Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
Add more filters

Publication year range
1.
EMBO J ; 43(12): 2294-2307, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719995

ABSTRACT

Organisms rely on mutations to fuel adaptive evolution. However, many mutations impose a negative effect on fitness. Cells may have therefore evolved mechanisms that affect the phenotypic effects of mutations, thus conferring mutational robustness. Specifically, so-called buffer genes are hypothesized to interact directly or indirectly with genetic variation and reduce its effect on fitness. Environmental or genetic perturbations can change the interaction between buffer genes and genetic variation, thereby unmasking the genetic variation's phenotypic effects and thus providing a source of variation for natural selection to act on. This review provides an overview of our understanding of mutational robustness and buffer genes, with the chaperone gene HSP90 as a key example. It discusses whether buffer genes merely affect standing variation or also interact with de novo mutations, how mutational robustness could influence evolution, and whether mutational robustness might be an evolved trait or rather a mere side-effect of complex genetic interactions.


Subject(s)
Evolution, Molecular , HSP90 Heat-Shock Proteins , Mutation , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Selection, Genetic , Genetic Variation , Humans , Animals , Genetic Fitness
2.
Mol Cell ; 76(2): 255-267, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31626749

ABSTRACT

For decades, mankind has dominated the battle against bacteria, yet the tide is slowly turning. Our antibacterial strategies are becoming less effective, allowing bacteria to get the upper hand. The alarming rise in antibiotic resistance is an important cause of anti-infective therapy failure. However, other factors are at play as well. It is widely recognized that bacterial populations display high levels of heterogeneity. Population heterogeneity generates phenotypes specialized in surviving antibiotic attacks. Nonetheless, the presence of antibiotic-insensitive subpopulations is not considered when initiating treatment. It is therefore time to reevaluate how we combat bacterial infections. We here focus on antibiotic persistence and heteroresistance, phenomena in which small fractions of the population are tolerant (persisters) and resistant to antibiotics, respectively. We discuss molecular mechanisms involved, their clinical importance, and possible therapeutic strategies. Moving forward, we argue that these heterogeneous phenotypes should no longer be ignored in clinical practice and that better diagnostic and therapeutic approaches are urgently needed.


Subject(s)
Anti-Bacterial Agents , Bacteria/metabolism , Bacterial Infections/drug therapy , Bacterial Infections/metabolism , Drug Resistance, Bacterial/drug effects , Microbial Viability/drug effects , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Bacteria/genetics , Bacterial Infections/genetics , Bacterial Infections/microbiology , Drug Resistance, Bacterial/genetics , Humans
3.
Mol Cell ; 75(5): 1031-1042.e4, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31327636

ABSTRACT

Every bacterial population harbors a small subpopulation of so-called persisters that are transiently antibiotic tolerant. These persisters are associated with the recalcitrance of chronic infections because they can recolonize the host after antibiotic removal. Although several effectors have been described to induce persistence, persister cell awakening is poorly understood. We previously reported that the toxin HokB induces persistence via pore formation, resulting in membrane depolarization and ATP leakage. We now delineate mechanisms responsible for the awakening of HokB-induced persisters. We show that HokB dimerization by the oxidoreductase DsbA is essential for pore formation and peptide stability. Pores are disassembled via DsbC-mediated monomerization, which targets HokB for DegQ-mediated degradation. Finally, pore disassembly allows membrane repolarization by the electron transport chain, supporting cells to resume growth. These results provide a detailed view of both the formation and awakening of HokB-induced persister cells.


Subject(s)
Bacterial Toxins/metabolism , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Membrane Potentials/physiology , Proteolysis , Serine Endopeptidases/metabolism , Bacterial Toxins/genetics , Cell Membrane/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Serine Endopeptidases/genetics
4.
Mol Cell ; 70(5): 763-764, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29883603

ABSTRACT

Antibiotic-tolerant persister cells are difficult to eradicate by conventional classes of antibiotics. Kim and colleagues have discovered a new class of synthetic retinoid antibiotics that kill Staphylococcus aureus persisters by disrupting their cytoplasmic membrane.


Subject(s)
Anti-Bacterial Agents , Staphylococcus aureus , Humans , Retinoids , Staphylococcal Infections
5.
EMBO Rep ; 24(8): e57309, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37395716

ABSTRACT

Recalcitrant infections pose a serious challenge by prolonging antibiotic therapies and contributing to the spread of antibiotic resistance, thereby threatening the successful treatment of bacterial infections. One potential contributing factor in persistent infections is antibiotic persistence, which involves the survival of transiently tolerant subpopulations of bacteria. This review summarizes the current understanding of antibiotic persistence, including its clinical significance and the environmental and evolutionary factors at play. Additionally, we discuss the emerging concept of persister regrowth and potential strategies to combat persister cells. Recent advances highlight the multifaceted nature of persistence, which is controlled by deterministic and stochastic elements and shaped by genetic and environmental factors. To translate in vitro findings to in vivo settings, it is crucial to include the heterogeneity and complexity of bacterial populations in natural environments. As researchers continue to gain a more holistic understanding of this phenomenon and develop effective treatments for persistent bacterial infections, the study of antibiotic persistence is likely to become increasingly complex.


Subject(s)
Anti-Bacterial Agents , Bacterial Infections , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria/genetics , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Biological Evolution , Environment , Drug Resistance, Bacterial/genetics
6.
Nucleic Acids Res ; 51(7): 3420-3435, 2023 04 24.
Article in English | MEDLINE | ID: mdl-36864742

ABSTRACT

Obg is a widely conserved and essential GTPase in bacteria, which plays a central role in a large range of important cellular processes, such as ribosome biogenesis, DNA replication, cell division and bacterial persistence. Nevertheless, the exact function of Obg in these processes and the interactions it makes within the associated pathways remain largely unknown. Here, we identify the DNA-binding TrpD2 protein YbiB as an interactor of the Escherichia coli Obg (ObgE). We show that both proteins interact with high affinity in a peculiar biphasic fashion, and pinpoint the intrinsically disordered and highly negatively charged C-terminal domain of ObgE as a main driver for this interaction. Molecular docking and X-ray crystallography, together with site-directed mutagenesis, are used to map the binding site of this ObgE C-terminal domain within a highly positively charged groove on the surface of the YbiB homodimer. Correspondingly, ObgE efficiently inhibits the binding of DNA to YbiB, indicating that ObgE competes with DNA for binding in the positive clefts of YbiB. This study thus forms an important step for the further elucidation of the interactome and cellular role of the essential bacterial protein Obg.


Subject(s)
Escherichia coli Proteins , Monomeric GTP-Binding Proteins , Escherichia coli Proteins/metabolism , Monomeric GTP-Binding Proteins/genetics , Molecular Docking Simulation , Escherichia coli/genetics , Escherichia coli/metabolism , Bacterial Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins/metabolism
7.
Plant J ; 116(1): 7-22, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37608631

ABSTRACT

Strigolactones are a class of phytohormones that are involved in many different plant developmental processes, including the rhizobium-legume nodule symbiosis. Although both positive and negative effects of strigolactones on the number of nodules have been reported, the influence of strigolactones on nodule development is still unknown. Here, by means of the ramosus (rms) mutants of Pisum sativum (pea) cv Terese, we investigated the impact of strigolactone biosynthesis (rms1 and rms5) and signaling (rms3 and rms4) mutants on nodule growth. The rms mutants had more red, that is, functional, and larger nodules than the wild-type plants. Additionally, the increased nitrogen fixation and senescence zones with consequently reduced meristematic and infection zones indicated that the rms nodules developed faster than the wild-type nodules. An enhanced expression of the nodule zone-specific molecular markers for meristem activity and senescence supported the enlarged, fast maturing nodules. Interestingly, the master nodulation regulator, NODULE INCEPTION, NIN, was strongly induced in nodules of all rms mutants but not prior to inoculation. Determination of sugar levels with both bulk and spatial metabolomics in roots and nodules, respectively, hints at slightly increased malic acid levels early during nodule primordia formation and reduced sugar levels at later stages, possibly the consequence of an increased carbon usage of the enlarged nodules, contributing to the enhanced senescence. Taken together, these results suggest that strigolactones regulate the development of nodules, which is probably mediated through NIN, and available plant sugars.


Subject(s)
Pisum sativum , Plant Growth Regulators , Pisum sativum/genetics , Plant Growth Regulators/metabolism , Plant Roots/metabolism , Nitrogen Fixation/physiology , Symbiosis/physiology , Sugars/metabolism , Root Nodules, Plant/metabolism , Gene Expression Regulation, Plant , Plant Proteins/metabolism
8.
Nat Prod Rep ; 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38465694

ABSTRACT

Covering: up to October 2023Many bioactive natural products are synthesized by microorganisms that are either difficult or impossible to cultivate under laboratory conditions, or that produce only small amounts of the desired compound. By transferring biosynthetic gene clusters (BGCs) into alternative host organisms that are more easily cultured and engineered, larger quantities can be obtained and new analogues with potentially improved biological activity or other desirable properties can be generated. Moreover, expression of cryptic BGCs in a suitable host can facilitate the identification and characterization of novel natural products. Heterologous expression therefore represents a valuable tool for natural product discovery and engineering as it allows the study and manipulation of their biosynthetic pathways in a controlled setting, enabling innovative applications. Bacillus is a genus of Gram-positive bacteria that is widely used in industrial biotechnology as a host for the production of proteins from diverse origins, including enzymes and vaccines. However, despite numerous successful examples, Bacillus species remain underexploited as heterologous hosts for the expression of natural product BGCs. Here, we review important advantages that Bacillus species offer as expression hosts, such as high secretion capacity, natural competence for DNA uptake, and the increasing availability of a wide range of genetic tools for gene expression and strain engineering. We evaluate different strain optimization strategies and other critical factors that have improved the success and efficiency of heterologous natural product biosynthesis in B. subtilis. Finally, future perspectives for using B. subtilis as a heterologous host are discussed, identifying research gaps and promising areas that require further exploration.

9.
Mol Cell ; 59(1): 9-21, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26051177

ABSTRACT

Within bacterial populations, a small fraction of persister cells is transiently capable of surviving exposure to lethal doses of antibiotics. As a bet-hedging strategy, persistence levels are determined both by stochastic induction and by environmental stimuli called responsive diversification. Little is known about the mechanisms that link the low frequency of persisters to environmental signals. Our results support a central role for the conserved GTPase Obg in determining persistence in Escherichia coli in response to nutrient starvation. Obg-mediated persistence requires the stringent response alarmone (p)ppGpp and proceeds through transcriptional control of the hokB-sokB type I toxin-antitoxin module. In individual cells, increased Obg levels induce HokB expression, which in turn results in a collapse of the membrane potential, leading to dormancy. Obg also controls persistence in Pseudomonas aeruginosa and thus constitutes a conserved regulator of antibiotic tolerance. Combined, our findings signify an important step toward unraveling shared genetic mechanisms underlying persistence.


Subject(s)
Bacterial Proteins/genetics , Bacterial Toxins/biosynthesis , Drug Resistance, Bacterial/physiology , Escherichia coli Proteins/biosynthesis , Escherichia coli/genetics , GTP-Binding Proteins/genetics , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/genetics , Cell Membrane/physiology , Escherichia coli Proteins/genetics , Membrane Potentials/genetics , Microbial Sensitivity Tests , Protein Structure, Tertiary/genetics
10.
Nucleic Acids Res ; 49(12): 7164-7178, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34139012

ABSTRACT

The rnlAB toxin-antitoxin operon from Escherichia coli functions as an anti-phage defense system. RnlA was identified as a member of the HEPN (Higher Eukaryotes and Prokaryotes Nucleotide-binding domain) superfamily of ribonucleases. The activity of the toxin RnlA requires tight regulation by the antitoxin RnlB, the mechanism of which remains unknown. Here we show that RnlA exists in an equilibrium between two different homodimer states: an inactive resting state and an active canonical HEPN dimer. Mutants interfering with the transition between states show that canonical HEPN dimerization via the highly conserved RX4-6H motif is required for activity. The antitoxin RnlB binds the canonical HEPN dimer conformation, inhibiting RnlA by blocking access to its active site. Single-alanine substitutions mutants of the highly conserved R255, E258, R318 and H323 show that these residues are involved in catalysis and substrate binding and locate the catalytic site near the dimer interface of the canonical HEPN dimer rather than in a groove located between the HEPN domain and the preceding TBP-like domain. Overall, these findings elucidate the structural basis of the activity and inhibition of RnlA and highlight the crucial role of conformational heterogeneity in protein function.


Subject(s)
Escherichia coli Proteins/chemistry , Ribonucleases/chemistry , Dimerization , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Multimerization , Ribonucleases/genetics , Ribonucleases/metabolism , Substrate Specificity
11.
Mol Biol Evol ; 38(8): 3345-3357, 2021 07 29.
Article in English | MEDLINE | ID: mdl-33871643

ABSTRACT

Bacterial persistence is a potential cause of antibiotic therapy failure. Antibiotic-tolerant persisters originate from phenotypic differentiation within a susceptible population, occurring with a frequency that can be altered by mutations. Recent studies have proven that persistence is a highly evolvable trait and, consequently, an important evolutionary strategy of bacterial populations to adapt to high-dose antibiotic therapy. Yet, the factors that govern the evolutionary dynamics of persistence are currently poorly understood. Theoretical studies predict far-reaching effects of bottlenecking on the evolutionary adaption of bacterial populations, but these effects have never been investigated in the context of persistence. Bottlenecking events are frequently encountered by infecting pathogens during host-to-host transmission and antibiotic treatment. In this study, we used a combination of experimental evolution and barcoded knockout libraries to examine how population bottlenecking affects the evolutionary dynamics of persistence. In accordance with existing hypotheses, small bottlenecks were found to restrict the adaptive potential of populations and result in more heterogeneous evolutionary outcomes. Evolutionary trajectories followed in small-bottlenecking regimes additionally suggest that the fitness landscape associated with persistence has a rugged topography, with distinct trajectories toward increased persistence that are accessible to evolving populations. Furthermore, sequencing data of evolved populations and knockout libraries after selection reveal various genes that are potentially involved in persistence, including previously known as well as novel targets. Together, our results do not only provide experimental evidence for evolutionary theories, but also contribute to a better understanding of the environmental and genetic factors that guide bacterial adaptation to antibiotic treatment.


Subject(s)
Biological Evolution , Drug Resistance, Bacterial/genetics , Escherichia coli , Gene Editing , Genetic Fitness , Population Dynamics
12.
Trends Genet ; 35(6): 401-411, 2019 06.
Article in English | MEDLINE | ID: mdl-31036343

ABSTRACT

All bacterial populations harbor a small fraction of transiently antibiotic-tolerant cells called persisters. These phenotypic variants compromise successful antibiotic treatment because they are held responsible for the relapse of many chronic infections. In addition, studies employing experimental evolution have demonstrated that persistence contributes to the development of antibiotic resistance. Persisters are typically described as dormant cells. However, recent findings indicate a role for active mechanisms in the formation and maintenance of the persister phenotype. This review summarizes novel insights into the molecular mechanisms of persister formation and awakening, focusing on changes in cell physiology mediated by persistence effectors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacterial Physiological Phenomena/drug effects , Drug Resistance, Bacterial , Bacteria/genetics , Bacteria/metabolism , Bacterial Physiological Phenomena/genetics , DNA Replication , Energy Metabolism/drug effects , Gene Expression Regulation, Bacterial/drug effects , Transcription, Genetic
13.
Anal Chem ; 94(45): 15781-15789, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36377427

ABSTRACT

Partitions in digital PCR (dPCR) assays do not reach the detection threshold at the same time. This heterogeneity in amplification results in intermediate endpoint fluorescence values (i.e., rain) and misclassification of partitions, which has a major impact on the accuracy of nucleic acid quantification. Rain most often results from a reduced amplification efficiency or template inaccessibility; however, exactly how these contribute to rain has not been described. We developed and experimentally validated an analytical model that mechanistically explains the relationship between amplification efficiency, template accessibility, and rain. Using Monte Carlo simulations, we show that a reduced amplification efficiency leads to broader threshold cycle (Ct) distributions that can be fitted using a log-normal probability distribution. From the fit parameters, the amplification efficiency can be calculated. Template inaccessibility, on the other hand, leads to a different rain pattern, in which a distinct exponential tail in the Ct distribution can be observed. Using our model, it is possible to determine if the amplification efficiency, template accessibility, or another source is the main contributor of rain in dPCR assays. We envision that this model will facilitate and speed up dPCR assay optimization and provide an indication for the accuracy of the assay.


Subject(s)
Rain , Polymerase Chain Reaction/methods , Monte Carlo Method
14.
PLoS Pathog ; 16(5): e1008431, 2020 05.
Article in English | MEDLINE | ID: mdl-32379814

ABSTRACT

Bacteria are well known for their extremely high adaptability in stressful environments. The clinical relevance of this property is clearly illustrated by the ever-decreasing efficacy of antibiotic therapies. Frequent exposures to antibiotics favor bacterial strains that have acquired mechanisms to overcome drug inhibition and lethality. Many strains, including life-threatening pathogens, exhibit increased antibiotic resistance or tolerance, which considerably complicates clinical practice. Alarmingly, recent studies show that in addition to resistance, tolerance levels of bacterial populations are extremely flexible in an evolutionary context. Here, we summarize laboratory studies providing insight in the evolution of resistance and tolerance and shed light on how the treatment conditions could affect the direction of bacterial evolution under antibiotic stress.


Subject(s)
Adaptation, Biological/drug effects , Bacteria/drug effects , Drug Resistance, Bacterial/drug effects , Adaptation, Biological/genetics , Adaptation, Physiological/drug effects , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Drug Resistance, Microbial/drug effects , Evolution, Molecular
15.
Nucleic Acids Res ; 47(W1): W151-W157, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31127271

ABSTRACT

IAMBEE is a web server designed for the Identification of Adaptive Mutations in Bacterial Evolution Experiments (IAMBEE). Input data consist of genotype information obtained from independently evolved clonal populations or strains that show the same adapted behavior (phenotype). To distinguish adaptive from passenger mutations, IAMBEE searches for neighborhoods in an organism-specific interaction network that are recurrently mutated in the adapted populations. This search for recurrently mutated network neighborhoods, as proxies for pathways is driven by additional information on the functional impact of the observed genetic changes and their dynamics during adaptive evolution. In addition, the search explicitly accounts for the differences in mutation rate between the independently evolved populations. Using this approach, IAMBEE allows exploiting parallel evolution to identify adaptive pathways. The web-server is freely available at http://bioinformatics.intec.ugent.be/iambee/ with no login requirement.


Subject(s)
Adaptation, Biological/genetics , Bacteria/genetics , Clonal Evolution/genetics , Databases, Genetic , Software , Genotype , Mutation/genetics , Mutation Rate , Phenotype , Web Browser
16.
Mol Microbiol ; 112(5): 1593-1608, 2019 11.
Article in English | MEDLINE | ID: mdl-31498933

ABSTRACT

Obg is a versatile GTPase that plays a pivotal role in bacterial persistence. We previously showed that the Escherichia coli homolog ObgE exerts this activity through transcriptional activation of a toxin-antitoxin module and subsequent membrane depolarization. Here, we assessed the role of G-domain functionality in ObgE-mediated persistence. Through screening of a mutant library, we identified five obgE alleles (with substitutions G166V, D246G, S270I, N283I and I313N) that have lost their persistence function and no longer activate hokB expression. These alleles support viability of a strain otherwise deprived of ObgE, indicating that ObgE's persistence function can be uncoupled from its essential role. Based on the ObgE crystal structure, we designed two additional mutant proteins (T193A and D286Y), one of which (D286Y) no longer affects persistence. Using isothermal titration calorimetry, stopped-flow experiments and kinetics, we subsequently assessed nucleotide binding and GTPase activity in all mutants. With the exception of the S270I mutant that is possibly affected in protein-protein interactions, all mutants that have lost their persistence function display severely reduced binding to GDP or the alarmone ppGpp. However, we find no clear relation between persistence and GTP or pppGpp binding nor with GTP hydrolysis. Combined, our results signify an important step toward understanding biochemical determinants underlying persistence.


Subject(s)
Bacterial Toxins/biosynthesis , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Escherichia coli/physiology , Monomeric GTP-Binding Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Structure-Activity Relationship , Transcriptional Activation/genetics
17.
Metab Eng ; 62: 287-297, 2020 11.
Article in English | MEDLINE | ID: mdl-32979485

ABSTRACT

Although high hydrostatic pressure (HHP) is an interesting parameter to be applied in bioprocessing, its potential is currently limited by the lack of bacterial chassis capable of surviving and maintaining homeostasis under pressure. While several efforts have been made to genetically engineer microorganisms able to grow at sublethal pressures, there is little information for designing backgrounds that survive more extreme pressures. In this investigation, we analyzed the genome of an extreme HHP-resistant mutant of E. coli MG1655 (designated as DVL1), from which we identified four mutations (in the cra, cyaA, aceA and rpoD loci) causally linked to increased HHP resistance. Analysing the functional effect of these mutations we found that the coupled effect of downregulation of cAMP/CRP, Cra and the glyoxylate shunt activity, together with the upregulation of RpoH and RpoS activity, could mechanistically explain the increased HHP resistance of the mutant. Using combinations of three mutations, we could synthetically engineer E. coli strains able to comfortably survive pressures of 600-800 MPa, which could serve as genetic backgrounds for HHP-based biotechnological applications.


Subject(s)
Bacteria , Escherichia coli , Escherichia coli/genetics , Hydrostatic Pressure , Mutation
18.
Appl Microbiol Biotechnol ; 104(9): 3757-3770, 2020 May.
Article in English | MEDLINE | ID: mdl-32170388

ABSTRACT

Plant growth-promoting bacteria show great potential for use in agriculture although efficient application remains challenging to achieve. Cells often lose viability during inoculant production and application, jeopardizing the efficacy of the inoculant. Since desiccation has been documented to be the primary stress factor affecting the decrease in survival, obtaining xerotolerance in plant growth-promoting bacteria is appealing. The molecular damage that occurs by drying bacteria has been broadly investigated, although a complete view is still lacking due to the complex nature of the process. Mechanic, structural, and metabolic changes that occur as a result of water depletion may potentially afflict lethal damage to membranes, DNA, and proteins. Bacteria respond to these harsh conditions by increasing production of exopolysaccharides, changing composition of the membrane, improving the stability of proteins, reducing oxidative stress, and repairing DNA damage. This review provides insight into the complex nature of desiccation stress in bacteria in order to facilitate strategic choices to improve survival and shelf life of newly developed inoculants. KEY POINTS: Desiccation-induced damage affects most major macromolecules in bacteria. Most bacteria are not xerotolerant despite multiple endogenous adaption mechanisms. Sensitivity to drying severely hampers inoculant quality.


Subject(s)
Agricultural Inoculants/metabolism , Bacteria/metabolism , Desiccation , Stress, Physiological , Temperature , Adaptation, Physiological , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/pathology
19.
Drug Resist Updat ; 38: 12-26, 2018 05.
Article in English | MEDLINE | ID: mdl-29857815

ABSTRACT

In addition to the well-known strategies of antibiotic resistance and biofilm formation, bacterial populations possess an additional survival strategy to endure hostile environments or antibiotic exposure. A small fraction of transiently antibiotic-tolerant phenotypical variants, called persister cells, is capable of surviving treatment with high doses of antibiotics. When antibiotic pressure drops, persisters that switch back to a normal phenotype can resume growth, ensuring survival of the bacterial population. Persister cells have been identified in every major pathogen, contribute to the antibiotic tolerance observed in biofilms, and are responsible for the recalcitrant nature of chronic infections. Also, evidence is accumulating that persister cells can contribute to the emergence of antibiotic resistance. Consequently, effective treatment of persister cells could greatly improve patient outcome. The small number of persisters and the redundancy in mechanisms of persister formation impede target-based development of anti-persister therapies. Nonetheless, the armory of anti-persister molecules is increasing. This review presents a comprehensive overview of anti-persister molecules and strategies described in literature to date and offers perspectives on potential anti-persistence targets and methods for the development of future therapies. Furthermore, we highlight in vivo model systems for pre-clinical testing and summarize ongoing clinical trials of candidate anti-persister therapeutics.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacteria/drug effects , Biofilms/drug effects , Drug Resistance, Bacterial , Animals , Anti-Bacterial Agents/adverse effects , Bacteria/genetics , Bacteria/growth & development , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , Drug Design , Gene Expression Regulation, Bacterial , Genotype , Humans , Microbial Viability/drug effects , Phenotype , Signal Transduction/drug effects
20.
Int J Mol Sci ; 21(1)2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31861427

ABSTRACT

Even though the Obg protein is essential for bacterial viability, the cellular functions of this universally conserved GTPase remain enigmatic. Moreover, the influence of GTP and GDP binding on the activity of this protein is largely unknown. Previously, we identified a mutant isoform of ObgE (the Obg protein of Escherichia coli) that triggers cell death. In this research we explore the biochemical requirements for the toxic effect of this mutant ObgE* isoform, using cell death as a readily accessible read-out for protein activity. Both the absence of the N-terminal domain and a decreased GTP binding affinity neutralize ObgE*-mediated toxicity. Moreover, a deletion in the region that connects the N-terminal domain to the G domain likewise abolishes toxicity. Taken together, these data indicate that GTP binding by ObgE* triggers a conformational change that is transmitted to the N-terminal domain to confer toxicity. We therefore conclude that ObgE*-GTP, but not ObgE*-GDP, is the active form of ObgE* that is detrimental to cell viability. Based on these data, we speculate that also for wild-type ObgE, GTP binding triggers conformational changes that affect the N-terminal domain and thereby control ObgE function.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Guanosine Triphosphate/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Escherichia coli Proteins/chemistry , Guanosine Triphosphate/chemistry , Models, Molecular , Monomeric GTP-Binding Proteins/chemistry , Mutant Proteins , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Isoforms , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL