IMPORTANCE: The bacterial pathogen Pseudomonas aeruginosa is responsible for a variety of chronic human infections. Even in the absence of identifiable resistance mutations, this pathogen can tolerate lethal antibiotic doses through phenotypic strategies like biofilm formation and metabolic quiescence. In this study, we determined that P. aeruginosa maintains greater metabolic activity in the stationary phase compared to the model organism, Escherichia coli, which has traditionally been used to study fluoroquinolone antibiotic tolerance. We demonstrate that hallmarks of E. coli fluoroquinolone tolerance are not conserved in P. aeruginosa, including the timing of cell death and necessity of the SOS DNA damage response for survival. The heightened sensitivity of stationary-phase P. aeruginosa to fluoroquinolones is attributed to maintained transcriptional and reductase activity. Our data suggest that perturbations that suppress transcription and respiration in P. aeruginosa may actually protect the pathogen against this important class of antibiotics.


Subject(s)
Levofloxacin , Pseudomonas Infections , Humans , Levofloxacin/pharmacology , Levofloxacin/metabolism , Pseudomonas aeruginosa/metabolism , Escherichia coli/genetics , Anti-Bacterial Agents/therapeutic use , Pseudomonas Infections/drug therapy , Fluoroquinolones/pharmacology , Microbial Sensitivity Tests
3.
Antimicrob Agents Chemother ; 67(4): e0163922, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36951560

ABSTRACT

Antibiotic tolerant bacteria and persistent cells that remain alive after a course of antibiotic treatment can foster the chronicity of infections and the development of antibiotic resistance. Elucidating how bacteria overcome antibiotic action and devising strategies to bolster a new drug's activity can allow us to preserve our antibiotic arsenal. Here, we investigate strategies to potentiate the activities of topoisomerase inhibitors against nongrowing Escherichia coli that are often recalcitrant to existing antibiotics. We focus on sensitizing bacteria to the fluoroquinolone (FQ) levofloxacin (Levo) and to the spiropyrimidinetrione zoliflodacin (Zoli)-the first antibiotic in its class of compounds in clinical development. We found that metabolic stimulation either alone or in combination with inhibiting the AcrAB-TolC efflux pump sensitized stationary-phase E. coli to Levo and Zoli. We demonstrate that the added metabolites increased proton motive force generation and ATP production in stationary-phase cultures without restarting growth. Instead, the stimulated bacteria increased transcription and translation, which rendered the populations more susceptible to topoisomerase inhibitors. Our findings illuminate potential vulnerabilities of antibiotic-tolerant bacteria that can be leveraged to sensitize them to new and existing classes of topoisomerase inhibitors. These approaches enable us to stay one step ahead of adaptive bacteria and safeguard the efficacy of our existing antibiotics.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Fluoroquinolones/pharmacology , Fluoroquinolones/metabolism , Topoisomerase Inhibitors/pharmacology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Bacteria
4.
Article in English | MEDLINE | ID: mdl-36845830

ABSTRACT

Antibiotic resistance is a major danger to public health that threatens to claim the lives of millions of people per year within the next few decades. Years of necessary administration and excessive application of antibiotics have selected for strains that are resistant to many of our currently available treatments. Due to the high costs and difficulty of developing new antibiotics, the emergence of resistant bacteria is outpacing the introduction of new drugs to fight them. To overcome this problem, many researchers are focusing on developing antibacterial therapeutic strategies that are "resistance-resistant"-regimens that slow or stall resistance development in the targeted pathogens. In this mini review, we outline major examples of novel resistance-resistant therapeutic strategies. We discuss the use of compounds that reduce mutagenesis and thereby decrease the likelihood of resistance emergence. Then, we examine the effectiveness of antibiotic cycling and evolutionary steering, in which a bacterial population is forced by one antibiotic toward susceptibility to another antibiotic. We also consider combination therapies that aim to sabotage defensive mechanisms and eliminate potentially resistant pathogens by combining two antibiotics or combining an antibiotic with other therapeutics, such as antibodies or phages. Finally, we highlight promising future directions in this field, including the potential of applying machine learning and personalized medicine to fight antibiotic resistance emergence and out-maneuver adaptive pathogens.

5.
J Appl Microbiol ; 132(6): 4020-4032, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35332984

ABSTRACT

AIMS: Bacterial persisters are rare phenotypic variants in clonal bacterial cultures that can endure antimicrobial therapy and potentially contribute to infection relapse. Here, we investigate the potential of leveraging microbial interactions to disrupt persisters as they resuscitate during the post-antibiotic treatment recovery period. METHODS AND RESULTS: We treated stationary-phase E. coli MG1655 with a DNA-damaging fluoroquinolone and co-cultured the cells with probiotic E. coli Nissle following antibiotic removal. We found that E. coli Nissle reduced the survival of fluoroquinolone persisters and their progeny by over three orders of magnitude within 24 h. Using a bespoke H-diffusion cell apparatus that we developed, we showed that E. coli Nissle antagonized the fluoroquinolone-treated cells in a contact-dependent manner. We further demonstrated that the fluoroquinolone-treated cells can still activate the SOS response as they recover from antibiotic treatment in the presence of E. coli Nissle and that the persisters depend on TolC-associated efflux systems to defend themselves against the action of E. coli Nissle. CONCLUSION: Our results demonstrate that probiotic bacteria, such as E. coli Nissle, have the potential to inhibit persisters as they resuscitate following antibiotic treatment. SIGNIFICANCE AND IMPACT OF THE STUDY: Bacterial persisters are thought to underlie chronic infections and they can lead to an increase in antibiotic-resistant mutants in their progenies. Our data suggest that we can leverage the knowledge we gain on the interactions between microbial strains/species that interfere with persister resuscitation, such as those involving probiotic E. coli Nissle and E. coli MG1655 (a K-12 strain), to bolster the activity of our existing antibiotics.


Subject(s)
Escherichia coli Proteins , Probiotics , Anti-Bacterial Agents/pharmacology , Escherichia coli , Fluoroquinolones/pharmacology , Probiotics/pharmacology
6.
Microorganisms ; 9(11)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34835403

ABSTRACT

Antibiotic persistence is a phenomenon in which rare cells of a clonal bacterial population can survive antibiotic doses that kill their kin, even though the entire population is genetically susceptible. With antibiotic treatment failure on the rise, there is growing interest in understanding the molecular mechanisms underlying bacterial phenotypic heterogeneity and antibiotic persistence. However, elucidating these rare cell states can be technically challenging. The advent of single-cell techniques has enabled us to observe and quantitatively investigate individual cells in complex, phenotypically heterogeneous populations. In this review, we will discuss current technologies for studying persister phenotypes, including fluorescent tags and biosensors used to elucidate cellular processes; advances in flow cytometry, mass spectrometry, Raman spectroscopy, and microfluidics that contribute high-throughput and high-content information; and next-generation sequencing for powerful insights into genetic and transcriptomic programs. We will further discuss existing knowledge gaps, cutting-edge technologies that can address them, and how advances in single-cell microbiology can potentially improve infectious disease treatment outcomes.

7.
Antimicrob Agents Chemother ; 65(8): e0028121, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34097492

ABSTRACT

Bacteria have a repertoire of strategies to overcome antibiotics in clinical use, complicating our ability to treat and cure infectious diseases. In addition to evolving resistance, bacteria within genetically clonal cultures can undergo transient phenotypic changes and tolerate high doses of antibiotics. These cells, termed persisters, exhibit heterogeneous phenotypes; the strategies that a bacterial population deploys to overcome one class of antibiotics can be distinct from those needed to survive treatment with drugs with another mode of action. It was previously reported that fluoroquinolones, which target DNA topoisomerases, retain the capacity to kill nongrowing bacteria that tolerate other classes of antibiotics. Here, we show that in Escherichia coli stationary-phase cultures and colony biofilms, persisters that survive treatment with the anionic fluoroquinolone delafloxacin depend on the AcrAB-TolC efflux pump. In contrast, we did not detect this dependence on AcrAB-TolC in E. coli persisters that survive treatment with three other fluoroquinolone compounds. We found that the loss of AcrAB-TolC activity via genetic mutations or chemical inhibition not only reduces delafloxacin persistence in nongrowing E. coli MG1655 or EDL933 (an E. coli O157:H7 strain), but it limits resistance development in progenies derived from delafloxacin persisters that were given the opportunity to recover in nutritive medium following antibiotic treatment. Our findings highlight the heterogeneity in defense mechanisms that persisters use to overcome different compounds within the same class of antibiotics. They further indicate that efflux pump inhibitors can potentiate the activity of delafloxacin against stationary-phase E. coli and block resistance development in delafloxacin persister progenies.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Anti-Bacterial Agents/pharmacology , Carrier Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fluoroquinolones/pharmacology
8.
J Bacteriol ; 203(14): e0001721, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33972352

ABSTRACT

Spores of firmicute species contain 100s of mRNAs, whose major function in Bacillus subtilis is to provide ribonucleotides for new RNA synthesis when spores germinate. To determine if this is a general phenomenon, RNA was isolated from spores of multiple firmicute species and relative mRNA levels determined by transcriptome sequencing (RNA-seq). Determination of RNA levels in single spores allowed calculation of RNA nucleotides/spore, and assuming mRNA is 3% of spore RNA indicated that only ∼6% of spore mRNAs were present at >1/spore. Bacillus subtilis, Bacillus atrophaeus, and Clostridioides difficile spores had 49, 42, and 51 mRNAs at >1/spore, and numbers of mRNAs at ≥1/spore were ∼10 to 50% higher in Geobacillus stearothermophilus and Bacillus thuringiensis Al Hakam spores and ∼4-fold higher in Bacillus megaterium spores. In all species, some to many abundant spore mRNAs (i) were transcribed by RNA polymerase with forespore-specific σ factors, (ii) encoded proteins that were homologs of those encoded by abundant B. subtilis spore mRNAs and are proteins in dormant spores, and (iii) were likely transcribed in the mother cell compartment of the sporulating cell. Analysis of the coverage of RNA-seq reads on mRNAs from all species suggested that abundant spore mRNAs were fragmented, as was confirmed by reverse transcriptase quantitative PCR (RT-qPCR) analysis of abundant B. subtilis and C. difficile spore mRNAs. These data add to evidence indicating that the function of at least the great majority of mRNAs in all firmicute spores is to be degraded to generate ribonucleotides for new RNA synthesis when spores germinate. IMPORTANCE Only ∼6% of mRNAs in spores of six firmicute species are at ≥1 molecule/spore, many abundant spore mRNAs encode proteins similar to B. subtilis spore proteins, and some abundant B. subtilis and C. difficile spore mRNAs were fragmented. Most of the abundant B. subtilis and other Bacillales spore mRNAs are transcribed under the control of the forespore-specific RNA polymerase σ factors, F or G, and these results may stimulate transcription analyses in developing spores of species other than B. subtilis. These findings, plus the absence of key nucleotide biosynthetic enzymes in spores, suggest that firmicute spores' abundant mRNAs are not translated when spores germinate but instead are degraded to generate ribonucleotides for new RNA synthesis by the germinated spore.


Subject(s)
Firmicutes/genetics , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Spores, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Firmicutes/enzymology , Firmicutes/metabolism , RNA, Bacterial/genetics , RNA, Messenger/genetics , Spores, Bacterial/metabolism
9.
Cell Signal ; 75: 109750, 2020 11.
Article in English | MEDLINE | ID: mdl-32846197

ABSTRACT

Antibiotics have vastly improved our quality of life since their discovery and introduction into modern medicine. Yet, widespread use and misuse have compromised the efficacy of these compounds and put our ability to cure infectious diseases in jeopardy. To defend themselves against antibiotics, bacteria have evolved an arsenal of survival strategies. In addition to acquiring mutations and genetic determinants that confer antibiotic resistance, bacteria can respond to environmental cues and adopt reversible phenotypic changes that transiently enhance their ability to survive adverse conditions, including those brought on by antibiotics. These antibiotic tolerant and persistent bacteria, which are prevalent in biofilms and can survive antimicrobial therapy without inheriting resistance, are thought to underlie treatment failure and infection relapse. At infection sites, bacteria encounter a range of signals originating from host immunity and the local microbiota that can induce transcriptomic and metabolic reprogramming. In this review, we will focus on the impact of host factors and microbial interactions on antibiotic tolerance and persistence. We will also outline current efforts in leveraging the knowledge of host-microbe and microbe-microbe interactions in designing therapies that potentiate antibiotic activity and reduce the burden caused by recurrent infections.


Subject(s)
Bacteria , Drug Resistance, Bacterial , Host Microbial Interactions , Social Networking , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/immunology , Humans
10.
Appl Environ Microbiol ; 86(8)2020 04 01.
Article in English | MEDLINE | ID: mdl-32033948

ABSTRACT

This study examined the microbicidal activity of 222-nm UV radiation (UV222), which is potentially a safer alternative to the 254-nm UV radiation (UV254) that is often used for surface decontamination. Spores and/or growing and stationary-phase cells of Bacillus cereus, Bacillus subtilis, Bacillus thuringiensis, Staphylococcus aureus, and Clostridioides difficile and a herpesvirus were all killed or inactivated by UV222 and at lower fluences than with UV254B. subtilis spores and cells lacking the major DNA repair protein RecA were more sensitive to UV222, as were spores lacking their DNA-protective proteins, the α/ß-type small, acid-soluble spore proteins. The spore cores' large amount of Ca2+-dipicolinic acid (∼25% of the core dry weight) also protected B. subtilis and C. difficile spores against UV222, while spores' proteinaceous coat may have given some slight protection against UV222 Survivors among B. subtilis spores treated with UV222 acquired a large number of mutations, and this radiation generated known mutagenic photoproducts in spore and cell DNA, primarily cyclobutane-type pyrimidine dimers in growing cells and an α-thyminyl-thymine adduct termed the spore photoproduct (SP) in spores. Notably, the loss of a key SP repair protein markedly decreased spore UV222 resistance. UV222-treated B. subtilis spores germinated relatively normally, and the generation of colonies from these germinated spores was not salt sensitive. The latter two findings suggest that UV222 does not kill spores by general protein damage, and thus, the new results are consistent with the notion that DNA damage is responsible for the killing of spores and cells by UV222IMPORTANCE Spores of a variety of bacteria are resistant to common decontamination agents, and many of them are major causes of food spoilage and some serious human diseases, including anthrax caused by spores of Bacillus anthracis Consequently, there is an ongoing need for efficient methods for spore eradication, in particular methods that have minimal deleterious effects on people or the environment. UV radiation at 254 nm (UV254) is sporicidal and commonly used for surface decontamination but can cause deleterious effects in humans. Recent work, however, suggests that 222-nm UV (UV222) may be less harmful to people than UV254 yet may still kill bacteria and at lower fluences than UV254 The present work has identified the damage by UV222 that leads to the killing of growing cells and spores of some bacteria, many of which are human pathogens, and UV222 also inactivates a herpesvirus.


Subject(s)
Bacillus/radiation effects , Clostridioides difficile/radiation effects , DNA Damage , Simplexvirus/radiation effects , Spores, Bacterial/radiation effects , Staphylococcus aureus/radiation effects , Bacillus/physiology , Clostridioides difficile/physiology , Simplexvirus/physiology , Spores, Bacterial/physiology , Staphylococcus aureus/physiology , Ultraviolet Rays/adverse effects
11.
Nat Commun ; 10(1): 1177, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862812

ABSTRACT

Bacterial persisters are able to tolerate high levels of antibiotics and give rise to new populations. Persister tolerance is generally attributed to minimally active cellular processes that prevent antibiotic-induced damage, which has led to the supposition that persister offspring give rise to antibiotic-resistant mutants at comparable rates to normal cells. Using time-lapse microscopy to monitor Escherichia coli populations following ofloxacin treatment, we find that persisters filament extensively and induce impressive SOS responses before returning to a normal appearance. Further, populations derived from fluoroquinolone persisters contain significantly greater quantities of antibiotic-resistant mutants than those from untreated controls. We confirm that resistance is heritable and that the enhancement requires RecA, SOS induction, an opportunity to recover from treatment, and the involvement of error-prone DNA polymerase V (UmuDC). These findings show that fluoroquinolones damage DNA in persisters and that the ensuing SOS response accelerates the development of antibiotic resistance from these survivors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/physiology , Ofloxacin/pharmacology , SOS Response, Genetics/physiology , DNA Damage/drug effects , DNA-Directed DNA Polymerase/metabolism , Escherichia coli Proteins/metabolism , Microscopy, Fluorescence , Rec A Recombinases/metabolism , SOS Response, Genetics/drug effects , Time-Lapse Imaging
12.
Proc Natl Acad Sci U S A ; 115(27): E6301-E6309, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29915065

ABSTRACT

Bacterial persisters are subpopulations of phenotypic variants in isogenic cultures that can survive lethal doses of antibiotics. Their tolerances are often attributed to reduced activities of antibiotic targets, which limit corruption and damage in persisters compared with bacteria that die from treatment. However, that model does not hold for nongrowing populations treated with ofloxacin, a fluoroquinolone, where antibiotic-induced damage is comparable between cells that live and those that die. To understand how those persisters achieve this feat, we employed a genetic system that uses orthogonal control of MazF and MazE, a toxin and its cognate antitoxin, to generate model persisters that are uniformly tolerant to ofloxacin. Despite this complete tolerance, MazF model persisters required the same DNA repair machinery (RecA, RecB, and SOS induction) to survive ofloxacin treatment as their nongrowing, WT counterparts and exhibited similar indicators of DNA damage from treatment. Further investigation revealed that, following treatment, the timing of DNA repair was critical to MazF persister survival because, when repair was delayed until after growth and DNA synthesis resumed, survival was compromised. In addition, we found that, with nongrowing, WT planktonic and biofilm populations, stalling the resumption of growth and DNA synthesis after the conclusion of fluoroquinolone treatment with a prevalent type of stress at infection sites (nutrient limitation) led to near complete survival. These findings illustrate that the timing of events, such as DNA repair, following fluoroquinolone treatment is important to persister survival and provide further evidence that knowledge of the postantibiotic recovery period is critical to understanding persistence phenotypes.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Microbial Viability/drug effects , Ofloxacin/pharmacology , DNA-Binding Proteins/genetics , Endoribonucleases/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics
13.
Science ; 356(6335): 247-248, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28428383
14.
Mol Cell ; 65(2): 205-206, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28107646

ABSTRACT

In this issue, Nelson and colleagues (2017) determined that guanidine, the prevalent protein denaturant, is the long-lost ligand sensed by the ykkC class of riboswitches, and identified that members of its regulon are involved in guanidine detoxification and export.


Subject(s)
Guanidine , Riboswitch , Bacteria/genetics , Guanidines , RNA, Bacterial
15.
mBio ; 6(6): e01588-15, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578677

ABSTRACT

UNLABELLED: Metabolism plays an important role in the persister phenotype, as evidenced by the number of strategies that perturb metabolism to sabotage this troublesome subpopulation. However, the absence of techniques to isolate high-purity populations of native persisters has precluded direct measurement of persister metabolism. To address this technical challenge, we studied Escherichia coli populations whose growth had been inhibited by the accumulation of the MazF toxin, which catalyzes RNA cleavage, as a model system for persistence. Using chromosomally integrated, orthogonally inducible promoters to express MazF and its antitoxin MazE, bacterial populations that were almost entirely tolerant to fluoroquinolone and ß-lactam antibiotics were obtained upon MazF accumulation, and these were subjected to direct metabolic measurements. While MazF model persisters were nonreplicative, they maintained substantial oxygen and glucose consumption. Metabolomic analysis revealed accumulation of all four ribonucleotide monophosphates (NMPs). These results are consistent with a MazF-catalyzed RNA futile cycle, where the energy derived from catabolism is dissipated through continuous transcription and MazF-mediated RNA degradation. When transcription was inhibited, oxygen consumption and glucose uptake decreased, and nucleotide triphosphates (NTPs) and NTP/NMP ratios increased. Interestingly, the MazF-inhibited cells were sensitive to aminoglycosides, and this sensitivity was blocked by inhibition of transcription. Thus, in MazF model persisters, futile cycles of RNA synthesis and degradation result in both significant metabolic demands and aminoglycoside sensitivity. IMPORTANCE: Metabolism plays a critical role in controlling each stage of bacterial persistence (shutdown, stasis, and reawakening). In this work, we generated an E. coli strain in which the MazE antitoxin and MazF toxin were artificially and independently inducible, and we used this strain to generate model persisters and study their metabolism. We found that even though growth of the model persisters was inhibited, they remained highly metabolically active. We further uncovered a futile cycle driven by continued transcription and MazF-mediated transcript degradation that dissipated the energy derived from carbon catabolism. Interestingly, the existence of this futile cycle acted as an Achilles' heel for MazF model persisters, rendering them vulnerable to killing by aminoglycosides.


Subject(s)
DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , RNA, Bacterial/metabolism , Anti-Bacterial Agents/pharmacology , DNA-Binding Proteins/genetics , Drug Tolerance , Endoribonucleases/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Fluoroquinolones/pharmacology , Gene Expression , Glucose/metabolism , Oxygen/metabolism , Promoter Regions, Genetic , RNA Stability , Ribonucleotides/metabolism , Substrate Cycling , Transcription, Genetic , beta-Lactams/pharmacology
16.
Pharmaceuticals (Basel) ; 8(4): 778-92, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26593926

ABSTRACT

Staphylococcus aureus is a notorious pathogen with a propensity to cause chronic, non-healing wounds. Bacterial persisters have been implicated in the recalcitrance of S. aureus infections, and this motivated us to examine the persistence of S. aureus to ciprofloxacin, a quinolone antibiotic. Upon treatment of exponential phase S. aureus with ciprofloxacin, we observed that survival was a non-monotonic function of ciprofloxacin concentration. Maximal killing occurred at 1 µg/mL ciprofloxacin, which corresponded to survival that was up to ~40-fold lower than that obtained with concentrations ≥ 5 µg/mL. Investigation of this phenomenon revealed that the non-monotonic response was associated with prophage induction, which facilitated killing of S. aureus persisters. Elimination of prophage induction with tetracycline was found to prevent cell lysis and persister killing. We anticipate that these findings may be useful for the design of quinolone treatments.

17.
Curr Protoc Microbiol ; 36: 17.9.1-17.9.14, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25641098

ABSTRACT

Bacterial persisters are cells with an impressive, yet transient, tolerance toward extraordinary concentrations of antibiotics. Persisters are believed to impose a significant burden on the healthcare system because of their role in the proclivity of infections to relapse. During antibiotic challenge, these rare, phenotypic variants enter a dormant state where antibiotic primary targets are rendered inactive, allowing them to survive. Once the antibiotic is removed, persisters reawaken and resume growth, leading to repopulation of the environment. Metabolism plays a pivotal role in coordinating the entry, maintenance, and exit from the persister state. However, the low abundance, transient nature, and similarity of persisters to other cell types have prevented their isolation, which is needed for direct metabolic measurements. In this unit, we describe a technique known as the aminoglycoside (AG) potentiation assay, which can be used to rapidly and specifically measure the breadth of persister metabolism in heterogeneous populations.


Subject(s)
Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , Bacteria/drug effects , Bacteria/growth & development , Bacteriological Techniques/methods , Drug Tolerance , Microbial Viability/drug effects
18.
Antimicrob Agents Chemother ; 59(5): 2713-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25712354

ABSTRACT

Bacterial persisters are phenotypic variants with an extraordinary capacity to tolerate antibiotics, and they are hypothesized to be a main cause of chronic and relapsing infections. Recent evidence has suggested that the metabolism of persisters can be targeted to develop therapeutic countermeasures; however, knowledge of persister metabolism remains limited due to difficulties associated with isolating these rare and transient phenotypic variants. By using a technique to measure persister catabolic activity, which is based on the ability of metabolites to enable aminoglycoside (AG) killing of persisters, we investigated the role of seven global transcriptional regulators (ArcA, Cra, cyclic AMP [cAMP] receptor protein [CRP], DksA, FNR, Lrp, and RpoS) on persister metabolism. We found that removal of CRP resulted in a loss of AG potentiation in persisters for all metabolites tested. These results highlight a central role for cAMP/CRP in persister metabolism, as its perturbation can significantly diminish the metabolic capabilities of persisters and effectively eliminate the ability of AGs to eradicate these troublesome bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aminoglycosides/pharmacology , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Cyclic AMP Receptor Protein/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gentamicins/pharmacology , Microbial Sensitivity Tests , Repressor Proteins/genetics
19.
Front Microbiol ; 5: 70, 2014.
Article in English | MEDLINE | ID: mdl-24624123

ABSTRACT

Bacterial persisters are phenotypic variants with extraordinary tolerances toward antibiotics. Persister survival has been attributed to inhibition of essential cell functions during antibiotic stress, followed by reversal of the process and resumption of growth upon removal of the antibiotic. Metabolism plays a critical role in this process, since it participates in the entry, maintenance, and exit from the persister phenotype. Here, we review the experimental evidence that demonstrates the importance of metabolism to persistence, highlight the successes and potential of targeting metabolism in the search for anti-persister therapies, and discuss the current methods and challenges to understand persister physiology.

20.
Curr Pharm Des ; 20(5): 771-92, 2014.
Article in English | MEDLINE | ID: mdl-23688084

ABSTRACT

Our incessant tug-of-war with multidrug resistant pathogenic bacteria has prompted researchers to explore novel methods of designing therapeutics in order to defend ourselves against infectious diseases. Combined advances in whole genome analysis, bioinformatics algorithms, and biochemical techniques have led to the discovery and subsequent characterization of an abundant array of functional small peptides in microorganisms and multicellular organisms. Typically classified as having 10 to 100 amino acids, many of these peptides have been found to have dual activities, executing important defensive and regulatory functions in their hosts. In higher organisms, such as mammals, plants, and fungi, host defense peptides have been shown to have immunomodulatory and antimicrobial properties. In microbes, certain growth-inhibiting peptides have been linked to the regulation of diverse cellular processes. Examples of these processes include quorum sensing, stress response, cell differentiation, biofilm formation, pathogenesis, and multidrug tolerance. In this review, we will present a comprehensive overview of the discovery, characteristics, and functions of host- and bacteria-derived peptides with antimicrobial activities. The advantages and possible shortcomings of using these peptides as antimicrobial agents and targets will also be discussed. We will further examine current efforts in engineering synthetic peptides to be used as therapeutics and/or drug delivery vehicles.


Subject(s)
Anti-Infective Agents/therapeutic use , Drug Design , Drug Resistance, Multiple , Drugs, Investigational/therapeutic use , Host-Pathogen Interactions/drug effects , Infections/drug therapy , Peptides/therapeutic use , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacokinetics , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacokinetics , Antimicrobial Cationic Peptides/therapeutic use , Cell Membrane Permeability/drug effects , Drugs, Investigational/chemistry , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacokinetics , Humans , Immunomodulation/drug effects , Infections/immunology , Infections/metabolism , Lipid Bilayers/metabolism , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/metabolism , Membrane Transport Modulators/pharmacokinetics , Membrane Transport Modulators/therapeutic use , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacokinetics , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL