Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Microbiology (Reading) ; 169(10)2023 10.
Article in English | MEDLINE | ID: mdl-37787650

ABSTRACT

Multidrug efflux pumps are molecular machines that sit in the bacterial cell membrane and pump molecules out from either the periplasm or cytoplasm to outside the cell. While involved in a variety of biological roles, they are primarily known for their contribution to antibiotic resistance by limiting the intracellular accumulation of antimicrobial compounds within bacteria. These transporters are often overexpressed in clinical isolates, leading to multidrug-resistant phenotypes. Efflux pumps are classified into several families based on their structure and understanding the characteristics of each family is important for the development of novel therapies to restore antibiotic potency.


Subject(s)
Anti-Bacterial Agents , Periplasm , Humans , Cytoplasm , Cell Membrane , Anti-Bacterial Agents/pharmacology , Membrane Transport Proteins/genetics
2.
Microbiology (Reading) ; 169(1)2023 01.
Article in English | MEDLINE | ID: mdl-36748572

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen frequently isolated from chronic infections of the cystic fibrosis lung and burn wounds, and is a major cause of antimicrobial-resistant nosocomial infections. P. aeruginosa is frequently co-isolated with the opportunistic fungal pathogen Candida albicans, with the presence of C. albicans in dual-species biofilms promoting tolerance to meropenem. Here, transcription profiling of mature P. aeruginosa single- or dual-species biofilms was carried out to understand the molecular mechanism(s) by which C. albicans enhances meropenem tolerance. C. albicans appeared to have a mild impact on the transcriptome of P. aeruginosa mature biofilms, with most differentially regulated genes being involved in interkingdom interactions (i.e. quorum sensing and phenazine biosynthesis). The addition of meropenem to mature single- or dual-species biofilms resulted in a significant bacterial transcriptional response, including the induction of the beta-lactamase, ampC, genes involved in biofilm formation. P. aeruginosa elicited a similar transcriptional response to meropenem in the presence of C. albicans, but C. albicans promoted the expression of additional efflux pumps, which could play roles in increasing the tolerance of P. aeruginosa to meropenem.


Subject(s)
Biofilms , Pseudomonas aeruginosa , Pseudomonas aeruginosa/genetics , Meropenem/pharmacology , Candida albicans/physiology , Quorum Sensing/genetics
3.
Microbiology (Reading) ; 169(4)2023 04.
Article in English | MEDLINE | ID: mdl-37074150

ABSTRACT

Escherichia coli is a facultative anaerobe found in a wide range of environments. Commonly described as the laboratory workhorse, E. coli is one of the best characterized bacterial species to date, however much of our understanding comes from studies involving the laboratory strain E. coli K-12. Resistance-nodulation-division efflux pumps are found in Gram-negative bacteria and can export a diverse range of substrates, including antibiotics. E. coli K-12 has six RND pumps; AcrB, AcrD, AcrF, CusA, MdtBC and MdtF, and it is frequently reported that all E. coli strains possess these six pumps. However, this is not true of E. coli ST11, a lineage of E. coli, which is primarily composed of the highly virulent important human pathogen, E. coli O157:H7. Here we show that acrF is absent from the pangenome of ST11 and that this lineage of E. coli has a highly conserved insertion within the acrF gene, which when translated encodes 13 amino acids and two stop codons. This insertion was found to be present in 97.59 % of 1787 ST11 genome assemblies. Non-function of AcrF in ST11 was confirmed in the laboratory as complementation with acrF from ST11 was unable to restore AcrF function in E. coli K-12 substr. MG1655 ΔacrB ΔacrF. This shows that the complement of RND efflux pumps present in laboratory bacterial strains may not reflect the situation in virulent strains of bacterial pathogens.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Membrane Proteins/metabolism
4.
J Antimicrob Chemother ; 78(9): 2228-2241, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37522316

ABSTRACT

BACKGROUND: Biofilms are the leading cause of nosocomial infections and are hard to eradicate due to their inherent antimicrobial resistance. Candida albicans is the leading cause of nosocomial fungal infections and is frequently co-isolated with the bacterium Pseudomonas aeruginosa from biofilms in the cystic fibrosis lung and severe burn wounds. The presence of C. albicans in multispecies biofilms is associated with enhanced antibacterial resistance, which is largely mediated through fungal extracellular carbohydrates sequestering the antibiotics. However, significantly less is known regarding the impact of polymicrobial biofilms on antifungal resistance. RESULTS: Here we show that, in dual-species biofilms, P. aeruginosa enhances the susceptibility of C. albicans to amphotericin B, an effect that was biofilm specific. Transcriptional analysis combined with gene ontology enrichment analysis identified several C. albicans processes associated with oxidative stress to be differentially regulated in dual-species biofilms, suggesting that P. aeruginosa exerts oxidative stress on C. albicans, likely through the secretion of phenazines. However, the mitochondrial superoxide dismutase SOD2 was significantly down-regulated in the presence of P. aeruginosa. Monospecies biofilms of the sod2Δ mutant were more susceptible to amphotericin B, and the susceptibility of these biofilms was further enhanced by exogenous phenazines. CONCLUSIONS: We propose that in dual-species biofilms, P. aeruginosa simultaneously induces mitochondrial oxidative stress, while down-regulating key detoxification enzymes, which prevents C. albicans mounting an appropriate oxidative stress response to amphotericin B, leading to fungal cell death. This work highlights the importance of understanding the impact of polymicrobial interactions on antimicrobial susceptibility.


Subject(s)
Amphotericin B , Candida albicans , Amphotericin B/pharmacology , Pseudomonas aeruginosa , Biofilms , Anti-Bacterial Agents/pharmacology , Phenazines , Antifungal Agents/pharmacology
5.
Chem Rev ; 121(9): 5479-5596, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33909410

ABSTRACT

Tripartite efflux pumps and the related type 1 secretion systems (T1SSs) in Gram-negative organisms are diverse in function, energization, and structural organization. They form continuous conduits spanning both the inner and the outer membrane and are composed of three principal components-the energized inner membrane transporters (belonging to ABC, RND, and MFS families), the outer membrane factor channel-like proteins, and linking the two, the periplasmic adaptor proteins (PAPs), also known as the membrane fusion proteins (MFPs). In this review we summarize the recent advances in understanding of structural biology, function, and regulation of these systems, highlighting the previously undescribed role of PAPs in providing a common architectural scaffold across diverse families of transporters. Despite being built from a limited number of basic structural domains, these complexes present a staggering variety of architectures. While key insights have been derived from the RND transporter systems, a closer inspection of the operation and structural organization of different tripartite systems reveals unexpected analogies between them, including those formed around MFS- and ATP-driven transporters, suggesting that they operate around basic common principles. Based on that we are proposing a new integrated model of PAP-mediated communication within the conformational cycling of tripartite systems, which could be expanded to other types of assemblies.


Subject(s)
Gram-Negative Bacteria/metabolism , Membrane Transport Proteins/metabolism , Type I Secretion Systems/metabolism , ATP-Binding Cassette Transporters , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Gram-Negative Bacteria/chemistry , Membrane Transport Proteins/chemistry , Molecular Dynamics Simulation , Protein Conformation , Structure-Activity Relationship , Type I Secretion Systems/chemistry
6.
J Antimicrob Chemother ; 78(1): 133-140, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36308324

ABSTRACT

BACKGROUND: Resistance nodulation division (RND) family efflux pumps, including the major pump AcrAB-TolC, are important mediators of intrinsic and evolved antibiotic resistance. Expression of these pumps is carefully controlled by a network of regulators that respond to different environmental cues. EnvR is a TetR family transcriptional regulator encoded upstream of the RND efflux pump acrEF. METHODS: Binding of EnvR protein upstream of acrAB was determined by electrophoretic mobility shift assays and the phenotypic consequence of envR overexpression on antimicrobial susceptibility, biofilm motility and invasion of eukaryotic cells in vitro was measured. Additionally, the global transcriptome of clinical Salmonella isolates overexpressing envR was determined by RNA-Seq. RESULTS: EnvR bound to the promoter region upstream of the genes coding for the major efflux pump AcrAB in Salmonella, inhibiting transcription and preventing production of AcrAB protein. The phenotype conferred by overexpression of envR mimicked deletion of acrB as it conferred multidrug susceptibility, decreased motility and decreased invasion into intestinal cells in vitro. Importantly, we demonstrate the clinical relevance of this regulatory mechanism because RNA-Seq revealed that a drug-susceptible clinical isolate of Salmonella had low acrB expression even though expression of its major regulator RamA was very high; this was caused by very high EnvR expression. CONCLUSIONS: In summary, we show that EnvR is a potent repressor of acrAB transcription in Salmonella, and can override binding by RamA so preventing MDR to clinically useful drugs. Finding novel tools to increase EnvR expression may form the basis of a new way to prevent or treat MDR infections.


Subject(s)
Bacterial Proteins , Salmonella typhimurium , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Drug Resistance, Microbial , Salmonella typhimurium/genetics , Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription, Genetic
7.
Bull Math Biol ; 84(5): 56, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35380320

ABSTRACT

Bacteria have developed resistance to antibiotics by various mechanisms, notable amongst these is the use of permeation barriers and the expulsion of antibiotics via efflux pumps. The resistance-nodulation-division (RND) family of efflux pumps is found in Gram-negative bacteria and a major contributor to multidrug resistance (MDR). In particular, Salmonella encodes five RND efflux pump systems: AcrAB, AcrAD, AcrEF, MdsAB and MdtAB which have different substrate ranges including many antibiotics. We produce a spatial partial differential equation (PDE) model governing the diffusion and efflux of antibiotic in Salmonella, via these RND efflux pumps. Using parameter fitting techniques on experimental data, we are able to establish the behaviour of multiple wild-type and efflux mutant Salmonella strains, which enables us to produce efflux profiles for each individual efflux pump system. By combining the model with a gene regulatory network (GRN) model of efflux regulation, we simulate how the bacteria respond to their environment. Finally, performing a parameter sensitivity analysis, we look into various different targets to inhibit the efflux pumps. The model provides an in silico framework with which to test these potential adjuvants to counter MDR.


Subject(s)
Drug Resistance, Multiple, Bacterial , Membrane Transport Proteins , Models, Biological , Salmonella , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Membrane Transport Proteins/genetics , Salmonella/drug effects , Salmonella/genetics
8.
J Antimicrob Chemother ; 76(10): 2558-2564, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34278432

ABSTRACT

BACKGROUND: Resistance-nodulation-division (RND) efflux pumps are important mediators of antibiotic resistance. RND pumps, including the principal multidrug efflux pump AcrAB-TolC in Salmonella, are tripartite systems with an inner membrane RND transporter, a periplasmic adaptor protein (PAP) and an outer membrane factor (OMF). We previously identified the residues required for binding between the PAP AcrA and the RND transporter AcrB and have demonstrated that PAPs can function with non-cognate transporters. AcrE and AcrD/AcrF are homologues of AcrA and AcrB, respectively. Here, we show that AcrE can interact with AcrD, which does not possess its own PAP, and establish that the residues previously identified in AcrB binding are also involved in AcrD binding. METHODS: The acrD and acrE genes were expressed in a strain lacking acrABDEF (Δ3RND). PAP residues involved in promiscuous interactions were predicted based on previously defined PAP-RND interactions and corresponding mutations generated in acrA and acrE. Antimicrobial susceptibility of the mutant strains was determined. RESULTS: Co-expression of acrD and acrE significantly decreased susceptibility of the Δ3RND strain to AcrD substrates, showing that AcrE can form a functional complex with AcrD. The substrate profile of Salmonella AcrD differed from that of Escherichia coli AcrD. Mutations targeting the previously defined PAP-RND interaction sites in AcrA/AcrE impaired efflux of AcrD-dependent substrates. CONCLUSIONS: These data indicate that AcrE forms an efflux-competent pump with AcrD and thus presents an alternative PAP for this pump. Mutagenesis of the conserved RND binding sites validates the interchangeability of AcrA and AcrE, highlighting them as potential drug targets for efflux inhibition.


Subject(s)
Periplasmic Proteins , Salmonella typhimurium , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Salmonella typhimurium/genetics , Serogroup
9.
J Antimicrob Chemother ; 76(5): 1130-1134, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33501980

ABSTRACT

OBJECTIVES: To characterize a blaOXA-58- and blaNDM-1-containing MDR plasmid from a rare Acinetobacter baumannii lineage and compare it with related plasmids to explore the distribution and evolution of a new plasmid group. METHODS: A. baumannii DETAB-P2 was isolated from a rectal swab of an intensive care patient. Antibiotic susceptibility was determined using broth microdilution. DETAB-P2 was mated with A. baumannii ATCC 17978 and putative transconjugants were characterized by S1/PFGE and Southern hybridization. WGS was performed on the Illumina and Oxford Nanopore platforms. MLST was performed with the Pasteur and Oxford schemes. Antibiotic resistance genes were identified with ABRicate. Plasmid sequence annotation was performed manually. Complete plasmids in GenBank with the same rep gene were used for comparative analyses. RESULTS: A. baumannii DETAB-P2 was ST138 by the Pasteur scheme and a novel Oxford type, ST2209. It transferred blaOXA-58 and blaNDM-1 to ATCC 17978 in the 100 072 bp plasmid pDETAB2 that also carried bleMBL, sul2, aacC2d, tet(39), msr(E)-mph(E) and putative mercury resistance and RND efflux system determinants. pDETAB2 represents a new plasmid type, GR34, and contained 16 pdif sites and several novel dif modules. Only a 10 kbp core sequence is shared amongst pDETAB2 and 18 further GR34 plasmids in GenBank, with diverse accessory regions comprised of various dif modules. CONCLUSIONS: GR34 plasmids are found in several Acinetobacter species from diverse environments. They display considerable variation in accessory content owing to the presence of pdif sites and an array of dif modules, some of which contain antibiotic resistance genes.


Subject(s)
Acinetobacter baumannii , Acinetobacter baumannii/genetics , Anti-Bacterial Agents/pharmacology , Humans , Microbial Sensitivity Tests , Multilocus Sequence Typing , Plasmids/genetics , beta-Lactamases/genetics
10.
PLoS Pathog ; 15(12): e1008101, 2019 12.
Article in English | MEDLINE | ID: mdl-31877175

ABSTRACT

Active efflux due to tripartite RND efflux pumps is an important mechanism of clinically relevant antibiotic resistance in Gram-negative bacteria. These pumps are also essential for Gram-negative pathogens to cause infection and form biofilms. They consist of an inner membrane RND transporter; a periplasmic adaptor protein (PAP), and an outer membrane channel. The role of PAPs in assembly, and the identities of specific residues involved in PAP-RND binding, remain poorly understood. Using recent high-resolution structures, four 3D sites involved in PAP-RND binding within each PAP protomer were defined that correspond to nine discrete linear binding sequences or "binding boxes" within the PAP sequence. In the important human pathogen Salmonella enterica, these binding boxes are conserved within phylogenetically-related PAPs, such as AcrA and AcrE, while differing considerably between divergent PAPs such as MdsA and MdtA, despite overall conservation of the PAP structure. By analysing these binding sequences we created a predictive model of PAP-RND interaction, which suggested the determinants that may allow promiscuity between certain PAPs, but discrimination of others. We corroborated these predictions using direct phenotypic data, confirming that only AcrA and AcrE, but not MdtA or MsdA, can function with the major RND pump AcrB. Furthermore, we provide functional validation of the involvement of the binding boxes by disruptive site-directed mutagenesis. These results directly link sequence conservation within identified PAP binding sites with functional data providing mechanistic explanation for assembly of clinically relevant RND-pumps and explain how Salmonella and other pathogens maintain a degree of redundancy in efflux mediated resistance. Overall, our study provides a novel understanding of the molecular determinants driving the RND-PAP recognition by bridging the available structural information with experimental functional validation thus providing the scientific community with a predictive model of pump-contacts that could be exploited in the future for the development of targeted therapeutics and efflux pump inhibitors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Membrane Transport Proteins/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bacterial Infections/drug therapy , Bacterial Proteins/drug effects , Bacterial Proteins/metabolism , Biological Transport/drug effects , Carrier Proteins/metabolism , Female , Membrane Transport Proteins/metabolism , Mice, Inbred BALB C , Periplasm/drug effects , Periplasm/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism
11.
J Antimicrob Chemother ; 75(4): 925-935, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31865379

ABSTRACT

BACKGROUND: Pseudomonas aeruginosa is an opportunistic bacterium that infects the airways of cystic fibrosis patients, surfaces of surgical and burn wounds, and indwelling medical devices. Patients are prone to secondary fungal infections, with Candida albicans being commonly co-isolated with P. aeruginosa. Both P. aeruginosa and C. albicans are able to form extensive biofilms on the surfaces of mucosa and medical devices. OBJECTIVES: To determine whether the presence of C. albicans enhances antibiotic tolerance of P. aeruginosa in a dual-species biofilm. METHODS: Single- and dual-species biofilms were established in microtitre plates and the survival of each species was measured following treatment with clinically relevant antibiotics. Scanning electron microscopy and confocal microscopy were used to visualize biofilm structure. RESULTS: C. albicans enhances P. aeruginosa biofilm tolerance to meropenem at the clinically relevant concentration of 5 mg/L. This effect is specific to biofilm cultures and is dependent upon C. albicans extracellular matrix polysaccharides, mannan and glucan, with C. albicans cells deficient in glycosylation structures not enhancing P. aeruginosa tolerance to meropenem. CONCLUSIONS: We propose that fungal mannan and glucan secreted into the extracellular matrix of P. aeruginosa/C. albicans dual-species biofilms play a central role in enhancing P. aeruginosa tolerance to meropenem, which has direct implications for the treatment of coinfected patients.


Subject(s)
Candida albicans , Pseudomonas aeruginosa , Biofilms , Drug Tolerance , Humans , Meropenem/pharmacology
13.
Biochem Soc Trans ; 47(3): 839-845, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31189732

ABSTRACT

The activity of any bacterial promoter is generally supposed to be set by its base sequence and the different transcription factors that bind in the local vicinity. Here, we review recent data indicating that the activity of the Escherichia coli lac operon promoter also depends upon its chromosomal location. Factors that affect promoter activity include the binding of nucleoid-associated proteins to neighbouring sequences, supercoiling and the activity of neighbouring promoters. We suggest that many bacterial promoters might be susceptible to similar position-dependent effects and we review recent data showing that the expression of mobile genes encoding antibiotic-resistance determinants is also location-dependent, both when carried on a bacterial chromosome or a conjugative plasmid.


Subject(s)
Chromosomal Position Effects , Drug Resistance, Microbial/genetics , Escherichia coli/genetics , Promoter Regions, Genetic , Chromosomes, Bacterial , DNA Transposable Elements , Lac Operon , Plasmids , Transcription, Genetic
14.
Molecules ; 24(23)2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31771095

ABSTRACT

Selenoesters and the selenium isostere of phthalic anhydride are bioactive selenium compounds with a reported promising activity in cancer, both due to their cytotoxicity and capacity to reverse multidrug resistance. Herein we evaluate the antiviral, the biofilm inhibitory, the antibacterial and the antifungal activities of these compounds. The selenoanhydride and 7 out of the 10 selenoesters were especially potent antiviral agents in Vero cells infected with herpes simplex virus-2 (HSV-2). In addition, the tested selenium derivatives showed interesting antibiofilm activity against Staphylococcus aureus and Salmonella enterica serovar Typhimurium, as well as a moderate antifungal activity in resistant strains of Candida spp. They were inactive against anaerobes, which may indicate that the mechanism of action of these derivatives depends on the presence of oxygen. The capacity to inhibit the bacterial biofilm can be of particular interest in the treatment of nosocomial infections and in the coating of surfaces of prostheses. Finally, the potent antiviral activity observed converts these selenium derivatives into promising antiviral agents with potential medical applications.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Antifungal Agents/chemical synthesis , Antiviral Agents/chemical synthesis , Phthalic Anhydrides/chemistry , Selenium Compounds/chemical synthesis , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Biofilms/drug effects , Candida/drug effects , Chlorocebus aethiops , Herpesvirus 2, Human/drug effects , Molecular Structure , Salmonella typhimurium/drug effects , Salmonella typhimurium/physiology , Selenium Compounds/chemistry , Selenium Compounds/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Vero Cells
15.
Proc Natl Acad Sci U S A ; 112(11): 3511-6, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25737552

ABSTRACT

The incidence of multidrug-resistant bacterial infections is increasing globally and the need to understand the underlying mechanisms is paramount to discover new therapeutics. The efflux pumps of Gram-negative bacteria have a broad substrate range and transport antibiotics out of the bacterium, conferring intrinsic multidrug resistance (MDR). The genomes of pre- and posttherapy MDR clinical isolates of Salmonella Typhimurium from a patient that failed antibacterial therapy and died were sequenced. In the posttherapy isolate we identified a novel G288D substitution in AcrB, the resistance-nodulation division transporter in the AcrAB-TolC tripartite MDR efflux pump system. Computational structural analysis suggested that G288D in AcrB heavily affects the structure, dynamics, and hydration properties of the distal binding pocket altering specificity for antibacterial drugs. Consistent with this hypothesis, recreation of the mutation in standard Escherichia coli and Salmonella strains showed that G288D AcrB altered substrate specificity, conferring decreased susceptibility to the fluoroquinolone antibiotic ciprofloxacin by increased efflux. At the same time, the substitution increased susceptibility to other drugs by decreased efflux. Information about drug transport is vital for the discovery of new antibacterials; the finding that one amino acid change can cause resistance to some drugs, while conferring increased susceptibility to others, could provide a basis for new drug development and treatment strategies.


Subject(s)
Amino Acid Substitution , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/genetics , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Binding Sites , Ciprofloxacin/pharmacology , Doxorubicin/chemistry , Doxorubicin/metabolism , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli Proteins/metabolism , Genetic Fitness , Genome, Bacterial , Humans , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Minocycline/pharmacology , Models, Molecular , Multidrug Resistance-Associated Proteins/metabolism , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Salmonella enterica/drug effects , Salmonella enterica/genetics , Salmonella enterica/isolation & purification , Substrate Specificity/drug effects , Water/chemistry
16.
J Antimicrob Chemother ; 70(2): 424-31, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25288678

ABSTRACT

OBJECTIVES: Enterobacteriaceae have multiple efflux pumps that confer intrinsic resistance to antibiotics. AcrB mediates clinically relevant multidrug resistance and is required for virulence and biofilm formation, making it an attractive target for the design of inhibitors. The aim of this study was to assess the viability of single transporters as a target for efflux inhibition using Salmonella Typhimurium as the model pathogen. METHODS: The expression of resistance-nodulation-division (RND) efflux pump genes in response to the inactivation of single or multiple homologues was measured using real-time RT-PCR. Phenotypes of mutants were characterized by measuring antimicrobial susceptibility, dye accumulation and the ability to cause infection in vitro. RESULTS: The expression of all RND efflux pump genes was increased when single or multiple acr genes were inactivated, suggesting a feedback mechanism that activates the transcription of homologous efflux pump genes. When two or three acr genes were inactivated, the mutants had further reduced efflux, altered susceptibility to antimicrobials (including increased susceptibility to some, but conversely and counterintuitively, decreased susceptibility to some others) and were more attenuated in the tissue culture model than mutants lacking single pumps were. CONCLUSIONS: These data indicate that it is critical to understand which pumps an inhibitor is active against and the effect of this on the expression of homologous systems. For some antimicrobials, an inhibitor with activity against multiple pumps will have a greater impact on susceptibility, but an unintended consequence of this may be decreased susceptibility to other drugs, such as aminoglycosides.


Subject(s)
Drug Resistance, Multiple, Bacterial/genetics , Gene Expression Regulation, Bacterial , Membrane Transport Proteins/genetics , Anti-Bacterial Agents/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Multiple, Bacterial/drug effects , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Gene Silencing , Genotype , Microbial Sensitivity Tests , Multigene Family , Mutation , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Virulence/genetics
17.
J Antimicrob Chemother ; 69(4): 982-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24302652

ABSTRACT

OBJECTIVES: The components of the AcrAB-TolC efflux pump function as a tripartite efflux system conferring resistance to multiple antibiotics and the individual components can also function in conjunction with other efflux pumps. This study aimed to establish whether redundancy exists between the homologous periplasmic adaptor proteins (PAPs) AcrA and AcrE and to measure the impact of this redundancy on antimicrobial resistance and the potential efficacy of inhibitor molecules. METHODS: The acrE gene was inactivated in Salmonella enterica serovar Typhimurium SL1344 and a ΔacrA mutant by insertion of the aph gene. The mutants were complemented with plasmids carrying acrA or acrE. The antimicrobial susceptibility of the mutants to various antimicrobials was determined and the accumulation or efflux of various substrates was measured. RESULTS: Inactivation of acrE alone had no phenotypic effect. However, the effect of inactivation of PAPs was additive; the acrA acrE mutant was more susceptible to certain antimicrobials and accumulated more Hoechst dye than single acrA, acrE or acrB mutants. In addition, the double mutant invaded human intestinal epithelial cells poorly. The phenotypic defects of the acrA acrE mutant were ameliorated by expression of either acrA or acrE, but the proteins exhibited some substrate specificity. CONCLUSIONS: These data show for the first time the level of redundancy between the PAPs AcrA and AcrE, and highlight the PAPs as excellent targets for inhibitor molecules that could be used to potentiate the action of clinical antimicrobials. However, the redundancy that exists between AcrA and AcrE means potential inhibitors must act on both targets to be effective.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Anti-Bacterial Agents/pharmacology , Biological Transport , Gene Deletion , Genetic Complementation Test , Humans , Microbial Sensitivity Tests
18.
J Antimicrob Chemother ; 69(3): 643-50, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24169580

ABSTRACT

OBJECTIVES: RamA regulates the AcrAB-TolC multidrug efflux system. Using Salmonella Typhimurium, we investigated the stability of RamA and its impact on antibiotic resistance. METHODS: To detect RamA, we introduced ramA::3XFLAG::aph into plasmid pACYC184 and transformed this into Salmonella Typhimurium SL1344ramA::cat and lon::aph mutants. An N-terminus-deleted mutant [pACYC184ramA(Δ2-21)::3XFLAG::aph] in which the first 20 amino acids of RamA were deleted was also constructed. To determine the abundance and half-life of FLAG-tagged RamA, we induced RamA with chlorpromazine (50 mg/L) and carried out western blotting using anti-FLAG antibody. Susceptibility to antibiotics and phenotypic characterization of the lon mutant was also carried out. RESULTS: We show that on removal of chlorpromazine, a known inducer of ramA, the abundance of RamA decreased to pre-induced levels. However, in cells lacking functional Lon, we found that the RamA protein was not degraded. We also demonstrated that the 21 amino acid residues of the RamA N-terminus are required for recognition by the Lon protease. Antimicrobial susceptibility and phenotypic tests showed that the lon mutant was more susceptible to fluoroquinolone antibiotics, was filamentous when observed by microscopy and grew poorly, but showed no difference in motility or the ability to form a biofilm. There was also no difference in the ability of the lon mutant to invade human intestinal cells (INT-407). CONCLUSIONS: In summary, we show that the ATP-dependent Lon protease plays an important role in regulating the expression of RamA and therefore multidrug resistance via AcrAB-TolC in Salmonella Typhimurium.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Bacterial , Multidrug Resistance-Associated Proteins/metabolism , Protease La/metabolism , Salmonella typhimurium/genetics , Trans-Activators/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Carrier Proteins/genetics , Drug Resistance, Bacterial , Multidrug Resistance-Associated Proteins/genetics , Protease La/genetics , Protein Stability , Salmonella typhimurium/drug effects , Trans-Activators/genetics
19.
mBio ; : e0237024, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39248573

ABSTRACT

Efflux pumps are well known to be an important mechanism for removing noxious substances such as antibiotics from bacteria. Given that many antibiotics function by accumulating inside bacteria, efflux pumps contribute to resistance. Efflux pump inactivation is a potential strategy to combat antimicrobial resistance, as bacteria would not be able to pump out antibiotics. We recently discovered that the impact of loss of efflux function is only apparent in actively growing cells. We demonstrated that the global transcriptome of Salmonella Typhimurium is drastically altered during slower growth leading to stationary-phase cells having a remodeled, less permeable envelope that prevents antibiotics entering the cell. Here, we investigated the effects of deleting the major efflux pump of Salmonella Typhimurium, AcrB, on global gene transcription across growth. We revealed that an acrB knockout entered stationary phase later than the wild-type strain SL1344 and displayed increased and prolonged expression of genes responsible for anaerobic energy metabolism. We devised a model linking efflux and membrane potential, whereby deactivation of AcrB prevents influx of protons across the inner membrane and thereby hyperpolarization. Knockout or deactivation of AcrB was demonstrated to increase membrane potential. We propose that the global transcription regulator ArcBA senses changes to the redox state of the quinol pool (linked to the membrane potential of the bacterium) and coordinates the shift from exponential to stationary phase via the key master regulators RpoS, Rsd, and Rmf. Inactivation of efflux pumps therefore influences the fundamental physiology of Salmonella, with likely impacts on multiple phenotypes.IMPORTANCEWe demonstrate for the first time that deactivation of efflux pumps brings about changes to gross bacterial physiology and metabolism. Rather than simply being a response to noxious substances, efflux pumps appear to play a key role in maintenance of membrane potential and thereby energy metabolism. This discovery suggests that efflux pump inhibition or inactivation might have unforeseen positive consequences on antibiotic activity. Given that stationary-phase bacteria are more resistant to antibiotic uptake, late entry into stationary phase would prolong antibiotic accumulation by bacteria. Furthermore, membrane hyperpolarization could result in increased generation of reactive species proposed to be important for the activity of some antibiotics. Finally, changes in gross physiology could also explain the decreased virulence of efflux mutants.

20.
mSphere ; 9(5): e0010924, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38578105

ABSTRACT

The two species that account for most cases of Acinetobacter-associated bacteremia in the United Kingdom are Acinetobacter lwoffii, often a commensal but also an emerging pathogen, and Acinetobacter baumannii, a well-known antibiotic-resistant species. While these species both cause similar types of human infection and occupy the same niche, A. lwoffii (unlike A. baumannii) has thus far remained susceptible to antibiotics. Comparatively little is known about the biology of A. lwoffii, and this is the largest study on it conducted to date, providing valuable insights into its behaviour and potential threat to human health. This study aimed to explain the antibiotic susceptibility, virulence, and fundamental biological differences between these two species. The relative susceptibility of A. lwoffii was explained as it encoded fewer antibiotic resistance and efflux pump genes than A. baumannii (9 and 30, respectively). While both species had markers of horizontal gene transfer, A. lwoffii encoded more DNA defense systems and harbored a far more restricted range of plasmids. Furthermore, A. lwoffii displayed a reduced ability to select for antibiotic resistance mutations, form biofilm, and infect both in vivo and in in vitro models of infection. This study suggests that the emerging pathogen A. lwoffii has remained susceptible to antibiotics because mechanisms exist to make it highly selective about the DNA it acquires, and we hypothesize that the fact that it only harbors a single RND system restricts the ability to select for resistance mutations. This provides valuable insights into how development of resistance can be constrained in Gram-negative bacteria. IMPORTANCE: Acinetobacter lwoffii is often a harmless commensal but is also an emerging pathogen and is the most common cause of Acinetobacter-derived bloodstream infections in England and Wales. In contrast to the well-studied and often highly drug-resistant A. baumannii, A. lwoffii has remained susceptible to antibiotics. This study explains why this organism has not evolved resistance to antibiotics. These new insights are important to understand why and how some species develop antibiotic resistance, while others do not, and could inform future novel treatment strategies.


Subject(s)
Acinetobacter Infections , Acinetobacter , Anti-Bacterial Agents , Biofilms , Microbial Sensitivity Tests , Acinetobacter/genetics , Acinetobacter/drug effects , Acinetobacter/pathogenicity , Virulence/genetics , Acinetobacter Infections/microbiology , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Biofilms/growth & development , Animals , Humans , Drug Resistance, Bacterial/genetics , Acinetobacter baumannii/genetics , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/pathogenicity , Mice , Gene Transfer, Horizontal , United Kingdom , Female , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL