Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 4731, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830889

ABSTRACT

Major antibiotic groups are losing effectiveness due to the uncontrollable spread of antimicrobial resistance (AMR) genes. Among these, ß-lactam resistance genes -encoding ß-lactamases- stand as the most common resistance mechanism in Enterobacterales due to their frequent association with mobile genetic elements. In this context, novel approaches that counter mobile AMR are urgently needed. Collateral sensitivity (CS) occurs when the acquisition of resistance to one antibiotic increases susceptibility to another antibiotic and can be exploited to eliminate AMR selectively. However, most CS networks described so far emerge as a consequence of chromosomal mutations and cannot be leveraged to tackle mobile AMR. Here, we dissect the CS response elicited by the acquisition of a prevalent antibiotic resistance plasmid to reveal that the expression of the ß-lactamase gene blaOXA-48 induces CS to colistin and azithromycin. We next show that other clinically relevant mobile ß-lactamases produce similar CS responses in multiple, phylogenetically unrelated E. coli strains. Finally, by combining experiments with surveillance data comprising thousands of antibiotic susceptibility tests, we show that ß-lactamase-induced CS is pervasive within Enterobacterales. These results highlight that the physiological side-effects of ß-lactamases can be leveraged therapeutically, paving the way for the rational design of specific therapies to block mobile AMR or at least counteract their effects.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Microbial Sensitivity Tests , beta-Lactamases , beta-Lactamases/genetics , beta-Lactamases/metabolism , Escherichia coli/genetics , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacology , Drug Collateral Sensitivity/genetics , Plasmids/genetics , Azithromycin/pharmacology , Colistin/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , beta-Lactam Resistance/genetics
2.
Proc Natl Acad Sci U S A ; 119(15): e2109370119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35385351

ABSTRACT

Collateral sensitivity is an evolutionary trade-off whereby acquisition of the adaptive phenotype of resistance to an antibiotic leads to the nonadaptive increased susceptibility to another. The feasibility of harnessing such a trade-off to design evolutionary-based approaches for treating bacterial infections has been studied using model strains. However, clinical application of collateral sensitivity requires its conservation among strains presenting different mutational backgrounds. Particularly relevant is studying collateral sensitivity robustness of already-antibiotic-resistant mutants when challenged with a new antimicrobial, a common situation in clinics that has hardly been addressed. We submitted a set of diverse Pseudomonas aeruginosa antibiotic-resistant mutants to short-term evolution in the presence of different antimicrobials. Ciprofloxacin selects different clinically relevant resistance mutations in the preexisting resistant mutants, which gave rise to the same, robust, collateral sensitivity to aztreonam and tobramycin. We then experimentally determined that alternation of ciprofloxacin with aztreonam is more efficient than ciprofloxacin­tobramycin alternation in driving the extinction of the analyzed antibiotic-resistant mutants. Also, we show that the combinations ciprofloxacin­aztreonam or ciprofloxacin­tobramycin are the most effective strategies for eliminating the tested P. aeruginosa antibiotic-resistant mutants. These findings support that the identification of conserved collateral sensitivity patterns may guide the design of evolution-based strategies to treat bacterial infections, including those due to antibiotic-resistant mutants. Besides, this is an example of phenotypic convergence in the absence of parallel evolution that, beyond the antibiotic-resistance field, could facilitate the understanding of evolution processes, where the selective forces giving rise to new, not clearly adaptive phenotypes remain unclear.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Drug Collateral Sensitivity , Drug Resistance, Bacterial , Pseudomonas Infections , Pseudomonas aeruginosa , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Ciprofloxacin/pharmacology , Ciprofloxacin/therapeutic use , Drug Collateral Sensitivity/genetics , Drug Resistance, Bacterial/genetics , Humans , Microbial Sensitivity Tests , Mutation , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics
3.
Int J Antimicrob Agents ; 56(1): 105994, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32335276

ABSTRACT

Antibiotic collateral sensitivity (CS) occurs when a bacterium that acquires resistance to a treatment drug exhibits decreased resistance to a different drug. Here we identify reciprocal CS networks and candidate genes in Burkholderia multivorans. Burkholderia multivorans was evolved to become resistant to each of six antibiotics. The antibiogram of the evolved strain was compared with the immediate parental strain to determine CS and cross-resistance. The evolution process was continued for each resistant strain. CS interactions were observed in 170 of 279 evolved strains. CS patterns grouped into two clusters based on the treatment drug being a ß-lactam antibiotic or not. Reciprocal pairs of CS antibiotics arose in ≥25% of all evolved strains. A total of 68 evolved strains were subjected to whole-genome sequencing and the resulting mutation patterns were correlated with antibiograms. Analysis revealed there was no single gene responsible for CS and that CS seen in B. multivorans is likely due to a combination of specific and non-specific mutations. The frequency of reciprocal CS, and the degree to which resistance changed, suggests a long-term treatment strategy; when resistance to one drug occurs, switch to use of the other member of the reciprocal pair. This switching could theoretically be continued indefinitely, allowing life-long treatment of chronic infections with just two antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burkholderia Infections/drug therapy , Burkholderia/drug effects , Burkholderia/genetics , Drug Collateral Sensitivity/genetics , Drug Resistance, Bacterial/genetics , Burkholderia/isolation & purification , Cystic Fibrosis/pathology , Drug Collateral Sensitivity/drug effects , Genome, Bacterial/genetics , Humans , Lung/microbiology , Lung/pathology , Microbial Sensitivity Tests , Whole Genome Sequencing , beta-Lactams/pharmacology
4.
PLoS Biol ; 18(1): e3000612, 2020 01.
Article in English | MEDLINE | ID: mdl-31986134

ABSTRACT

Antibiotic resistance increasingly limits the success of antibiotic treatments, and physicians require new ways to achieve efficient treatment despite resistance. Resistance mechanisms against a specific antibiotic class frequently confer increased susceptibility to other antibiotic classes, a phenomenon designated collateral sensitivity (CS). An informed switch of antibiotic may thus enable the efficient treatment of resistant strains. CS occurs in many pathogens, but the mechanisms that generate hypersusceptibility are largely unknown. We identified several molecular mechanisms of CS against the antibiotic nitrofurantoin (NIT). Mutants that are resistant against tigecycline (tetracycline), mecillinam (ß-lactam), and protamine (antimicrobial peptide) all show CS against NIT. Their hypersusceptibility is explained by the overexpression of nitroreductase enzymes combined with increased drug uptake rates, or increased drug toxicity. Increased toxicity occurs through interference of the native drug-response system for NIT, the SOS response, with growth. A mechanistic understanding of CS will help to develop drug switches that combat resistance.


Subject(s)
Drug Collateral Sensitivity/genetics , Nitrofurantoin/pharmacology , Activation, Metabolic/drug effects , Activation, Metabolic/genetics , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/drug effects , Microbial Sensitivity Tests , Mutation/drug effects , Nitrofurantoin/pharmacokinetics , Organisms, Genetically Modified , Prodrugs/pharmacokinetics , Salmonella enterica/drug effects , Salmonella enterica/genetics , Salmonella enterica/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
5.
Mol Biol Evol ; 37(5): 1394-1406, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31851309

ABSTRACT

Evolutionary adaptation of bacteria to nonantibiotic selective forces, such as osmotic stress, has been previously associated with increased antibiotic resistance, but much less is known about potentially sensitizing effects of nonantibiotic stressors. In this study, we use laboratory evolution to investigate adaptation of Enterococcus faecalis, an opportunistic bacterial pathogen, to a broad collection of environmental agents, ranging from antibiotics and biocides to extreme pH and osmotic stress. We find that nonantibiotic selection frequently leads to increased sensitivity to other conditions, including multiple antibiotics. Using population sequencing and whole-genome sequencing of single isolates from the evolved populations, we identify multiple mutations in genes previously linked with resistance to the selecting conditions, including genes corresponding to known drug targets or multidrug efflux systems previously tied to collateral sensitivity. Finally, we hypothesized based on the measured sensitivity profiles that sequential rounds of antibiotic and nonantibiotic selection may lead to hypersensitive populations by harnessing the orthogonal collateral effects of particular pairs of selective forces. To test this hypothesis, we show experimentally that populations evolved to a sequence of linezolid (an oxazolidinone antibiotic) and sodium benzoate (a common preservative) exhibit increased sensitivity to more stressors than adaptation to either condition alone. The results demonstrate how sequential adaptation to drug and nondrug environments can be used to sensitize bacteria to antibiotics and highlight new potential strategies for exploiting shared constraints governing adaptation to diverse environmental challenges.


Subject(s)
Drug Collateral Sensitivity/genetics , Drug Resistance, Bacterial/genetics , Enterococcus faecalis/genetics , Selection, Genetic , Stress, Physiological/genetics , Anti-Bacterial Agents , Linezolid , Sodium Benzoate , Triclosan
SELECTION OF CITATIONS
SEARCH DETAIL
...