Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.276
Filter
1.
Commun Biol ; 7(1): 1051, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187619

ABSTRACT

Efflux-mediated ß-lactam resistance is a major public health concern, reducing the effectiveness of ß-lactam antibiotics against many bacteria. Structural analyses show the efflux protein TolC in Gram-negative bacteria acts as a channel for antibiotics, impacting bacterial susceptibility and virulence. This study examines ß-lactam drug efflux mediated by TolC using experimental and computational methods. Molecular dynamics simulations of drug-free TolC reveal essential movements and key residues involved in TolC opening. A whole-gene-saturation mutagenesis assay, mutating each TolC residue and measuring fitness effects under ß-lactam selection, is performed. Here we show the TolC-mediated efflux of three antibiotics: oxacillin, piperacillin, and carbenicillin. Steered molecular dynamics simulations identify general and drug-specific efflux mechanisms, revealing key positions at TolC's periplasmic entry affecting efflux motions. Our findings provide insights into TolC's structural dynamics, aiding the design of new antibiotics to overcome bacterial efflux mechanisms.


Subject(s)
Anti-Bacterial Agents , Bacterial Outer Membrane Proteins , Molecular Dynamics Simulation , beta-Lactam Resistance , beta-Lactam Resistance/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Escherichia coli/drug effects , Microbial Sensitivity Tests , Protein Conformation
2.
Microbiol Spectr ; 12(9): e0381523, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39041813

ABSTRACT

Reliable detection of mecA and mecC-mediated beta-lactam resistance using automated antimicrobial susceptibility test systems is critical for patient care. The aim of this study was to compare the performance of the new cefoxitin screen test (oxsf02n) on the Vitek 2 card (Vitek 2) and BD Phoenix PMC-100 Gram-Positive AST Panel (Phoenix) against the reference method for the detection of mecA (and mecC)-mediated beta-lactam resistance. Two hundred fifty clinical fresh and stock Staphylococcus spp. isolates were included. There were 120 mecA-positive, 10 mecC-positive, and 120 mecA and mecC-negative isolates. Cefoxitin screen and oxacillin tests were performed on Vitek 2 and Phoenix and by their respective reference methods (disk diffusion for the cefoxitin screen test and broth microdilution for oxacillin) for all isolates. PCR testing was also performed to confirm the presence of mecA and/or mecC genes. Results from each system were compared to the reference methods. Statistical hypotheses were evaluated both for Vitek 2 compared to the reference methods and Vitek 2 compared to the Phoenix. Compared to the reference method, the Vitek 2 cefoxitin screen test had 100% sensitivity/98% specificity and the Phoenix cefoxitin screen test had 84% sensitivity/100% specificity for the detection of mecA (and mecC)-mediated beta-lactam resistance. When the oxacillin test was combined with the cefoxitin screen for Vitek 2, the sensitivity and specificity were unchanged. However, when the oxacillin and cefoxitin screen tests were combined for the Phoenix, the sensitivity increased to 100% and the specificity remained unchanged (100%). When considering cefoxitin alone, the Vitek 2 screen test showed a higher sensitivity than the Phoenix for the detection of mecA and mecC-mediated beta-lactam resistance. However, currently, both systems use a combination of the cefoxitin and oxacillin tests to interpret the final result, and both reached a high level of performance when cefoxitin and oxacillin results were combined.IMPORTANCEThis research marks the inaugural evaluation of the revamped cefoxitin screen test version in Vitek 2, juxtaposing it against reference methods and a primary competitor BD Phoenix.


Subject(s)
Anti-Bacterial Agents , Cefoxitin , Microbial Sensitivity Tests , Cefoxitin/pharmacology , Microbial Sensitivity Tests/methods , Anti-Bacterial Agents/pharmacology , Humans , Bacterial Proteins/genetics , Oxacillin/pharmacology , Staphylococcus/drug effects , Staphylococcus/genetics , beta-Lactam Resistance , Staphylococcal Infections/microbiology , Penicillin-Binding Proteins/genetics
3.
J Antimicrob Chemother ; 79(9): 2170-2178, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39001778

ABSTRACT

BACKGROUND: ß-lactam antibiotics, which inhibit penicillin-binding protein 3 (PBP3) that is required for cell division, play a key role in treating P. aeruginosa infections. Some sequence variations in PBP3 have been associated with ß-lactam resistance but the effects of variations on antibiotic susceptibility and on cell division have not been quantified. Antibiotic efflux can also reduce susceptibility. OBJECTIVES: To quantify the effects of PBP3 variations on ß-lactam susceptibility and cell morphology in P. aeruginosa. METHODS: Nineteen PBP3 variants were expressed from a plasmid in the reference strain P. aeruginosa PAO1 and genome engineering was used to construct five mutants expressing PBP3 variants from the chromosome. The effects of the variations on ß-lactam minimum inhibitory concentration (MIC) and cell morphology were measured. RESULTS: Some PBP3 variations reduced susceptibility to a variety of ß-lactam antibiotics including meropenem, ceftazidime, cefepime and ticarcillin with different variations affecting different antibiotics. None of the tested variations reduced susceptibility to imipenem or piperacillin. Antibiotic susceptibility was further reduced when PBP3 variants were expressed in mutant bacteria overexpressing the MexAB-OprM efflux pump, with some variations conferring clinical levels of resistance. Some PBP3 variations, and sub-MIC levels of ß-lactams, reduced bacterial growth rates and inhibited cell division, causing elongated cells. CONCLUSIONS: PBP3 variations in P. aeruginosa can increase the MIC of multiple ß-lactam antibiotics, although not imipenem or piperacillin. PBP3 variations, or the presence of sub-lethal levels of ß-lactams, result in elongated cells indicating that variations reduce the activity of PBP3 and may reduce bacterial fitness.


Subject(s)
Anti-Bacterial Agents , Cell Division , Microbial Sensitivity Tests , Penicillin-Binding Proteins , Pseudomonas aeruginosa , beta-Lactams , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Penicillin-Binding Proteins/genetics , beta-Lactams/pharmacology , Anti-Bacterial Agents/pharmacology , Cell Division/drug effects , beta-Lactam Resistance/genetics , Genetic Variation , Plasmids/genetics
4.
Lasers Surg Med ; 56(7): 673-681, 2024 09.
Article in English | MEDLINE | ID: mdl-39039622

ABSTRACT

OBJECTIVE: In this study, we evaluated the effectiveness of antimicrobial blue light (aBL; 410 nm wavelength) against ß-lactamase-carrying bacteria and the effect of aBL on the activity of ß-lactamases. METHODS: Pseudomonas aeruginosa, Escherichia coli, and Klebsiella pneumoniae strains carrying ß-lactamases as well as a purified ß-lactamase enzymes were studied. ß-lactamase activity was assessed using a chromogenic cephalosporin hydrolysis assay. Additionally, we evaluated the role of porphyrins in the photoreaction, as well as protein degradation by sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Finally, we investigated the bactericidal effect of combined aBL-ceftazidime exposure against a metallo-ß-lactamase expressing P. aeruginosa strain. RESULTS: Our study demonstrated that aBL effectively killed ß-lactamase-producing bacteria and reduced ß-lactamase activity. After an aBL exposure of 1.52 J/cm2, a 50% reduction in enzymatic activity was observed in P. aeruginosa. Additionally, we found a 40% decrease in the photoreaction activity of porphyrins following an aBL exposure of 64.8 J/cm2. We also revealed that aBL reduced ß-lactamase activity via protein degradation (after 136.4 J/cm2). Additionally, aBL markedly improved the bactericidal effect of ceftazidime (by >4-log10) in the metallo-ß-lactamase P. aeruginosa strain. CONCLUSION: Our results provide evidence that aBL compromises bacterial ß-lactamase activity, offering a potential approach to overcome ß-lactam resistance in bacteria.


Subject(s)
Blue Light , Escherichia coli , Klebsiella pneumoniae , Pseudomonas aeruginosa , beta-Lactam Resistance , beta-Lactamases , Anti-Bacterial Agents/pharmacology , beta-Lactam Resistance/radiation effects , beta-Lactamases/metabolism , Ceftazidime/pharmacology , Escherichia coli/drug effects , Escherichia coli/radiation effects , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/radiation effects , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/radiation effects
5.
Front Cell Infect Microbiol ; 14: 1395260, 2024.
Article in English | MEDLINE | ID: mdl-39081869

ABSTRACT

Patients with severe carbapenem-resistant Acinetobacter baumannii (CRAB) infections currently face significant treatment challenges. When patients display signs of infection and the clinical suspicion of CRAB infections is high, appropriate treatment should be immediately provided. However, current treatment plans and clinical data for CRAB are limited. Inherent and acquired resistance mechanisms, as well as host factors, significantly restrict options for empirical medication. Moreover, inappropriate drug coverage can have detrimental effects on patients. Most existing studies have limitations, such as a restricted sample size, and are predominantly observational or non-randomized, which report significant variability in patient infection severity and comorbidities. Therefore, a gold-standard therapy remains lacking. Current and future treatment options of infections due to CRAB were described in this review. The dose and considerable side effects restrict treatment options for polymyxins, and high doses of ampicillin-sulbactam or tigecycline appear to be the best option at the time of initial treatment. Moreover, new drugs such as durlobactam and cefiderocol have substantial therapeutic capabilities and may be effective salvage treatments. Bacteriophages and antimicrobial peptides may serve as alternative treatment options in the near future. The advantages of a combination antimicrobial regimen appear to predominate those of a single regimen. Despite its significant nephrotoxicity, colistin is considered a primary treatment and is often used in combination with antimicrobials, such as tigecycline, ampicillin-sulbactam, meropenem, or fosfomycin. The Infectious Diseases Society of America (IDSA) has deemed high-dose ampicillin-sulbactam, which is typically combined with high-dose tigecycline, polymyxin, and other antibacterial agents, the best option for treating serious CRAB infections. A rational combination of drug use and the exploration of new therapeutic drugs can alleviate or prevent the effects of CRAB infections, shorten hospital stays, and reduce patient mortality.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Anti-Bacterial Agents , Carbapenems , Acinetobacter baumannii/drug effects , Humans , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Carbapenems/therapeutic use , Carbapenems/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Drug Therapy, Combination , beta-Lactam Resistance , Polymyxins/therapeutic use , Polymyxins/pharmacology , Drug Resistance, Multiple, Bacterial
6.
J Bacteriol ; 206(8): e0013024, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-38995039

ABSTRACT

c-di-AMP is an essential second messenger that binds and regulates several proteins of different functions within bacterial cells. Among those, PstA is a structurally conserved c-di-AMP-binding protein, but its function is largely unknown. PstA is structurally similar to PII signal transduction proteins, although it specifically binds c-di-AMP rather than other PII ligands such as ATP and α-ketoglutarate. In Listeria monocytogenes, we found that PstA increases ß-lactam susceptibility at normal and low c-di-AMP levels, but increases ß-lactam resistance upon c-di-AMP accumulation. Examining a PstA mutant defective for c-di-AMP binding, we found the apo form of PstA to be toxic for ß-lactam resistance, and the c-di-AMP-bound form to be beneficial. Intriguingly, a role for PstA in ß-lactam resistance is only prominent in aerobic cultures, and largely diminished under hypoxic conditions, suggesting that PstA function is linked to aerobic metabolism. However, PstA does not control aerobic growth rate, and has a modest influence on the tricarboxylic acid cycle and membrane potential-an indicator of cellular respiration. The regulatory role of PstA in ß-lactam resistance is unrelated to reactive oxygen species or oxidative stress. Interestingly, during aerobic growth, PstA function requires the cytochrome bd oxidase (CydAB), a component of the respiratory electron transport chain. The requirement for CydAB might be related to its function in maintaining a membrane potential, or redox stress response activities. Altogether, we propose a model in which apo-PstA diminishes ß-lactam resistance by interacting with an effector protein, and this activity can be countered by c-di-AMP binding or a by-product of redox stress. IMPORTANCE: PstA is a structurally conserved c-di-AMP-binding protein that is broadly present among Firmicutes bacteria. Furthermore, PstA binds c-di-AMP at high affinity and specificity, indicating an important role in the c-di-AMP signaling network. However, the molecular function of PstA remains elusive. Our findings reveal contrasting roles of PstA in ß-lactam resistance depending on c-di-AMP-binding status. We also define physiological conditions for PstA function during aerobic growth. Future efforts can exploit these conditions to identify PstA interaction partners under ß-lactam stress.


Subject(s)
Bacterial Proteins , Listeria monocytogenes , beta-Lactam Resistance , Listeria monocytogenes/drug effects , Listeria monocytogenes/genetics , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Listeria monocytogenes/growth & development , beta-Lactam Resistance/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cytochrome b Group/metabolism , Cytochrome b Group/genetics , beta-Lactams/pharmacology , Anti-Bacterial Agents/pharmacology , Gene Expression Regulation, Bacterial
7.
J Antimicrob Chemother ; 79(8): 2040-2047, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38973602

ABSTRACT

BACKGROUND: Viridans group streptococci (VGS) bloodstream infection (BSI) frequently occurs in cancer patients receiving chemotherapy, and is associated with infective endocarditis (IE) in up to 20% of cases in the general population. OBJECTIVES: In cancer patients receiving chemotherapy with VGS BSI, we aimed to: (i) determine the incidence of infective complications including IE, (ii) assess the utility of echocardiography in this patient population, (iii) determine the duration and type of antimicrobial therapy received for monomicrobial infections, and (iv) determine the evolution of antimicrobial resistance. METHODS: VGS BSIs (excluding Streptococcus pneumoniae and Streptococcus pseudopneumoniae) in cancer patients receiving chemotherapy were identified from a statewide public pathology database between 2013 and 2022 at our tertiary centre. Medical records were accessed for clinical, microbiological and radiological data. RESULTS: Of 581 patient episodes screened, 183 episodes involving 171 patients met inclusion criteria. Of these, 51% were bone marrow transplantation (BMT) patients, 40% were non-BMT haematology patients, and 8% were solid organ malignancy patients. The median age was 55 years, and 96% were neutropenic at the time of blood culture collection. A transthoracic echocardiogram was performed for 71% of episodes, and one patient met modified Duke's criteria for definite IE, although this diagnosis was not suspected on clinical grounds. Other complications were uncommon. Benzylpenicillin resistance was rare (2.9%) and did not change over time. Most episodes (75%) were treated with piperacillin/tazobactam. For monomicrobial BSIs, the median antibiotic duration was 5 days (IQR 2-7) post-neutropenia resolution. CONCLUSIONS: Infective complications and antimicrobial resistance are rare in cancer patients with VGS BSI. This may provide a safe opportunity to limit both investigations (e.g. echocardiogram) and prolonged exposure to broad-spectrum antimicrobials.


Subject(s)
Anti-Bacterial Agents , Bacteremia , Neoplasms , Streptococcal Infections , Viridans Streptococci , beta-Lactam Resistance , Humans , Middle Aged , Neoplasms/drug therapy , Neoplasms/complications , Female , Male , Viridans Streptococci/drug effects , Viridans Streptococci/isolation & purification , Streptococcal Infections/microbiology , Streptococcal Infections/epidemiology , Aged , Bacteremia/microbiology , Bacteremia/drug therapy , Bacteremia/epidemiology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/adverse effects , Adult , Incidence , Retrospective Studies , Echocardiography , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Endocarditis, Bacterial/microbiology , Endocarditis, Bacterial/drug therapy
8.
mBio ; 15(8): e0121024, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39028200

ABSTRACT

The high-level resistance to next-generation ß-lactams frequently found in Staphylococcus aureus isolates lacking mec, which encodes the transpeptidase PBP2a traditionally associated with methicillin-resistant Staphylococcus aureus (MRSA), has remained incompletely understood for decades. A new study by Lai et al. found that the co-occurrence of mutations in pbp4 and gdpP, which respectively cause increased PBP4-mediated cell wall crosslinking and elevated cyclic-di-AMP levels, produces synergistic ß-lactam resistance rivaling that of PBP2a-producing MRSA (L.-Y. Lai, N. Satishkumar, S. Cardozo, V. Hemmadi, et al., mBio 15:e02889-23. 2024, https://doi.org/10.1128/mbio.02889-23). The combined mutations are sufficient to explain the high-level ß-lactam resistance of some mec-lacking strains, but the mechanism of synergy remains elusive and an avenue for further research. Importantly, the authors establish that co-occurrence of these mutations leads to antibiotic therapy failure in a Caenorhabditis elegans infection model. These results underscore the need to consider this unique and novel ß-lactam resistance mechanism during the clinical diagnosis of MRSA, rather than relying on mec as a diagnostic.


Subject(s)
Anti-Bacterial Agents , Caenorhabditis elegans , Methicillin-Resistant Staphylococcus aureus , Penicillin-Binding Proteins , Staphylococcal Infections , beta-Lactams , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , beta-Lactams/pharmacology , Anti-Bacterial Agents/pharmacology , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Caenorhabditis elegans/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , beta-Lactam Resistance/genetics , Mutation , Microbial Sensitivity Tests , Cell Wall/metabolism , Cell Wall/drug effects , Humans , Cyclic AMP/metabolism , beta Lactam Antibiotics
9.
Pharmacotherapy ; 44(8): 658-674, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38949413

ABSTRACT

Antibiotic resistance has become a global threat as it is continuously growing due to the evolution of ß-lactamases diminishing the activity of classic ß-lactam (BL) antibiotics. Recent antibiotic discovery and development efforts have led to the availability of ß-lactamase inhibitors (BLIs) with activity against extended-spectrum ß-lactamases as well as Klebsiella pneumoniae carbapenemase (KPC)-producing carbapenem-resistant organisms (CRO). Nevertheless, there is still a lack of drugs that target metallo-ß-lactamases (MBL), which hydrolyze carbapenems efficiently, and oxacillinases (OXA) often present in carbapenem-resistant Acinetobacter baumannii. This review aims to provide a snapshot of microbiology, pharmacology, and clinical data for currently available BL/BLI treatment options as well as agents in late stage development for CRO harboring various ß-lactamases including MBL and OXA-enzymes.


Subject(s)
Anti-Bacterial Agents , Carbapenems , Gram-Negative Bacteria , beta-Lactamase Inhibitors , beta-Lactams , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactams/pharmacology , beta-Lactams/therapeutic use , Carbapenems/pharmacology , Carbapenems/therapeutic use , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , beta-Lactam Resistance , beta-Lactamases
10.
Diagn Microbiol Infect Dis ; 110(1): 116358, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002448

ABSTRACT

The VITEK®2 AES ß-lactam phenotypes of 488 Enterobacterales from North and Latin America generated by the VITEK®2 were compared to the resistance genotypes provided by whole genome sequencing (WGS). The AES provided phenotypic reports for 447 (91.6 %) isolates, including isolates harbouring carbapenemases (195; 43.6 %), ESBLs (103; 23.0 %) and transferable AmpCs (tAmpC; 28; 6.3 %) genes, as well as wildtype isolates (WT; 121; 27.1 %). Overall, the AES report was accurate for 433/447 (96.9 %) isolates. The AES accurately reported carbapenemase, ESBL, and tAmpC phenotypes for 93.7 %, 93.7 %, and 98.4 % of isolates, respectively, and sensitivity/specificity rates were 96.4 %/91.7 %, 98.1 %/92.4 %, 82.1 %/99.5 %, and 100 %/98.8 %. 14 isolates carrying carbapenemase (7 total; 3 KPC, 2 MBL, 2 OXA-48-like), ESBL (2), and tAmpC-encoding genes (5) were not correctly identified by AES. The AES phenotypic report detected resistance mechanisms among Enterobacterales rapidly and could significantly aid future antimicrobial stewardship initiatives and patient care.


Subject(s)
Bacterial Proteins , Enterobacteriaceae Infections , Enterobacteriaceae , Microbial Sensitivity Tests , Phenotype , Whole Genome Sequencing , beta-Lactamases , Latin America , Humans , Enterobacteriaceae/genetics , Enterobacteriaceae/drug effects , beta-Lactamases/genetics , Enterobacteriaceae Infections/microbiology , Bacterial Proteins/genetics , beta-Lactam Resistance/genetics , Anti-Bacterial Agents/pharmacology , North America , beta-Lactams/pharmacology , Genotype , Genome, Bacterial/genetics
12.
mBio ; 15(7): e0141924, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38920394

ABSTRACT

Pseudomonas aeruginosa encodes the beta-lactamase AmpC, which promotes resistance to beta-lactam antibiotics. Expression of ampC is induced by anhydro-muropeptides (AMPs) released from the peptidoglycan (PG) cell wall upon beta-lactam treatment. AmpC can also be induced via genetic inactivation of PG biogenesis factors such as the endopeptidase DacB that cleaves PG crosslinks. Mutants in dacB occur in beta-lactam-resistant clinical isolates of P. aeruginosa, but it has remained unclear why DacB inactivation promotes ampC induction. Similarly, the inactivation of lytic transglycosylase (LT) enzymes such as SltB1 that cut PG glycans has also been associated with ampC induction and beta-lactam resistance. Given that LT enzymes are capable of producing AMP products that serve as ampC inducers, this latter observation has been especially difficult to explain. Here, we show that ampC induction in sltB1 or dacB mutants requires another LT enzyme called MltG. In Escherichia coli, MltG has been implicated in the degradation of nascent PG strands produced upon beta-lactam treatment. Accordingly, in P. aeruginosa sltB1 and dacB mutants, we detected the MltG-dependent production of pentapeptide-containing AMP products that are signatures of nascent PG degradation. Our results therefore support a model in which SltB1 and DacB use their PG-cleaving activity to open space in the PG matrix for the insertion of new material. Thus, their inactivation mimics low-level beta-lactam treatment by reducing the efficiency of new PG insertion into the wall, causing the degradation of some nascent PG material by MltG to produce the ampC-inducing signal. IMPORTANCE: Inducible beta-lactamases like the ampC system of Pseudomonas aeruginosa are a common determinant of beta-lactam resistance among gram-negative bacteria. The regulation of ampC is elegantly tuned to detect defects in cell wall synthesis caused by beta-lactam drugs. Studies of mutations causing ampC induction in the absence of drug therefore promise to reveal new insights into the process of cell wall biogenesis in addition to aiding our understanding of how resistance to beta-lactam antibiotics arises in the clinic. In this study, the ampC induction phenotype for mutants lacking a glycan-cleaving enzyme or an enzyme that cuts cell wall crosslinks was used to uncover a potential role for these enzymes in making space in the wall matrix for the insertion of new material during cell growth.


Subject(s)
Bacterial Proteins , Cell Wall , Pseudomonas aeruginosa , beta-Lactamases , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Cell Wall/metabolism , Cell Wall/drug effects , beta-Lactamases/genetics , beta-Lactamases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , beta-Lactam Resistance/genetics , Phenotype , Peptidoglycan/metabolism , Anti-Bacterial Agents/pharmacology , beta-Lactams/pharmacology , beta-Lactams/metabolism , Gene Expression Regulation, Bacterial
13.
J Environ Manage ; 363: 121288, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38850900

ABSTRACT

Inappropriate antibiotic use not only amplifies the threat of antimicrobial resistance (AMR), moreover exacerbates the spread of resistant bacterial strains and genes in the environment, underscoring the critical need for effective research and interventions. Our aim is to assess the prevalence and resistance characteristics of ß-lactam resistant bacteria (BLRB) and ß-lactamase resistant bacterial genes (BLRBGs) under various environmental conditions within Delhi NCR, India. Using a culture-dependent method, we isolated 130 BLRB from 75 different environmental samples, including lakes, ponds, the Yamuna River, agricultural soil, aquatic weeds, drains, dumping yards, STPs, and gaushalas. Tests for antibiotic susceptibility were conducted in addition to phenotypic and genotypic identification of BLs and integron genes. The water and sediment samples recorded an average bacterial abundance of 3.6 × 106 CFU/mL and an average ampicillin-resistant bacterial count of 2.2 × 106 CFU/mL, which can be considered a potent reservoir of BLRB and BLRBGs. The majority of the BLRB discovered are opportunistic pathogens from the Bacillus, Aeromonas, Pseudomonas, Enterobacter, Escherichia, and Klebsiella genera, with Multiple Antibiotic Resistance (MAR) index ≥0.2 against a wide variety of ß-lactams and ß-lactamase (BLs) inhibitor combinations. The antibiotic resistance pattern was similar in the case of bacteria isolated from STPs. Meanwhile, bacteria isolated from other sources were diverse in their antibiotic resistance profile. Interestingly, we discovered that 10 isolates of various origins produce both Extended Spectrum BLs and Metallo BLs, as well as found harboring blaTEM, blaCTX, blaOXA, blaSHV, int-1, and int-3 genes. Enterobacter cloacae (S50/A), a common nosocomial pathogen isolated from Yamuna River sediment samples at Nizamuddin point, possesses three BLRBGs (blaTEM, blaCTX, and blaOXA) and a MAR index of 1.0, which is a major cause for concern. Therefore, identifying the source, origin and dissemination of BLRB and BLRGs in the environment is of the utmost importance for designing effective mitigation approaches to reduce a load of antimicrobial resistance factors in the environmental settings.


Subject(s)
Anti-Bacterial Agents , India , Anti-Bacterial Agents/pharmacology , beta-Lactamases/genetics , beta-Lactam Resistance , Bacteria/drug effects , Bacteria/genetics , Microbial Sensitivity Tests , beta-Lactams/pharmacology
14.
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
15.
J Antimicrob Chemother ; 79(7): 1577-1580, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38742706

ABSTRACT

BACKGROUND: The blaB, blaGOB and blaCME genes are thought to confer ß-lactam resistance to Elizabethkingia anophelis, based on experiments conducted primarily on Escherichia coli. OBJECTIVES: To determine the individual contributions of ß-lactamase genes to increased MICs in E. anophelis and to assess their impact on the in vivo efficacy of carbapenem therapy. METHODS: Scarless gene deletion of one or more ß-lactamase gene(s) was performed in three clinical E. anophelis isolates. MICs were determined by broth microdilution. Hydrolytic activity and expressions of ß-lactamase genes were measured by an enzymatic assay and quantitative RT-PCR, respectively. In vivo efficacy was determined using Galleria mellonella and murine thigh infection models. RESULTS: The presence of blaB resulted in >16-fold increases, while blaGOB caused 4-16-fold increases of carbapenem MICs. Hydrolysis of carbapenems was highest in lysates of blaB-positive strains, possibly due to the constitutionally higher expression of blaB. Imipenem was ineffective against blaB-positive isolates in vivo in terms of improvement of the survival of wax moth larvae and reduction of murine bacterial load. The deletion of blaB restored the efficacy of imipenem. The blaB gene was also responsible for a >4-fold increase of ampicillin/sulbactam and piperacillin/tazobactam MICs. The presence of blaCME, but not blaB or blaGOB, increased the MICs of ceftazidime and cefepime by 8-16- and 4-8-fold, respectively. CONCLUSIONS: The constitutionally and highly expressed blaB gene in E. anophelis was responsible for increased MICs of carbapenems and led to their poor in vivo efficacy. blaCME increased the MICs of ceftazidime and cefepime.


Subject(s)
Anti-Bacterial Agents , Flavobacteriaceae Infections , Flavobacteriaceae , Microbial Sensitivity Tests , beta-Lactamases , beta-Lactams , Animals , beta-Lactamases/genetics , beta-Lactamases/metabolism , Flavobacteriaceae/drug effects , Flavobacteriaceae/genetics , Flavobacteriaceae Infections/microbiology , Flavobacteriaceae Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Mice , beta-Lactams/pharmacology , Disease Models, Animal , Carbapenems/pharmacology , Moths/microbiology , Humans , beta-Lactam Resistance/genetics , Female
16.
Adv Sci (Weinh) ; 11(28): e2400858, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38747156

ABSTRACT

Small molecule can be utilized to restore the effectiveness of existing major classes of antibiotics against antibiotic-resistant bacteria. In this study, it is demonstrated that celastrol, a natural compound, can modify the bacterial cell wall and subsequently render bacteria more suceptible to ß-lactam antibiotics. It is shown that celastrol leads to incomplete cell wall crosslinking by modulating levels of c-di-AMP, a secondary messenger, in methicillin-resistant Staphylococcus aureus (MRSA). This mechanism enables celastrol to act as a potentiator, effectively rendering MRSA susceptible to a range of penicillins and cephalosporins. Restoration of in vivo susceptibility of MRSA to methicillin is also demonstrated using a sepsis animal model by co-administering methicillin along with celastrol at a much lower amount than that of methicillin. The results suggest a novel approach for developing potentiators for major classes of antibiotics by exploring molecules that re-program metabolic pathways to reverse ß-lactam-resistant strains to susceptible strains.


Subject(s)
Anti-Bacterial Agents , Methicillin-Resistant Staphylococcus aureus , Peptidoglycan , beta-Lactam Resistance , Methicillin-Resistant Staphylococcus aureus/drug effects , Peptidoglycan/metabolism , beta-Lactam Resistance/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Microbial Sensitivity Tests , Mice , Pentacyclic Triterpenes/pharmacology , Staphylococcal Infections/drug therapy , Cell Wall/metabolism , Cell Wall/drug effects , beta-Lactams/pharmacology , Triterpenes/pharmacology , Triterpenes/metabolism
17.
Int J Antimicrob Agents ; 64(1): 107185, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38692492

ABSTRACT

OBJECTIVES: Using a random forest algorithm, we previously found that teicoplanin-associated gene A (tcaA) might play a role in resistance of methicillin-resistant Staphylococcus aureus (MRSA) to ß-lactams, which we have investigated further here. METHODS: Representative MRSA strains of prevalent clones were selected to identify the role of tcaA in the MRSA response to ß-lactams. tcaA genes were deleted by homologous recombination in the selected MRSA strains, and antibiotic susceptibility tests were applied to evaluate the effect of tcaA on the minimum inhibitory concentrations (MICs) of glycopeptides and ß-lactams. Scanning electron microscopy, RNA sequencing, and quantitative reverse transcription-polymerase chain reaction were performed to explore the mechanism of tcaA in MRSA resistance to ß-lactams. RESULTS: The MIC of penicillin plus clavulanate decreased from 3 mg/L to 0.064 mg/L and that of oxacillin decreased from 16 to 0.5 mg/L when tcaA was knocked out in the LAC strain. Compared with wild-type MRSA isolates, when tcaA was deleted, all selected strains were more susceptible to ß-lactams. Susceptibility to ceftobiprole was restored in the ceftobiprole-resistant strain when tcaA was deleted. tcaA knockout caused "log-like" abnormal division of MRSA, and tcaA deficiency mediated low expression of mecA, ponA, and murA2. CONCLUSIONS: Machine learning is a reliable tool for identifying drug resistance-related genes. tcaA may be involved in S. aureus cell division and may affect mecA, ponA, and murA2 expression. Furthermore, tcaA is a potential resistance breaker target for ß-lactams, including ceftobiprole, in MRSA.


Subject(s)
Anti-Bacterial Agents , Cephalosporins , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , beta-Lactam Resistance , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Humans , beta-Lactam Resistance/genetics , Bacterial Proteins/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , beta-Lactams/pharmacology , Gene Knockout Techniques
18.
BMC Genomics ; 25(1): 508, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778284

ABSTRACT

BACKGROUND: Enzymatic degradation mediated by beta-lactamases constitutes one of the primary mechanisms of resistance to beta-lactam antibiotics in gram-negative bacteria. This enzyme family comprises four molecular classes, categorized into serine beta-lactamases (Classes A, C, and D) and zinc-dependent metallo-beta-lactamases (Class B). Gram-negative bacteria producing beta-lactamase are of significant concern, particularly due to their prevalence in nosocomial infections. A comprehensive understanding of the evolution and dissemination of this enzyme family is essential for effective control of these pathogens. In this study, we conducted the prospecting, phylogenetic analysis, and in silico analysis of beta-lactamases and homologous proteins identified in 1827 bacterial genomes with phenotypic data on beta-lactam resistance. These genomes were distributed among Klebsiella pneumoniae (45%), Acinetobacter baumannii (31%), Pseudomonas aeruginosa (14%), Escherichia coli (6%), and Enterobacter spp. (4%). Using an HMM profile and searching for conserved domains, we mined 2514, 8733, 5424, and 2957 proteins for molecular classes A, B, C, and D, respectively. This set of proteins encompasses canonical subfamilies of beta-lactamases as well as hypothetical proteins and other functional groups. Canonical beta-lactamases were found to be phylogenetically distant from hypothetical proteins, which, in turn, are closer to other representatives of the penicillin-binding-protein (PBP-like) and metallo-beta-lactamase (MBL) families. The catalytic amino acid residues characteristic of beta-lactamases were identified from the sequence alignment and revealed that motifs are less conserved in homologous groups than in beta-lactamases. After comparing the frequency of protein groups in genomes of resistant strains with those of sensitive ones applying Fisher's exact test and relative risk, it was observed that some groups of homologous proteins to classes B and C are more common in the genomes of resistant strains, particularly to carbapenems. We identified the beta-lactamase-like domain widely distributed in gram-negative species of the ESKAPEE group, which highlights its importance in the context of beta-lactam resistance. Some hypothetical homologous proteins have been shown to potentially possess promiscuous activity against beta-lactam antibiotics, however, they do not appear to expressly determine the resistance phenotype. The selective pressure due to the widespread use of antibiotics may favor the optimization of these functions for specialized resistance enzymes.


Subject(s)
Gram-Negative Bacteria , Phylogeny , beta-Lactamases , beta-Lactamases/metabolism , beta-Lactamases/genetics , beta-Lactamases/chemistry , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/enzymology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , beta-Lactams/pharmacology , beta-Lactams/metabolism , Anti-Bacterial Agents/pharmacology , Genome, Bacterial , beta-Lactam Resistance/genetics , beta Lactam Antibiotics
19.
J Hosp Infect ; 149: 56-64, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735628

ABSTRACT

BACKGROUND: Pseudomonas aeruginosa bloodstream infections (PA-BSIs) are a serious disease and a therapeutic challenge due to increasing resistance to carbapenems. Our objectives were to describe the prevalence and risk factors associated with carbapenem resistance (CR) and mortality in children with PA-BSI. METHODS: A retrospective, multi-centre study was carried out, including patients aged <20 years with PA-BSI in four tertiary hospitals in Madrid (Spain) during 2010-2020. Risk factors for CR PA-BSIs and 30-day mortality were evaluated in a multi-variable logistic regression model. RESULTS: In total, 151 patients with PA-BSI were included, with a median age of 29 months (interquartile range: 3.5-87.1). Forty-five (29.8%) cases were CR, 9.9% multi-drug resistant and 6.6% extensively drug resistant. The prevalence of CR remained stable throughout the study period, with 26.7% (12/45) of CR mediated by VIM-type carbapenemase. Patients with BSIs produced by CR-PA were more likely to receive inappropriate empiric treatment (53.3% vs 5.7%, P<0.001) and to have been previously colonized by CR-PA (8.9% vs 0%, P=0.002) than BSIs caused by carbapenem-susceptible P. aeruginosa. CR was associated with carbapenem treatment in the previous month (adjusted odds ratio (aOR) 11.15) and solid organ transplantation (aOR 7.64). The 30-day mortality was 23.2%, which was associated with mechanical ventilation (aOR 4.24), sepsis (aOR 5.72), inappropriate empiric antibiotic therapy (aOR 5.86), and source control as a protective factor (aOR 0.16). CONCLUSION: This study shows a concerning prevalence of CR in children with PA-BSIs, leading to high mortality. Inappropriate empiric treatment and sepsis were associated with mortality. The high prevalence of CR with an increased risk of inappropriate empiric treatment should be closely monitored.


Subject(s)
Bacteremia , Carbapenems , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Pseudomonas Infections/mortality , Pseudomonas Infections/epidemiology , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Child, Preschool , Child , Risk Factors , Male , Female , Pseudomonas aeruginosa/drug effects , Retrospective Studies , Infant , Carbapenems/pharmacology , Carbapenems/therapeutic use , Adolescent , Bacteremia/mortality , Bacteremia/microbiology , Bacteremia/epidemiology , Bacteremia/drug therapy , Spain/epidemiology , Prevalence , Tertiary Care Centers/statistics & numerical data , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Survival Analysis , beta-Lactam Resistance
20.
Exp Clin Transplant ; 22(4): 300-306, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38742321

ABSTRACT

OBJECTIVES: In this study, we analyzed the effects of carbapenem-resistant Pseudomonas aeruginosa infection and mixed infection on the perioperative prognosis of lung transplant recipients and studied statistics on antibiotic resistance in P aeruginosa. MATERIALS AND METHODS: This was a retrospective casecontrol study. We collected data on lung transplant recipients with combined lower respiratory tract P aeruginosa infection within 48 hours after lung transplant at the China-Japan Friendship Hospital from August 2018 to April 2022. We grouped recipients according to P aeruginosa resistance to carbapenem antibiotics and summarized the clinical characteristics of carbapenem-resistant P aeruginosa infection. We analyzed the effects of carbapenemresistant P aeruginosa infection and mixed infections on all-cause mortality 30 days after lung transplant by Cox regression. We used the Kaplan-Meier method to plot survival curves. RESULTS: Patients in the carbapenem-resistant P aeruginosa group had a higher all-cause mortality rate than those in the carbapenem-sensitive P aeruginosa group at both 7 days (6 patients [22.3%] vs 2 patients [4.5%]; P = .022) and 30 days (12 patients [44.4%] vs 7 patients [15.9%]; P = .003) after lung transplant. In multivariate analysis, both carbapenemresistant P aeruginosa infection and P aeruginosa combined with bacterial infection were independent risk factors for death 30 days after transplant in lung transplant recipients (P < .05). In subgroup analysis, carbapenem-resistant P aeruginosa combined with bacterial infection increased the risk of death 30 days after transplant in lung transplant recipients compared with carbapenem-sensitive P aeruginosa combined with bacterial infection (12 patients [60%] vs 6 patients [19.4%]; P < .001). CONCLUSIONS: Combined lower respiratory tract carbapenem-resistant P aeruginosa infection and P aeruginosa combined with bacterial infection early after lung transplant increased the risk of 30-day mortality after lung transplant.


Subject(s)
Anti-Bacterial Agents , Carbapenems , Coinfection , Lung Transplantation , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/isolation & purification , Retrospective Studies , Pseudomonas Infections/mortality , Pseudomonas Infections/microbiology , Pseudomonas Infections/diagnosis , Pseudomonas Infections/drug therapy , Risk Factors , Lung Transplantation/adverse effects , Lung Transplantation/mortality , Carbapenems/pharmacology , Female , Male , Middle Aged , Time Factors , Anti-Bacterial Agents/therapeutic use , Adult , Treatment Outcome , Risk Assessment , beta-Lactam Resistance
SELECTION OF CITATIONS
SEARCH DETAIL