Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
J Antimicrob Chemother ; 75(11): 3218-3229, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32766760

ABSTRACT

BACKGROUND: Rapid antimicrobial susceptibility testing (RAST) of bacteria causing bloodstream infections is critical for implementation of appropriate antibiotic regimens. OBJECTIVES: We have established a procedure to prepare standardized bacterial inocula for Enterobacterales-containing clinical blood cultures and assessed antimicrobial susceptibility testing (AST) data generated with the WASPLabTM automated reading system. METHODS: A total of 258 blood cultures containing Enterobacterales were examined. Bacteria were enumerated by flow cytometry using the UF-4000 system and adjusted to an inoculum of 106 cfu/mL. Disc diffusion plates were automatically streaked, incubated for 6, 8 and 18 h and imaged using the fully automated WASPLabTM system. Growth inhibition zones were compared with those obtained with inocula prepared from primary subcultures following the EUCAST standard method. Due to time-dependent variations of the inhibition zone diameters, early AST readings were interpreted using time-adjusted tentative breakpoints and areas of technical uncertainty. RESULTS AND CONCLUSIONS: Inhibition zones obtained after 18 h incubation using an inoculum of 106 cfu/mL prepared directly from blood cultures were highly concordant with those of the EUCAST standard method based on primary subcultures, with categorical agreement (CA) of 95.8%. After 6 and 8 h incubation, 89.5% and 93.0% of the isolates produced interpretable results, respectively, with CA of >98.5% and very low numbers of clinical categorization errors for both the 6 h and 8 h readings. Overall, with the standardized and automated RAST method, consistent AST data from blood cultures containing Enterobacterales can be generated after 6-8 h of incubation and subsequently confirmed by standard reading of the same plate after 18 h.


Subject(s)
Anti-Bacterial Agents , Blood Culture , Anti-Bacterial Agents/pharmacology , Bacteria , Microbial Sensitivity Tests
2.
Int J Antimicrob Agents ; 56(1): 106019, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32422315

ABSTRACT

The co-occurrence of aminoglycoside and ß-lactam resistance was assessed in 3358 consecutive Escherichia coli clinical isolates collected in 2014 in the greater Zurich area, Switzerland. Non-susceptibility to at least one of the tested aminoglycosides was observed in 470/3358 E. coli strains (14%). In strains categorized as broad-spectrum ß-lactamase (BSBL)-producers (1241/3358 isolates), extended-spectrum ß-lactamase (ESBL)-producers (262/3358) and AmpC-producers (66/3358), resistance to aminoglycoside was found in 23%, 52% and 20% of the isolates, respectively. In contrast, aminoglycoside-susceptible strains were rarely resistant to ß-lactams (33/1777, 1.9%). The genomes of 439 aminoglycoside-resistant E. coli were sequenced and aminoglycoside and ß-lactam genotypes were analysed. The most prevalent aminoglycoside resistance genes were aph(3')-Ia (133 strains, 30.3%), aac(3)-IId (100 strains, 22.8%), and aac(6')-Ib-cr (52 strains, 11.8%). The most frequent associations with ß-lactam resistance genes were aph(3')-Ia or aac(3)-IId with blaTEM-1 (94 and 72 strains, respectively), and aac(3)-IIa/aac(6')-Ib-cr with blaCTX-M-15/blaOXA-1 (23 strains). These results indicate a frequent association of aac(3) and aph(3') genotypes with BSBL production, and a frequent co-occurrence of aac(6') genes with ESBL production. The high rate of co-occurrence of aminoglycoside resistance and ß-lactamase production must be considered in combination therapy.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , beta-Lactams/pharmacology , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/drug therapy , Escherichia coli Infections/epidemiology , Genome, Bacterial/genetics , Humans , Microbial Sensitivity Tests , Switzerland/epidemiology , Whole Genome Sequencing , beta-Lactamases/biosynthesis , beta-Lactamases/genetics
3.
Am J Infect Control ; 48(9): 1090-1099, 2020 09.
Article in English | MEDLINE | ID: mdl-32311380

ABSTRACT

Antimicrobial resistance (AMR) continues to threaten global health. Although global and national AMR action plans are in place, infection prevention and control is primarily discussed in the context of health care facilities with home and everyday life settings barely addressed. As seen with the recent global SARS-CoV-2 pandemic, everyday hygiene measures can play an important role in containing the threat from infectious microorganisms. This position paper has been developed following a meeting of global experts in London, 2019. It presents evidence that home and community settings are important for infection transmission and also the acquisition and spread of AMR. It also demonstrates that the targeted hygiene approach offers a framework for maximizing protection against colonization and infections, thereby reducing antibiotic prescribing and minimizing selection pressure for the development of antibiotic resistance. If combined with the provision of clean water and sanitation, targeted hygiene can reduce the circulation of resistant bacteria in homes and communities, regardless of a country's Human Development Index (overall social and economic development). Achieving a reduction of AMR strains in health care settings requires a mirrored reduction in the community. The authors call upon national and international policy makers, health agencies, and health care professionals to further recognize the importance of targeted hygiene in the home and everyday life settings for preventing and controlling infection, in a unified quest to tackle AMR.


Subject(s)
Anti-Bacterial Agents/adverse effects , Drug Resistance, Bacterial , Global Health/standards , Hygiene/standards , Prescription Drug Overuse/prevention & control , Bacterial Infections/drug therapy , Humans , Sanitation/standards
4.
J Antimicrob Chemother ; 75(6): 1495-1505, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32155259

ABSTRACT

BACKGROUND: Disc diffusion is a reliable, accurate and cost-efficient procedure for antimicrobial susceptibility testing (AST) but requires long (18-24 h) incubation times. Reading of disc diffusion after short incubation times (6-8 h) by automated systems is feasible but should be categorized with time-adapted breakpoints to reduce errors. OBJECTIVES: This study systematically compared early readings (6 and 8 h) of disc diffusion using an automated system with that of the standard 18 h EUCAST method. Time-adapted tentative breakpoints were proposed to discriminate susceptible from resistant isolates and areas of technical uncertainty were defined to minimize the risk of errors. METHODS: A total of 1106 Enterobacterales isolates with a wide variety of resistance mechanisms and resistance profiles were included. All isolates were analysed for susceptibility to amoxicillin/clavulanic acid, ceftriaxone, cefepime, meropenem, ciprofloxacin and gentamicin using the automated WASPLabTM system. Part of the collection (515 isolates) was also analysed for susceptibility to an additional 10 antibiotics. RESULTS: Separation between WT and non-WT populations was poorer at early incubation times than following standard incubation. Editing of rapid automated AST results after 6 and 8 h incubation with time-adapted breakpoints resulted in 84.0% and 88.5% interpretable results with assignment to the resistant or susceptible category. Major error and very major error rates for the 6 h readings were only 0.4% and 0.3%, virtually identical to those of 18 h AST reading. CONCLUSIONS: Time-adapted clinical breakpoints in disc diffusion testing for Enterobacterales allow for accurate automated AST interpretation after shortened incubation times for a large number of antibiotics, with the additional possibility of subsequent confirmation after 18 h incubation.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Anti-Bacterial Agents/pharmacology , Gentamicins , Microbial Sensitivity Tests , Uncertainty
5.
FEMS Microbiol Rev ; 44(2): 171-188, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31981358

ABSTRACT

Antibiotic resistance is one of the major challenges facing modern medicine worldwide. The past few decades have witnessed rapid progress in our understanding of the multiple factors that affect the emergence and spread of antibiotic resistance at the population level and the level of the individual patient. However, the process of translating this progress into health policy and clinical practice has been slow. Here, we attempt to consolidate current knowledge about the evolution and ecology of antibiotic resistance into a roadmap for future research as well as clinical and environmental control of antibiotic resistance. At the population level, we examine emergence, transmission and dissemination of antibiotic resistance, and at the patient level, we examine adaptation involving bacterial physiology and host resilience. Finally, we describe new approaches and technologies for improving diagnosis and treatment and minimizing the spread of resistance.


Subject(s)
Bacterial Infections/microbiology , Bacterial Physiological Phenomena , Drug Resistance, Bacterial/physiology , Evolution, Molecular , Preventive Medicine/trends , Animals , Bacterial Infections/transmission , Humans
6.
Protein Sci ; 29(3): 758-767, 2020 03.
Article in English | MEDLINE | ID: mdl-31891426

ABSTRACT

Aminoglycosides were one of the first classes of broad-spectrum antibacterial drugs clinically used to effectively combat infections. The rise of resistance to these drugs, mediated by enzymatic modification, has since compromised their utility as a treatment option, prompting intensive research into the molecular function of resistance enzymes. Here, we report the crystal structure of aminoglycoside nucleotidyltransferase ANT(4')-IIb in apo and tobramycin-bound forms at a resolution of 1.6 and 2.15 Å, respectively. ANT(4')-IIb was discovered in the opportunistic pathogen Pseudomonas aeruginosa and conferred resistance to amikacin and tobramycin. Analysis of the ANT(4')-IIb structures revealed a two-domain organization featuring a mixed ß-sheet and an α-helical bundle. ANT(4')-IIb monomers form a dimer required for its enzymatic activity, as coordination of the aminoglycoside substrate relies on residues contributed by both monomers. Despite harbouring appreciable primary sequence diversity compared to previously characterized homologues, the ANT(4')-IIb structure demonstrates a surprising level of structural conservation highlighting the high plasticity of this general protein fold. Site-directed mutagenesis of active site residues and kinetic analysis provides support for a catalytic mechanism similar to those of other nucleotidyltransferases. Using the molecular insights provided into this ANT(4')-IIb-represented enzymatic group, we provide a hypothesis for the potential evolutionary origin of these aminoglycoside resistance determinants.


Subject(s)
Nucleotidyltransferases/chemistry , Pseudomonas aeruginosa/enzymology , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Conformation
7.
EBioMedicine ; 46: 184-192, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31307955

ABSTRACT

BACKGROUND: Interpretative reading of antimicrobial susceptibility test (AST) results allows inferring biochemical resistance mechanisms from resistance phenotypes. For aminoglycosides, however, correlations between resistance pathways inferred on the basis of the European Committee on Antimicrobial Susceptibility Testing (EUCAST) clinical breakpoints and expert rules versus genotypes are generally poor. This study aimed at developing and validating a decision tree based on resistance phenotypes determined by disc diffusion and based on epidemiological cut-offs (ECOFFs) to infer the corresponding resistance mechanisms in Escherichia coli. METHODS: Phenotypic antibiotic susceptibility of thirty wild-type and 458 aminoglycoside-resistant E. coli clinical isolates was determined by disc diffusion and the genomes were sequenced. Based on well-defined cut-offs, we developed a phenotype-based algorithm (Aminoglycoside Resistance Mechanism Inference Algorithm - ARMIA) to infer the biochemical mechanisms responsible for the corresponding aminoglycoside resistance phenotypes. The mechanisms inferred from susceptibility to kanamycin, tobramycin and gentamicin were analysed using ARMIA- or EUCAST-based AST interpretation and validated by whole genome sequencing (WGS) of the host bacteria. FINDINGS: ARMIA-based inference of resistance mechanisms and WGS data were congruent in 441/458 isolates (96·3%). In contrast, there was a poor correlation between resistance mechanisms inferred using EUCAST CBPs/expert rules and WGS data (418/488, 85·6%). Based on the assumption that resistance mechanisms can result in therapeutic failure, EUCAST produced 63 (12·9%) very major errors (vME), compared to only 2 (0·4%) vME with ARMIA. When used for detection and identification of resistance mechanisms, ARMIA resolved >95% vMEs generated by EUCAST-based AST interpretation. INTERPRETATION: This study demonstrates that ECOFF-based analysis of AST data of only four aminoglycosides provides accurate information on the resistance mechanisms in E. coli. Since aminoglycoside resistance mechanisms, despite having in certain cases a minimal effect on the minimal inhibitory concentration, may compromise the bactericidal activity of aminoglycosides, prompt detection of resistance mechanisms is crucial for therapy. Using ARMIA as an interpretative rule set for editing AST results allows for better predictions of in vivo activity of this drug class.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli/drug effects , Algorithms , Escherichia coli/classification , Escherichia coli/genetics , Genome, Bacterial , Genome-Wide Association Study/methods , Genomics/methods , Humans , Microbial Sensitivity Tests , Phenotype , Population Surveillance
9.
Mol Cell Proteomics ; 17(3): 442-456, 2018 03.
Article in English | MEDLINE | ID: mdl-29259044

ABSTRACT

Resistance to ß-lactams in Acinetobacter baumannii involves various mechanisms. To decipher them, whole genome sequencing (WGS) and real-time quantitative polymerase chain reaction (RT-qPCR) were complemented by mass spectrometry (MS) in selected reaction monitoring mode (SRM) in 39 clinical isolates. The targeted label-free proteomic approach enabled, in one hour and using a single method, the quantitative detection of 16 proteins associated with antibiotic resistance: eight acquired ß-lactamases (i.e. GES, NDM-1, OXA-23, OXA-24, OXA-58, PER, TEM-1, and VEB), two resident ß-lactamases (i.e. ADC and OXA-51-like) and six components of the two major efflux systems (i.e. AdeABC and AdeIJK). Results were normalized using "bacterial quantotypic peptides," i.e. peptide markers of the bacterial quantity, to obtain precise protein quantitation (on average 8.93% coefficient of variation for three biological replicates). This allowed to correlate the levels of resistance to ß-lactam with those of the production of acquired as well as resident ß-lactamases or of efflux systems. SRM detected enhanced ADC or OXA-51-like production and absence or increased efflux pump production. Precise protein quantitation was particularly valuable to detect resistance mechanisms mediated by regulated genes or by overexpression of chromosomal genes. Combination of WGS and MS, two orthogonal and complementary techniques, allows thereby interpretation of the resistance phenotypes at the molecular level.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Drug Resistance, Microbial/physiology , beta-Lactams/pharmacology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/metabolism , Bacterial Proteins/genetics , Genomics , Phenotype , Proteomics , beta-Lactamases/genetics , beta-Lactamases/metabolism
10.
ACS Infect Dis ; 3(2): 132-143, 2017 02 10.
Article in English | MEDLINE | ID: mdl-27785912

ABSTRACT

Modification of aminoglycosides by N-acetyltransferases (AACs) is one of the major mechanisms of resistance to these antibiotics in human bacterial pathogens. More than 50 enzymes belonging to the AAC(6') subfamily have been identified in Gram-negative and Gram-positive clinical isolates. Our understanding of the molecular function and evolutionary origin of these resistance enzymes remains incomplete. Here we report the structural and enzymatic characterization of AAC(6')-Ig and AAC(6')-Ih from Acinetobacter spp. The crystal structure of AAC(6')-Ig in complex with tobramycin revealed a large substrate-binding cleft remaining partially unoccupied by the substrate, which is in stark contrast with the previously characterized AAC(6')-Ib enzyme. Enzymatic analysis indicated that AAC(6')-Ig and -Ih possess a broad specificity against aminoglycosides but with significantly lower turnover rates as compared to other AAC(6') enzymes. Structure- and function-informed phylogenetic analysis of AAC(6') enzymes led to identification of at least three distinct subfamilies varying in oligomeric state, active site composition, and drug recognition mode. Our data support the concept of AAC(6') functionality originating through convergent evolution from diverse Gcn5-related-N-acetyltransferase (GNAT) ancestral enzymes, with AAC(6')-Ig and -Ih representing enzymes that may still retain ancestral nonresistance functions in the cell as provided by their particular active site properties.


Subject(s)
Acetyltransferases/chemistry , Acetyltransferases/metabolism , Acinetobacter/enzymology , Drug Resistance, Microbial , Acinetobacter/chemistry , Aminoglycosides/pharmacology , Crystallography, X-Ray , Evolution, Molecular , Models, Molecular , Phylogeny , Protein Binding , Protein Conformation , Tobramycin/pharmacology
11.
Microb Drug Resist ; 23(5): 556-562, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27854148

ABSTRACT

This study was conducted to investigate the molecular epidemiology of resistance in Acinetobacter baumannii isolates collected at a hospital in Riyadh, Saudi Arabia, from January through December 2010. Twenty-seven A. baumannii were highly resistant (MIC90 > 256 µg/ml) to ceftazidime, cefepime, and aztreonam. Imipenem resistance was seen in 24 isolates, of which 18 had an minimum inhibitory concentration (MIC) >32 µg/mL. Ciprofloxacin, gentamicin, and amikacin resistance was found in 93%, 52%, and 37% of all the isolates, respectively. Moreover, 8 (30%) isolates showed colistin resistance, and 15 (56%) were found to have MICs ≥4 µg/mL for tigecycline. The frequency of ADC, GES-1, GES-11, and GES-5 were 96.3% (n = 26), 18.5% (n = 5), 11% (n = 3), and 3.7% (n = 1), respectively. OXA-23 was found in 63% (n = 17) of the isolates; ISAba1 was found upstream of OXA-23 in 16. OXA-24/40 was detected in only one strain. Multilocus sequence typing (MLST) and pulsed-field gel electrophoresis (PFGE) analysis revealed that the 27 strains were distributed in 8 sequence types (STs) and 16 clonal pulsotypes (A-P). Five singleton STs were identified, including ST15 and ST113-ST116. The emergence of multidrug-resistant A. baumannii is becoming a major concern in Saudi Arabia. Metallo-ß-lactamases have no role in carbapenem resistance in this collection. The spread of OXA-23 in our strains occurred across different STs and pulsotypes, unlike what has been observed in many other countries. PFGE typing was more discriminatory than MLST. The high frequency of colistin and tigecycline resistance found in the isolates calls for continuous monitoring. This study describes the first identification of GES-5 conferring carbapenem resistance in A. baumannii.


Subject(s)
Acinetobacter Infections/epidemiology , Acinetobacter baumannii/genetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Hospitals , beta-Lactamases/genetics , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/classification , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Carbapenems/pharmacology , Cephalosporins/pharmacology , Ciprofloxacin/pharmacology , Clone Cells , Colistin/pharmacology , Electrophoresis, Gel, Pulsed-Field , Gene Expression , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Microbial Sensitivity Tests , Minocycline/analogs & derivatives , Minocycline/pharmacology , Molecular Epidemiology , Multilocus Sequence Typing , Saudi Arabia/epidemiology , Serogroup , Tigecycline , beta-Lactamases/metabolism
12.
J Antibiot (Tokyo) ; 70(4): 400-403, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27965516

ABSTRACT

Analysis of whole-genome sequences of 133 strains of Acinetobacter detected two genes for new types of aminoglycoside 3'-O-phosphotransferase [APH(3')], type VIII in Acinetobacter rudis and IX in A. gerneri. The enzymes were related to each other (49% identity) and to APH(3')-VI (61% and 51% identity, respectively), which is intrinsic to A. guillouiae. The cloned genes conferred kanamycin and amikacin resistance to Escherichia coli but were cryptic or expressed at low levels in the original hosts. The chromosomal location of both genes and the genetic events for acquisition of an ancestral aphA gene by A. rudis and A. gerneri, and loss by A. bereziniae were supported by the molecular phylogenetic tree of these genes. These data confirm that nonpathogenic susceptible bacterial species can be considered as potential reservoirs of resistance genes.


Subject(s)
Acinetobacter/metabolism , Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , Kanamycin Kinase/metabolism , Acinetobacter/drug effects , Amikacin/pharmacology , Chromosome Mapping , Chromosomes, Bacterial/genetics , Cloning, Molecular , DNA, Bacterial/genetics , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Kanamycin/pharmacology , Kanamycin Kinase/chemistry , Microbial Sensitivity Tests
13.
Can J Infect Dis Med Microbiol ; 2016: 4379686, 2016.
Article in English | MEDLINE | ID: mdl-27597874

ABSTRACT

Background. This study described various molecular and epidemiological characters determining antibiotic resistance patterns in Pseudomonas aeruginosa isolates. Methods. A total of 34 carbapenem-resistant P. aeruginosa clinical isolates were isolated from samples collected at a tertiary hospital in Riyadh, Saudi Arabia, from January to December 2011. Susceptibility testing, serotyping, molecular characterization of carbapenem resistance, and pulsed-field gel electrophoresis (PFGE) were performed. Results. All isolates were resistant to ceftazidime, and more than half were highly resistant (minimum inhibitory concentration (MIC) > 256 mg/L). Fifteen isolates had MIC values ≥64 mg/L for any of the carbapenems examined. Vietnamese extended-spectrum ß-lactamase (VEB-1) (n = 16/34) and oxacillinase (OXA-10) (n = 14/34) were the most prevalent extended-spectrum ß-lactamase and penicillinase, respectively. Verona imipenemase (VIM-1, VIM-2, VIM-4, VIM-11, and VIM-28) and imipenemase (IMP-7) variants were found in metallo-ß-lactamase producers. A decrease in outer membrane porin gene (oprD) expression was seen in nine isolates, and an increase in efflux pump gene (MexAB) expression was detected in five isolates. Six serotypes (O:1, O:4, O:7, O:10, O:11, and O:15) were found among the 34 isolates. The predominant serotype was O:11 (16 isolates), followed by O:15 (nine isolates). PFGE analysis of the 34 carbapenem-resistant P. aeruginosa isolates revealed 14 different pulsotypes. Conclusions. These results revealed diverse mechanisms conferring carbapenem resistance to P. aeruginosa isolates from Saudi Arabia.

14.
mBio ; 7(3)2016 05 31.
Article in English | MEDLINE | ID: mdl-27247231

ABSTRACT

UNLABELLED: Overexpression of chromosomal resistance-nodulation-cell division (RND)-type efflux systems with broad substrate specificity contributes to multidrug resistance (MDR) in Acinetobacter baumannii We have shown that modulation of expression of the structural genes for the efflux systems AdeABC and AdeIJK confers MDR and results in numerous alterations of membrane-associated cellular functions, in particular biofilm formation. However, the contribution of these RND pumps to cell fitness and virulence has not yet been studied. The biological cost of an antibiotic resistance mechanism is a key parameter in determining its stability and dissemination. From an entirely sequenced susceptible clinical isolate, we have generated a set of isogenic derivatives having single point mutations resulting in overexpression of each efflux system or with every pump deleted by allelic replacement. We found that overproduction of the pumps results in a significant decrease in fitness of the bacterial host when measured by competition experiments in vitro Fitness and virulence were also evaluated in vivo both in systemic and pulmonary infection models in immunocompetent mice. A diminished competitiveness of the AdeABC-overexpressing mutant was observed only after intraperitoneal inoculation, but not after intranasal inoculation, the latter mimicking the most frequent type of human A. baumannii infection. However, in mice infected intranasally, this mutant was more virulent and stimulated an enhanced neutrophil activation in the lungs. Altogether, these data account for the observation that adeABC overexpression is common in MDR A. baumannii frequently found in ventilator-associated pneumonia. IMPORTANCE: Overproduction of the RND AdeABC efflux system is observed with a high incidence in multidrug-resistant Acinetobacter baumannii and results in increased resistance to several antibiotics of choice for the treatment of infections caused by this nosocomial pathogen. It was therefore important to study the biological cost of the overexpression of the adeABC structural operon which is normally tightly regulated. Fitness diminution of an overexpressing mutant detected in vitro and in vivo in a model that mimics sepsis was not observed in a pulmonary infection model in which the mutant was more virulent. This points out that increased virulence can occur independently from prolonged persistence in the infected organ and can account for the elevated incidence of this resistance mechanism in clinical isolates. The study also indicates that transposon libraries will identify only virulence genes that are expressed under physiological conditions but not those that are tightly regulated.


Subject(s)
Acinetobacter baumannii/genetics , Acinetobacter baumannii/pathogenicity , Drug Resistance, Multiple, Bacterial/genetics , Genetic Fitness , Membrane Transport Proteins/genetics , Acinetobacter Infections/microbiology , Acinetobacter baumannii/growth & development , Acinetobacter baumannii/metabolism , Animals , Bacterial Proteins/genetics , Biofilms/growth & development , Cell Division/genetics , Humans , Immunocompetence , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Neutrophils/immunology , Virulence
15.
J Antimicrob Chemother ; 71(3): 601-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26645270

ABSTRACT

OBJECTIVES: The aac(6')-Ih gene encoding aminoglycoside 6'-N-acetyltransferase type I subtype h [AAC(6')-Ih] is plasmid-borne in Acinetobacter baumannii where it confers high-level amikacin resistance, but its origin remains unknown. We searched for the gene in the genomes of a collection of 133 Acinetobacter spp. and studied its species specificity, expression and dissemination. METHODS: Gene copy number was determined by quantitative PCR, expression by quantitative RT-PCR, MIC by microdilution and transfer by plasmid mobilization. RESULTS: The aac(6')-Ih gene was present in the chromosome of the two Acinetobacter gyllenbergii of the collection and was detected in all seven A. gyllenbergii clinical isolates. They had indistinguishable flanking regions indicating that the gene was intrinsic to this species. A. baumannii PIS Aba23 promoters were provided by insertion of ISAba23, which disrupted the Pnative promoter in A. gyllenbergii. Both types of promoters were similarly potent in Escherichia coli and A. baumannii. Aminoglycoside MICs for A. baumannii harbouring pIP1858 were higher than for A. gyllenbergii due to gene dosage. The non-self-transferable plasmid could be mobilized to other A. baumannii cells by the broad host range plasmid RP4. CONCLUSIONS: We have found the origin of aac(6')-Ih in A. gyllenbergii, a species isolated, although rarely, in humans, and documented that dissemination of this gene is restricted to the Acinetobacter genus.


Subject(s)
Acetyltransferases/genetics , Acetyltransferases/metabolism , Acinetobacter baumannii/classification , Acinetobacter baumannii/enzymology , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , DNA Transposable Elements , Gene Dosage , Gene Transfer, Horizontal , Humans , Microbial Sensitivity Tests , Plasmids/analysis , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction
16.
mBio ; 6(4): e00806, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26265719

ABSTRACT

UNLABELLED: Vancomycin resistance in Gram-positive bacteria results from the replacement of the D-alanyl-D-alanine target of peptidoglycan precursors with D-alanyl-D-lactate or D-alanyl-D-serine (D-Ala-D-Ser), to which vancomycin has low binding affinity. VanT is one of the proteins required for the production of D-Ala-D-Ser-terminating precursors by converting L-Ser to D-Ser. VanT is composed of two domains, an N-terminal membrane-bound domain, likely involved in L-Ser uptake, and a C-terminal cytoplasmic catalytic domain which is related to bacterial alanine racemases. To gain insight into the molecular function of VanT, the crystal structure of the catalytic domain of VanTG from VanG-type resistant Enterococcus faecalis BM4518 was determined. The structure showed significant similarity to type III pyridoxal 5'-phosphate (PLP)-dependent alanine racemases, which are essential for peptidoglycan synthesis. Comparative structural analysis between VanTG and alanine racemases as well as site-directed mutagenesis identified three specific active site positions centered around Asn696 which are responsible for the L-amino acid specificity. This analysis also suggested that VanT racemases evolved from regular alanine racemases by acquiring additional selectivity toward serine while preserving that for alanine. The 4-fold-lower relative catalytic efficiency of VanTG against L-Ser versus L-Ala implied that this enzyme relies on its membrane-bound domain for L-Ser transport to increase the overall rate of d-Ser production. These findings illustrate how vancomycin pressure selected for molecular adaptation of a housekeeping enzyme to a bifunctional enzyme to allow for peptidoglycan remodeling, a strategy increasingly observed in antibiotic-resistant bacteria. IMPORTANCE: Vancomycin is one of the drugs of last resort against Gram-positive antibiotic-resistant pathogens. However, bacteria have evolved a sophisticated mechanism which remodels the drug target, the D-alanine ending precursors in cell wall synthesis, into precursors terminating with D-lactate or D-serine, to which vancomycin has less affinity. D-Ser is synthesized by VanT serine racemase, which has two unusual characteristics: (i) it is one of the few serine racemases identified in bacteria and (ii) it contains a membrane-bound domain involved in L-Ser uptake. The structure of the catalytic domain of VanTG showed high similarity to alanine racemases, and we identified three specific active site substitutions responsible for L-Ser specificity. The data provide the molecular basis for VanT evolution to a bifunctional enzyme coordinating both transport and racemization. Our findings also illustrate the evolution of the essential alanine racemase into a vancomycin resistance enzyme in response to antibiotic pressure.


Subject(s)
Enterococcus faecalis/enzymology , Racemases and Epimerases/chemistry , Catalytic Domain , Crystallography, X-Ray , Enterococcus faecalis/genetics , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Conformation , Racemases and Epimerases/genetics , Substrate Specificity
17.
Antimicrob Agents Chemother ; 59(9): 5647-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26149994

ABSTRACT

Corynebacterium striatum BM4687 was resistant to gentamicin and tobramycin but susceptible to kanamycin A and amikacin, a phenotype distinct among Gram-positive bacteria. Analysis of the entire genome of this strain did not detect any genes for known aminoglycoside resistance enzymes. Yet, annotation of the coding sequences identified 12 putative acetyltransferases or GCN5-related N-acetyltransferases. A total of 11 of these coding sequences were also present in the genomes of other Corynebacterium spp. The 12th coding sequence had 55 to 60% amino acid identity with acetyltransferases in Actinomycetales. The gene was cloned in Escherichia coli, where it conferred resistance to aminoglycosides by acetylation. The protein was purified to homogeneity, and its steady-state kinetic parameters were determined for dibekacin and kanamycin B. The product of the turnover of dibekacin was purified, and its structure was elucidated by high-field nuclear magnetic resonance (NMR), indicating transfer of the acetyl group to the amine at the C-3 position. Due to the unique profile of the reaction, it was designated aminoglycoside 3-N-acetyltransferase type XI.


Subject(s)
Acetyltransferases/metabolism , Corynebacterium/enzymology , Anti-Bacterial Agents/pharmacology , Corynebacterium/drug effects , Dibekacin/pharmacology , Kanamycin/pharmacology , Microbial Sensitivity Tests , Molecular Structure
18.
PLoS Genet ; 11(4): e1005170, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25898178

ABSTRACT

Enterococcus faecalis BM4518 is resistant to vancomycin by synthesis of peptidoglycan precursors ending in D-alanyl-D-serine. In the chromosomal vanG locus, transcription of the resistance genes from the PYG resistance promoter is inducible and, upstream from these genes, there is an unusual three-component regulatory system encoded by the vanURS(G) operon from the P(UG) regulatory promoter. In contrast to the other van operons in enterococci, the vanG operon possesses the additional vanU(G) gene which encodes a transcriptional regulator whose role remains unknown. We show by DNase I footprinting, RT-qPCR, and reporter proteins activities that VanU(G), but not VanR(G), binds to P(UG) and negatively autoregulates the vanURS(G) operon and that it also represses PYG where it overlaps with VanR(G) for binding. In clinical isolate BM4518, the transcription level of the resistance genes was dependent on vancomycin concentration whereas, in a ΔvanUG mutant, resistance was expressed at a maximum level even at low concentrations of the inducer. The binding competition between VanU(G) and VanR(G) on the P(YG) resistance promoter allowed rheostatic activation of the resistance operon depending likely on the level of VanR(G) phosphorylation by the VanS(G) sensor. In addition, there was cross-talk between VanS(G) and VanR'(G), a VanR(G) homolog, encoded elsewhere in the chromosome indicating a sophisticated and subtle regulation of vancomycin resistance expression by a complex two-component system.


Subject(s)
Bacterial Proteins/genetics , Drug Resistance, Microbial/genetics , Enterococcus faecalis/genetics , Gram-Positive Bacterial Infections/genetics , Transcription Factors/genetics , Vancomycin Resistance/genetics , DNA Footprinting , Enterococcus faecalis/pathogenicity , Gene Expression Regulation, Bacterial , Gram-Positive Bacterial Infections/microbiology , Humans , Operon/genetics , Peptidoglycan/genetics , Transcription, Genetic , Vancomycin/therapeutic use
19.
J Mol Biol ; 427(12): 2229-43, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25900373

ABSTRACT

One of the main mechanisms of resistance to lincosamide and aminoglycoside antibiotics is their inactivation by O-nucleotidylyltransferases (NTases). Significant sequence variation of lincomycin nucleotidylyltransferase (Lnu) and aminoglycoside nucleotidylyltransferase (ANT) enzymes plus lack of detailed information about the molecular basis for specificity of these enzymes toward chemically distinct antibiotic scaffolds hinders development of a general strategy to curb this resistance mechanism. We conducted an extensive sequence analysis identifying 129 putative antibiotic NTases constituting six distinct subfamilies represented by Lnu(A), Lnu(B), Lnu(C), Lnu(D), Lnu(F)/(G) plus ANT(2") enzymes. Since only the Lnu(B) enzyme has been previously studied in detail, we biochemically characterized the Lnu(A) and Lnu(D) enzymes, with the former representing the most sequence distinct Lnu ortholog. We also determined the crystal structure of the Lnu(A) enzyme in complex with a lincosamide. These data suggested that, while sharing the N-terminal nucleotidylyltransferase domain, the groups of antibiotic NTases feature structurally distinct C-terminal domains (CTDs) adapted to accommodate antibiotics. Comparative structural analysis among antibiotic NTases rationalized their specificity toward lincosamides versus aminoglycosides through active-site plasticity, which allows retention of general catalytic activity while accepting alterations at multiple, specific positions contributed by both domains. Based on this structural analysis, we suggest that antibiotic NTases evolved from an ancestral nucleotidylyltransferase along independent paths according to the identified groups, characterized by structural changes in the active site and recruitment of structurally diverse CTDs. These data show the complexity of enzyme-driven antibiotic resistance and provide a basis for broadly active inhibitors by identifying the key unifying features of antibiotic NTases.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Drug Resistance, Bacterial , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Catalytic Domain , Cluster Analysis , Crystallography, X-Ray , Lincosamides/chemistry , Lincosamides/metabolism , Molecular Sequence Data , Nucleotidyltransferases/genetics , Phylogeny , Protein Binding , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
20.
J Antimicrob Chemother ; 70(7): 1942-5, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25802283

ABSTRACT

OBJECTIVES: The objective was to study a new vanG-type locus in Clostridium argentinense vanGCar and to determine its impact on glycopeptide susceptibility of the host. METHODS: The whole genome of C. argentinense NCIB 10714 was sequenced using Illumina single-reads sequencing technology. The presence of vanGCar in seven C. argentinense strains was tested by PCR and its expression was tested by quantitative RT-PCR (qRT-PCR). Glycopeptide susceptibility was determined by the Etest procedure. RESULTS: The vanGCar locus contained four genes encoding a carboxypeptidase, a d-alanine:d-serine ligase, a serine transporter and a serine racemase, and was present in the seven C. argentinense studied. An AraC-type transcriptional regulator was found upstream from the genes. C. argentinense NCIB 10714 was susceptible to vancomycin and to teicoplanin. qRT-PCR experiments revealed that vanGCar was not expressed without or with induction by a subinhibitory concentration of vancomycin. CONCLUSIONS: The new vanGCar locus was cryptic in C. argentinense and intrinsic to this species. Emergence of vancomycin resistance in C. argentinense due to decryptification of the vanGCar gene cluster could occur.


Subject(s)
Clostridium/drug effects , Clostridium/genetics , Genes, Bacterial , Vancomycin Resistance , Anti-Bacterial Agents/pharmacology , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Disk Diffusion Antimicrobial Tests , Gene Order , Genetic Loci , Genome, Bacterial , Glycopeptides/pharmacology , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...