Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
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
2.
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
3.
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
4.
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
5.
mBio ; 6(2)2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25805730

ABSTRACT

UNLABELLED: Acinetobacter baumannii is a nosocomial pathogen of increasing importance due to its multiple resistance to antibiotics and ability to survive in the hospital environment linked to its capacity to form biofilms. To fully characterize the contribution of AdeABC, AdeFGH, and AdeIJK resistance-nodulation-cell division (RND)-type efflux systems to acquired and intrinsic resistance, we constructed, from an entirely sequenced susceptible A. baumannii strain, a set of isogenic mutants overexpressing each system following introduction of a point mutation in their cognate regulator or a deletion for the pump by allelic replacement. Pairwise comparison of every derivative with the parental strain indicated that AdeABC and AdeFGH are tightly regulated and contribute to acquisition of antibiotic resistance when overproduced. AdeABC had a broad substrate range, including ß-lactams, fluoroquinolones, tetracyclines-tigecycline, macrolides-lincosamides, and chloramphenicol, and conferred clinical resistance to aminoglycosides. Importantly, when combined with enzymatic resistance to carbapenems and aminoglycosides, this pump contributed in a synergistic fashion to the level of resistance of the host. In contrast, AdeIJK was expressed constitutively and was responsible for intrinsic resistance to the same major drug classes as AdeABC as well as antifolates and fusidic acid. Surprisingly, overproduction of AdeABC and AdeIJK altered bacterial membrane composition, resulting in decreased biofilm formation but not motility. Natural transformation and plasmid transfer were diminished in recipients overproducing AdeABC. It thus appears that alteration in the expression of efflux systems leads to multiple changes in the relationship between the host and its environment, in addition to antibiotic resistance. IMPORTANCE: Increased expression of chromosomal genes for RND-type efflux systems plays a major role in bacterial multidrug resistance. Acinetobacter baumannii has recently emerged as an important human pathogen responsible for epidemics of hospital-acquired infections. Besides its remarkable ability to horizontally acquire resistance determinants, it has a broad intrinsic resistance due to low membrane permeability, endogenous resistance genes, and antibiotic efflux. The study of isogenic mutants from a susceptible A. baumannii clinical isolate overproducing or deleted for each of the three major RND-type pumps demonstrated their major contribution to intrinsic resistance and to the synergism between overproduction of an efflux system and acquisition of a resistance gene. We have also shown that modulation of expression of the structural genes for the efflux systems results in numerous alterations in membrane-associated cellular functions, in particular, in a decrease in biofilm formation and resistance gene acquisition.


Subject(s)
Acinetobacter baumannii/drug effects , Acinetobacter baumannii/metabolism , Anti-Bacterial Agents/metabolism , Biofilms/growth & development , Drug Resistance, Bacterial , Membrane Transport Proteins/metabolism , Acinetobacter baumannii/genetics , Acinetobacter baumannii/physiology , Biological Transport, Active , Genome, Bacterial , Humans , Membrane Transport Proteins/genetics , Molecular Sequence Data , Sequence Analysis, DNA , Substrate Specificity
6.
Genome Biol Evol ; 6(10): 2866-82, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25313016

ABSTRACT

Bacterial genomics has greatly expanded our understanding of microdiversification patterns within a species, but analyses at higher taxonomical levels are necessary to understand and predict the independent rise of pathogens in a genus. We have sampled, sequenced, and assessed the diversity of genomes of validly named and tentative species of the Acinetobacter genus, a clade including major nosocomial pathogens and biotechnologically important species. We inferred a robust global phylogeny and delimited several new putative species. The genus is very ancient and extremely diverse: Genomes of highly divergent species share more orthologs than certain strains within a species. We systematically characterized elements and mechanisms driving genome diversification, such as conjugative elements, insertion sequences, and natural transformation. We found many error-prone polymerases that may play a role in resistance to toxins, antibiotics, and in the generation of genetic variation. Surprisingly, temperate phages, poorly studied in Acinetobacter, were found to account for a significant fraction of most genomes. Accordingly, many genomes encode clustered regularly interspaced short palindromic repeats (CRISPR)-Cas systems with some of the largest CRISPR-arrays found so far in bacteria. Integrons are strongly overrepresented in Acinetobacter baumannii, which correlates with its frequent resistance to antibiotics. Our data suggest that A. baumannii arose from an ancient population bottleneck followed by population expansion under strong purifying selection. The outstanding diversification of the species occurred largely by horizontal transfer, including some allelic recombination, at specific hotspots preferentially located close to the replication terminus. Our work sets a quantitative basis to understand the diversification of Acinetobacter into emerging resistant and versatile pathogens.


Subject(s)
Acinetobacter/genetics , Genome, Bacterial/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genomics/methods , Interspersed Repetitive Sequences/genetics , Phylogeny
7.
mBio ; 5(5): e01972-14, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25336457

ABSTRACT

The amikacin resistance gene aphA6 was first detected in the nosocomial pathogen Acinetobacter baumannii and subsequently in other genera. Analysis of 133 whole-genome sequences covering the taxonomic diversity of Acinetobacter spp. detected aphA6 in the chromosome of 2 isolates of A. guillouiae, which is an environmental species, 1 of 8 A. parvus isolates, and 5 of 34 A. baumannii isolates. The gene was also present in 29 out of 36 A. guillouiae isolates screened by PCR, indicating that it is ancestral to this species. The Pnative promoter for aphA6 in A. guillouiae and A. parvus was replaced in A. baumannii by PaphA6, which was generated by use of the insertion sequence ISAba125, which brought a -35 sequence. Study of promoter strength in Escherichia coli and A. baumannii indicated that PaphA6 was four times more potent than Pnative. There was a good correlation between aminoglycoside MICs and aphA6 transcription in A. guillouiae isolates that remained susceptible to amikacin. The marked topology differences of the phylogenetic trees of aphA6 and of the hosts strongly support its recent direct transfer within Acinetobacter spp. and also to evolutionarily remote bacterial genera. Concomitant expression of aphA6 must have occurred because, contrary to the donors, it can confer resistance to the new hosts. Mobilization and expression of aphA6 via composite transposons and the upstream IS-generating hybrid PaphA6, followed by conjugation, seems the most plausible mechanism. This is in agreement with the observation that, in the recipients, aphA6 is carried by conjugative plasmids and flanked by IS that are common in Acinetobacter spp. Our data indicate that resistance genes can also be found in susceptible environmental bacteria. Importance: We speculated that the aphA6 gene for an enzyme that confers resistance to amikacin, the most active aminoglycoside for the treatment of nosocomial infections due to Acinetobacter spp., originated in this genus before disseminating to phylogenetically distant genera pathogenic for humans. Using a combination of whole-genome sequencing of a collection of Acinetobacter spp. covering the breadth of the known taxonomic diversity of the genus, gene cloning, detailed promoter analysis, study of heterologous gene expression, and comparative analysis of the phylogenetic trees of aphA6 and of the bacterial hosts, we found that aphA6 originated in Acinetobacter guillouiae, an amikacin-susceptible environmental species. The gene conferred, upon mobilization, high-level resistance to the new hosts. This work stresses that nonpathogenic bacteria can act as reservoirs of resistance determinants, and it provides an example of the use of a genomic library to study the origin and dissemination of an antibiotic resistance gene to human pathogens.


Subject(s)
Acinetobacter/enzymology , Acinetobacter/genetics , Kanamycin Kinase/genetics , Acinetobacter/drug effects , Acinetobacter/isolation & purification , Amino Acid Sequence , Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Base Sequence , Cluster Analysis , Conjugation, Genetic , Drug Resistance, Bacterial , Environmental Microbiology , Escherichia coli/enzymology , Escherichia coli/genetics , Evolution, Molecular , Gene Transfer, Horizontal , Interspersed Repetitive Sequences , Microbial Sensitivity Tests , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic , Sequence Homology
8.
Vaccine ; 32(32): 4051-8, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24886957

ABSTRACT

Protein-subunit vaccines as boosting strategies against tuberculosis (TB) infection are currently in the pipeline of TB vaccine research. Their main limitation is represented by their poor immunogenicity, which makes it necessary to couple protein-subunits with adjuvant molecules. In this study, we employed replication-deficient invasive Escherichia coli strains to deliver Mycobacterium tuberculosis proteins to the cytoplasm of non-phagocytic eukaryotic cells using various priming and prime-boosting vaccination protocols. Our results demonstrate that intranasal administration of invasive E. coli expressing the M. tuberculosis protective antigen MPT64 to mice primed with a recombinant BCG strain over-expressing MPT64 on its surface, decrease bacterial burden in mice spleens. Our data suggest that replication-deficient invasive E. coli may represent a suitable platform for BCG/rBCG priming followed by homologous-boosting immunization strategies.


Subject(s)
Antigens, Bacterial/immunology , Escherichia coli , Immunization, Secondary/methods , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Administration, Intranasal , Animals , BCG Vaccine/immunology , Bacterial Load , Female , HeLa Cells , Humans , Mice, Inbred C57BL , Mycobacterium tuberculosis , Recombinant Proteins/immunology , Spleen/microbiology
9.
mBio ; 5(2): e00915, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24757213

ABSTRACT

Gene amplification is believed to play an important role in antibiotic resistance but has been rarely documented in clinical settings because of its unstable nature. We report a rise in MICs from 0.5 to 16 µg/ml in successive Acinetobacter baumannii isolated over 4 days from a patient being treated with tobramycin for an infection by multidrug-resistant A. baumannii, resulting in therapeutic failure. Isolates were characterized by whole-genome sequencing, real-time and reverse transcriptase PCR, and growth assays to determine the mechanism of tobramycin resistance and its fitness cost. Tobramycin resistance was associated with two amplification events of different chromosomal fragments containing the aphA1 aminoglycoside resistance gene part of transposon Tn6020. The first amplification event involved low amplification (6 to 10 copies) of a large DNA fragment that was unstable and conferred tobramycin MICs of ≤ 8 µg/ml. The second event involved moderate (10 to 30 copies) or high (40 to 110 copies) amplification of Tn6020. High copy numbers were associated with tobramycin MICs of 16 µg/ml, impaired fitness, and genetic instability, whereas lower copy numbers resulted in tobramycin MICs of ≤8 µg/ml and no fitness cost and were stably maintained in vitro. Exposure in vitro to tobramycin of the initial susceptible isolate and of the A. baumannii AB0057 reference strain led to similar aphA1 amplifications and elevated tobramycin MICs. To the best of our knowledge, this is the first report of in vivo development of antibiotic resistance secondary to gene amplifications resulting in therapy failure. IMPORTANCE A combination of whole-genome sequencing and mapping were used to detect an antibiotic resistance mechanism, gene amplification, which has been presumed for a long time to be of major importance but has rarely been reported in clinical settings because of its unstable nature. Two gene amplification events in a patient with an Acinetobacter baumannii infection treated with tobramycin were identified. One gene amplification event led to high levels of resistance and was rapidly reversible, while the second event led to low and more stable resistance since it incurred low fitness cost on the host. Gene amplification, with an associated rise in tobramycin MICs, could be readily reproduced in vitro from initially susceptible strains exposed to increasing concentrations of tobramycin, suggesting that gene amplification in A. baumannii may be a more common mechanism than currently believed. This report underscores the importance of rapid molecular techniques for surveillance of drug resistance.


Subject(s)
Acid Phosphatase/genetics , Acinetobacter Infections/drug therapy , Acinetobacter baumannii/enzymology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Bacterial , Gene Amplification , Tobramycin/therapeutic use , Acid Phosphatase/metabolism , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Acinetobacter baumannii/isolation & purification , DNA Transposable Elements , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Gene Dosage , Genome, Bacterial , Humans , Male , Microbial Sensitivity Tests , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Treatment Failure , Young Adult
10.
RNA ; 20(3): 382-91, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24398977

ABSTRACT

In Gram-negative bacteria, acquired 16S rRNA methyltransferases ArmA and NpmA confer high-level resistance to all clinically useful aminoglycosides by modifying, respectively, G1405 and A1408 in the A-site. These enzymes must coexist with several endogenous methyltransferases that are essential for fine-tuning of the decoding center, such as RsmH and RsmI in Escherichia coli, which methylate C1402 and RsmF C1407. The resistance methyltransferases have a contrasting distribution--ArmA has spread worldwide, whereas a single clinical isolate producing NpmA has been reported. The rate of dissemination of resistance depends on the fitness cost associated with its expression. We have compared ArmA and NpmA in isogenic Escherichia coli harboring the corresponding structural genes and their inactive point mutants cloned under the control of their native constitutive promoter in the stable plasmid pGB2. Growth rate determination and competition experiments showed that ArmA had a fitness cost due to methylation of G1405, whereas NpmA conferred only a slight disadvantage to the host due to production of the enzyme. MALDI MS indicated that ArmA impeded one of the methylations at C1402 by RsmI, and not at C1407 as previously proposed, whereas NpmA blocked the activity of RsmF at C1407. A dual luciferase assay showed that methylation at G1405 and A1408 and lack of methylation at C1407 affect translation accuracy. These results indicate that resistance methyltransferases impair endogenous methylation with different consequences on cell fitness.


Subject(s)
Aminoglycosides/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli Proteins/genetics , Genetic Fitness , Methylation/drug effects , Methyltransferases/metabolism , Protein Biosynthesis/drug effects , RNA, Ribosomal, 16S/genetics , Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Methyltransferases/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Binding , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
J Infect Dis ; 208(7): 1142-51, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23812239

ABSTRACT

BACKGROUND: Colistin resistance is of concern since it is increasingly needed to treat infections caused by bacteria resistant to all other antibiotics and has been associated with poorer outcomes. Longitudinal data from in vivo series are sparse. METHODS: Under a quality-improvement directive to intensify infection-control measures, extremely drug-resistant (XDR) bacteria undergo phenotypic and molecular analysis. RESULTS: Twenty-eight XDR Acinetobacter baumannii isolates were longitudinally recovered during colistin therapy. Fourteen were susceptible to colistin, and 14 were resistant to colistin. Acquisition of colistin resistance did not alter resistance to other antibiotics. Isolates had low minimum inhibitory concentrations of an investigational aminoglycoside, belonged to multi-locus sequence type 94, were indistinguishable by pulsed-field gel electrophoresis and optical mapping, and harbored a novel pmrC1A1B allele. Colistin resistance was associated with point mutations in the pmrA1 and/or pmrB genes. Additional pmrC homologs, designated eptA-1 and eptA-2, were at distant locations from the operon. Compared with colistin-susceptible isolates, colistin-resistant isolates displayed significantly enhanced expression of pmrC1A1B, eptA-1, and eptA-2; lower growth rates; and lowered fitness. Phylogenetic analysis suggested that colistin resistance emerged from a single progenitor colistin-susceptible isolate. CONCLUSIONS: We provide insights into the in vivo evolution of colistin resistance in a series of XDR A. baumannii isolates recovered during therapy of infections and emphasize the importance of antibiotic stewardship and surveillance.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Colistin/therapeutic use , Drug Resistance, Bacterial , Transcription Factors/genetics , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/isolation & purification , Adult , Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Genotype , Humans , Longitudinal Studies , Microbial Sensitivity Tests , Molecular Typing , Operon , Point Mutation , Wound Infection/drug therapy
12.
Antimicrob Agents Chemother ; 57(7): 2989-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23587960

ABSTRACT

Increased expression of chromosomal genes for resistance-nodulation-cell division (RND)-type efflux systems plays a major role in the multidrug resistance (MDR) of Acinetobacter baumannii. However, the relative contributions of the three most prevalent pumps, AdeABC, AdeFGH, and AdeIJK, have not been evaluated in clinical settings. We have screened 14 MDR clinical isolates shown to be distinct on the basis of multilocus sequence typing (MLST) and pulsed-field gel electrophoresis (PFGE) for the presence and overexpression of the three Ade efflux systems and analyzed the sequences of the regulators AdeRS, a two-component system, for AdeABC and AdeL, a LysR-type regulator, for AdeFGH. Gene adeB was detected in 13 of 14 isolates, and adeG and the intrinsic adeJ gene were detected in all strains. Significant overexpression of adeB was observed in 10 strains, whereas only 7 had moderately increased levels of expression of AdeFGH, and none overexpressed AdeIJK. Thirteen strains had reduced susceptibility to tigecycline, but there was no correlation between tigecycline MICs and the levels of AdeABC expression, suggesting the presence of other mechanisms for tigecycline resistance. No mutations were found in the highly conserved LysR regulator of the nine strains expressing AdeFGH. In contrast, functional mutations were found in conserved domains of AdeRS in all the strains that overexpressed AdeABC with two mutational hot spots, one in AdeS near histidine 149 suggesting convergent evolution and the other in the DNA binding domain of AdeR compatible with horizontal gene transfer. This report outlines the high incidence of AdeABC efflux pump overexpression in MDR A. baumannii as a result of a variety of single mutations in the corresponding two-component regulatory system.


Subject(s)
Acinetobacter baumannii/genetics , Bacterial Proteins/biosynthesis , Drug Resistance, Multiple, Bacterial/genetics , Membrane Transport Proteins/biosynthesis , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Acinetobacter baumannii/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Electrophoresis, Gel, Pulsed-Field , Humans , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Minocycline/analogs & derivatives , Minocycline/pharmacology , Multilocus Sequence Typing , Mutation , Tigecycline , Transcription Factors/genetics
13.
Bioeng Bugs ; 3(2): 86-92, 2012.
Article in English | MEDLINE | ID: mdl-22095052

ABSTRACT

Efficient delivery of large intact vectors into mammalian cells remains problematical. Here we evaluate delivery by bacterial invasion of two large BACs of more than 150 kb in size into various cells. First, we determined the effect of several drugs on bacterial delivery of a small plasmid into different cell lines. Most drugs tested resulted in a marginal increase of the overall efficiency of delivery in only some cell lines, except the lysosomotropic drug chloroquine, which was found to increase the efficiency of delivery by 6-fold in B16F10 cells. Bacterial invasion was found to be significantly advantageous compared with lipofection in delivering large intact BACs into mouse cells, resulting in 100% of clones containing intact DNA. Furthermore, evaluation of expression of the human hypoxanthine phosphoribosyltransferase (HPRT) gene from its genomic locus, which was present in one of the BACs, showed that single copy integrations of the HPRT-containing BAC had occurred in mouse B16F10 cells and that expression of HPRT from each human copy was 0.33 times as much as from each endogenous mouse copy. These data provide new evidence that bacterial delivery is a convenient and efficient method to transfer large intact therapeutic genes into mammalian cells.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Gene Transfer Techniques , Genetic Vectors , Animals , Cell Line , Escherichia coli/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Mice , Plasmids
14.
Proc Natl Acad Sci U S A ; 107(39): 16964-9, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20833818

ABSTRACT

Inducible vancomycin resistance in enterococci is due to a sophisticated mechanism that combines synthesis of cell wall peptidoglycan precursors with low affinity for glycopeptides and elimination of the normal target precursors. Although this dual mechanism, which involves seven genes organized in two operons, is predicted to have a high fitness cost, resistant enterococci have disseminated worldwide. We have evaluated the biological cost of VanB-type resistance due to acquisition of conjugative transposon Tn1549 in Enterococcus faecium and Enterococcus faecalis. Because fitness was dependent on the integration site of Tn1549, an isogenic set of E. faecalis was constructed to determine the cost of inducible or constitutive expression of resistance or of carriage of Tn1549. A luciferase gene was inserted in the integrase gene of the transposon to allow differential quantification of the strains in cocultures and in the digestive tract of gnotobiotic mice. Both in vitro and in vivo, carriage of inactivated or inducible Tn1549 had no cost for the host in the absence of induction by vancomycin. In contrast, induced or constitutively resistant strains not only had reduced fitness but were severely impaired in colonization ability and dissemination among mice. These data indicate that tight regulation of resistance expression drastically reduces the biological cost associated with vancomycin resistance in Enterococcus spp. and accounts for the widespread dissemination of these strains. Our findings are in agreement with the observation that regulation of expression is common in horizontally acquired resistance and represents an efficient evolutionary pathway for resistance determinants to become selectively neutral.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , DNA Transposable Elements , Enterococcus faecalis/genetics , Enterococcus faecium/genetics , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Animals , Enterococcus faecalis/drug effects , Enterococcus faecium/drug effects , Mice , Mice, Inbred C3H , Transcription, Genetic
15.
Antimicrob Agents Chemother ; 53(6): 2354-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19332680

ABSTRACT

We have quantified the biological cost of VanA-type glycopeptide resistance due to the acquisition of the resistance operon by methicillin-resistant Staphylococcus aureus (MRSA) from Enterococcus sp. Exponential growths of recipient strain HIP11713, its transconjugant VRSA-1, VRSA-5, and VRSA-6 were compared in the absence or, except for HIP11713, in the presence of vancomycin. Induction of resistance was performed by adding vancomycin in both the preculture and the culture or the culture at only 1/50 the MIC. In the absence of vancomycin, the growth rates of the vancomycin-resistant S. aureus (VRSA) strains were similar to that of susceptible MRSA strain HIP11713. When resistance was induced, and under both conditions, there was a significant reduction of the growth rate of the VRSA strains relative to that of HIP11713 and to those of their noninduced counterparts, corresponding to a ca. 20% to 38% reduction in fitness. Competition experiments between isogenic VRSA-1 and HIP11713 mixed at a 1:1, 1:100, or 100:1 ratio revealed a competitive disadvantage of 0.4% to 3% per 10 generations of the transconjugant versus the recipient. This slight fitness burden can be attributed to the basal level of expression of the van genes in the absence of induction combined with a gene dosage effect due to the presence of the van operon on multicopy plasmids. These data indicate that VanA-type resistance, when induced, is highly costly for the MRSA host, whereas in the absence of induction, its biological cost is minimal. Thus, the potential for the dissemination of VRSA clinical isolates should not be underestimated.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/drug effects , Vancomycin Resistance , Bacterial Proteins/genetics , Carbon-Oxygen Ligases/genetics , Conjugation, Genetic , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/growth & development , Microbial Sensitivity Tests
16.
J Cell Mol Med ; 13(9A): 2938-48, 2009 Sep.
Article in English | MEDLINE | ID: mdl-18657227

ABSTRACT

The use of genomic DNA rather than cDNA or mini-gene constructs in gene therapy might be advantageous as these contain intronic and long-range control elements vital for accurate expression. For gene therapy of cystic fibrosis though, no bacterial artificial chromosome (BAC), containing the whole CFTR gene is available. We have used Red homologous recombination to add a to a previously described vector to construct a new BAC vector with a 250.3-kb insert containing the whole coding region of the CFTR gene along with 40.1 kb of DNA 5' to the gene and 25 kb 3' to the gene. This includes all the known control elements of the gene. We evaluated expression by RT-PCR in CMT-93 cells and showed that the gene is expressed both from integrated copies of the BAC and also from episomes carrying the oriP/EBNA-1 element. Sequencing of the human CFTR mRNA from one clone showed that the BAC is functional and can generate correctly spliced mRNA in the mouse background. The BAC described here is the only CFTR genomic construct available on a convenient vector that can be readily used for gene expression studies or in vivo studies to test its potential application in gene therapy for cystic fibrosis.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gene Expression Regulation , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Electrophoresis, Gel, Pulsed-Field , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , In Situ Hybridization, Fluorescence , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombination, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
17.
Vaccine ; 26(16): 1934-41, 2008 Apr 07.
Article in English | MEDLINE | ID: mdl-18342411

ABSTRACT

Naturally invasive bacteria have been successfully used for mucosal delivery of DNA vaccines against bacterial, viral and tumour antigens. Recently, an alternative delivery system based on a genetically modified mutant of the non-pathogenic commensal bacterium Escherichia coli, was developed and successfully used to deliver therapeutic genes and immunogenic proteins to epithelial cells in vivo. In this work, we used these recombinant invasive bacteria to deliver DNA vaccines against two Mycobacterium tuberculosis proteins (FbpA, and HtpX) following intranasal administration. Both DNA vaccines were able to induce an antigen-specific T-cell response. Moreover, mice immunized with the recombinant bacteria carrying the DNA vaccine encoding HtpX, were significatively protected from challenge with M. tuberculosis.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Escherichia coli/metabolism , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis/immunology , Tuberculosis/prevention & control , Vaccination , Acyltransferases/biosynthesis , Acyltransferases/genetics , Administration, Intranasal , Animals , Antigens, Bacterial/biosynthesis , Bacterial Proteins/biosynthesis , Cells, Cultured , Cytokines/biosynthesis , Female , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Immunization Schedule , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccines, DNA/administration & dosage
18.
Genomics ; 90(5): 610-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17822869

ABSTRACT

Hemophilia A is caused by mutations in the gene encoding factor VIII (F8) and is an important target for gene therapy. The F8 gene contains 26 exons spread over approximately 186 kb and no work using the intact genomic locus has been carried out. We have constructed a 250-kb BAC carrying all 26 exons, the introns, and more than 40 kb of upstream and 20 kb of downstream DNA. This F8 BAC was further retrofitted with either the oriP/EBNA-1 elements from Epstein-Barr virus, which allow episomal maintenance in mammalian cells, or alphoid DNA, which allows human artificial chromosome formation in some human cell lines. Lipofection of the oriP/EBNA-1-containing version into mouse Hepa1-6 cells resulted in expression of F8 mRNA spanning the F8 gene. The >300-kb BAC carrying alphoid DNA was successfully delivered to 293A and HT1080 cells using bacterial delivery, resulting in greater than endogenous levels of F8 mRNA expression.


Subject(s)
Chromosomes, Artificial, Bacterial , DNA/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Factor VIII/genetics , RNA, Messenger/genetics , Recombination, Genetic , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cells, Cultured , DNA/analysis , Factor VIII/metabolism , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Gene Targeting , Genetic Vectors , Humans , Kidney/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Plasmids , Polymerase Chain Reaction , RNA, Messenger/metabolism , Transfection
19.
Infect Immun ; 73(3): 1847-51, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731086

ABSTRACT

Nasal carriage is a major risk factor for Staphylococcus aureus infection, especially for methicillin-resistant strains (MRSA). Using a mouse model of nasal carriage, we have compared several S. aureus strains and demonstrated increased colonization levels by MRSA in cystic fibrosis transmembrane conductance regulator-deficient mice and Toll-like receptor 2 (TLR2)-deficient mice but not TLR4-deficient mice.


Subject(s)
Carrier State/microbiology , Methicillin Resistance , Nose/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/isolation & purification , Animals , Colony Count, Microbial , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CFTR , Mice, Knockout , Receptors, Cell Surface/genetics , Staphylococcus aureus/drug effects , Toll-Like Receptor 2 , Toll-Like Receptor 4
20.
J Mol Microbiol Biotechnol ; 6(3-4): 211-8, 2003.
Article in English | MEDLINE | ID: mdl-15153774

ABSTRACT

Intracellular bacteria can act as DNA delivery vectors into mammalian cells. Transfer of genetic information can be monitored by screening for cellular expression of a reporter gene under the control of an eukaryotic promoter. However, intracellular bacteria can also efficiently deliver heterologous proteins in the cell cytosol. We have studied the activity of the eukaryotic PCMV promoter in Escherichia coli and Salmonella typhimurium using the lacZ and gfp genes as reporters and determined its strength relative to those of PRSV and PSV40 in E. coli. We found substantial heterologous activity of fragments carrying the PCMV and PRSV promoters, the strength of PRSV being greater than that of PCMV, but did not detect any PSV40 activity in E. coli. The green fluorescent protein (GFP) synthesized in E. coli was transferred to COS-1 cells where it was detectable and stable. Insertion of a transcription terminator or deletion of the bacterial ribosome binding site downstream from PCMV led to the silencing of the promoter in bacteria but not in mammalian cells. These observations should incite to exert caution when interpreting data on the DNA transfer from bacteria to mammallian cells when the genes of interest are under the control of the PCMV or PRSV promoter.


Subject(s)
Escherichia coli/genetics , Luminescent Proteins/biosynthesis , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Salmonella typhimurium/genetics , beta-Galactosidase/biosynthesis , Animals , COS Cells , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , Escherichia coli/metabolism , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins , Lac Operon , Plasmids , Salmonella typhimurium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...