Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.057
Filter
1.
Proc Natl Acad Sci U S A ; 121(25): e2401831121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38875147

ABSTRACT

Ovoid-shaped bacteria, such as Streptococcus pneumoniae (pneumococcus), have two spatially separated peptidoglycan (PG) synthase nanomachines that locate zonally to the midcell of dividing cells. The septal PG synthase bPBP2x:FtsW closes the septum of dividing pneumococcal cells, whereas the elongasome located on the outer edge of the septal annulus synthesizes peripheral PG outward. We showed previously by sm-TIRFm that the septal PG synthase moves circumferentially at midcell, driven by PG synthesis and not by FtsZ treadmilling. The pneumococcal elongasome consists of the PG synthase bPBP2b:RodA, regulators MreC, MreD, and RodZ, but not MreB, and genetically associated proteins Class A aPBP1a and muramidase MpgA. Given its zonal location separate from FtsZ, it was of considerable interest to determine the dynamics of proteins in the pneumococcal elongasome. We found that bPBP2b, RodA, and MreC move circumferentially with the same velocities and durations at midcell, driven by PG synthesis. However, outside of the midcell zone, the majority of these elongasome proteins move diffusively over the entire surface of cells. Depletion of MreC resulted in loss of circumferential movement of bPBP2b, and bPBP2b and RodA require each other for localization and circumferential movement. Notably, a fraction of aPBP1a molecules also moved circumferentially at midcell with velocities similar to those of components of the core elongasome, but for shorter durations. Other aPBP1a molecules were static at midcell or diffusing over cell bodies. Last, MpgA displayed nonprocessive, subdiffusive motion that was largely confined to the midcell region and less frequently detected over the cell body.


Subject(s)
Bacterial Proteins , Penicillin-Binding Proteins , Streptococcus pneumoniae , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/genetics , Peptidoglycan/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Peptidoglycan Glycosyltransferase/genetics
2.
J Struct Biol ; 216(2): 108086, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38527711

ABSTRACT

Staphylococcus aureus, an ESKAPE pathogen, is a major clinical concern due to its pathogenicity and manifold antimicrobial resistance mechanisms. The commonly used ß-lactam antibiotics target bacterial penicillin-binding proteins (PBPs) and inhibit crosslinking of peptidoglycan strands that comprise the bacterial cell wall mesh, initiating a cascade of effects leading to bacterial cell death. S. aureus PBP1 is involved in synthesis of the bacterial cell wall during division and its presence is essential for survival of both antibiotic susceptible and resistant S. aureus strains. Here, we present X-ray crystallographic data for S. aureus PBP1 in its apo form as well as acyl-enzyme structures with distinct classes of ß-lactam antibiotics representing the penicillins, carbapenems, and cephalosporins, respectively: oxacillin, ertapenem and cephalexin. Our structural data suggest that the PBP1 active site is readily accessible for substrate, with little conformational change in key structural elements required for its covalent acylation of ß-lactam inhibitors. Stopped-flow kinetic analysis and gel-based competition assays support the structural observations, with even the weakest performing ß-lactams still having comparatively high acylation rates and affinities for PBP1. Our structural and kinetic analysis sheds insight into the ligand-PBP interactions that drive antibiotic efficacy against these historically useful antimicrobial targets and expands on current knowledge for future drug design and treatment of S. aureus infections.


Subject(s)
Penicillin-Binding Proteins , Staphylococcus aureus , Staphylococcus aureus/metabolism , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/genetics , Crystallography, X-Ray , Kinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , beta-Lactams/pharmacology , beta-Lactams/metabolism , beta-Lactams/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Catalytic Domain , Protein Conformation , Models, Molecular
3.
Antimicrob Agents Chemother ; 68(4): e0117923, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38415648

ABSTRACT

Streptococcus mitis/oralis group isolates with reduced carbapenem susceptibility have been reported, but its isolation rate in Japan is unknown. We collected 356 clinical α-hemolytic streptococcal isolates and identified 142 of them as S. mitis/oralis using partial sodA sequencing. The rate of meropenem non-susceptibility was 17.6% (25/142). All 25 carbapenem-non-susceptible isolates harbored amino acid substitutions in/near the conserved motifs in PBP1A, PBP2B, and PBP2X. Carbapenem non-susceptibility is common among S. mitis/oralis group isolates in Japan.


Subject(s)
Carbapenems , Streptococcus mitis , Penicillin-Binding Proteins/genetics , Streptococcus mitis/genetics , Streptococcus mitis/metabolism , Carbapenems/pharmacology , Japan , Amino Acid Substitution , Microbial Sensitivity Tests , Streptococcus/metabolism , Viridans Streptococci/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
4.
Antimicrob Agents Chemother ; 68(9): e0055524, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39058024

ABSTRACT

Enterococci exhibit intrinsic resistance to cephalosporins, mediated in part by the class B penicillin-binding protein (bPBP) Pbp4 that exhibits low reactivity toward cephalosporins and thus can continue crosslinking peptidoglycan despite exposure to cephalosporins. bPBPs partner with cognate SEDS (shape, elongation, division, and sporulation) glycosyltransferases to form the core catalytic complex of peptidoglycan synthases that synthesize peptidoglycan at discrete cellular locations, although the SEDS partner for Pbp4 is unknown. SEDS-bPBP peptidoglycan synthases of enterococci have not been studied, but some SEDS-bPBP pairs can be predicted based on sequence similarity. For example, FtsW (SEDS)-PbpB (bPBP) is predicted to form the catalytic core of the peptidoglycan synthase that functions at the division septum (the divisome). However, PbpB is readily inactivated by cephalosporins, raising the question-how could the FtsW-PbpB synthase continue functioning to enable growth in the presence of cephalosporins? In this work, we report that the FtsW-PbpB peptidoglycan synthase is required for cephalosporin resistance of Enterococcus faecalis, despite the fact that PbpB is inactivated by cephalosporins. Moreover, Pbp4 associates with the FtsW-PbpB synthase and the TPase activity of Pbp4 is required to enable growth in the presence of cephalosporins in an FtsW-PbpB-synthase-dependent manner. Overall, our results implicate a model in which Pbp4 directly interacts with the FtsW-PbpB peptidoglycan synthase to provide TPase activity during cephalosporin treatment, thereby maintaining the divisome SEDS-bPBP peptidoglycan synthase in a functional state competent to synthesize crosslinked peptidoglycan. These results suggest that two bPBPs coordinate within the FtsW-PbpB peptidoglycan synthase to drive cephalosporin resistance in E. faecalis.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Cephalosporin Resistance , Cephalosporins , Enterococcus faecalis , Penicillin-Binding Proteins , Peptidoglycan Glycosyltransferase , Peptidoglycan , Enterococcus faecalis/drug effects , Enterococcus faecalis/enzymology , Enterococcus faecalis/genetics , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Peptidoglycan Glycosyltransferase/metabolism , Peptidoglycan Glycosyltransferase/genetics , Cephalosporin Resistance/genetics , Cephalosporins/pharmacology , Anti-Bacterial Agents/pharmacology , Peptidoglycan/metabolism , Microbial Sensitivity Tests , Peptidyl Transferases/metabolism , Peptidyl Transferases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics
5.
Biochem Biophys Res Commun ; 729: 150368, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38986258

ABSTRACT

Penicillin-binding protein 2 (PBP2), a vital protein involved in bacterial cell-wall synthesis, serves a target for ß-lactam antibiotics. Acinetobacter baumannii is a pathogen notorious for multidrug resistance; therefore, exploration of PBPs is pivotal in the development of new antimicrobial strategies. In this study, the tertiary structure of PBP2 from A. baumannii (abPBP2) was elucidated using X-ray crystallography. The structural analysis demonstrated notable movement in the head domain, potentially critical for its glycosyltransferase function, suggesting that abPBP2 assumes a fully closed conformation. Our findings offer valuable information for developing novel antimicrobial agents targeting abPBP2 that are applicable in combating multidrug-resistant infections.


Subject(s)
Acinetobacter baumannii , Penicillin-Binding Proteins , Protein Conformation , Acinetobacter baumannii/metabolism , Acinetobacter baumannii/chemistry , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/genetics , Crystallography, X-Ray , Models, Molecular , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Amino Acid Sequence
6.
Small ; 20(24): e2311764, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38506607

ABSTRACT

The development of novel method for drug-resistant bacteria detection is imperative. A simultaneous dual-gene Test of methicillin-resistant Staphylococcus aureus (MRSA) is developed using an Argonaute-centered portable biosensor (STAR). This is the first report concerning Argonaute-based pathogenic bacteria detection. Simply, the species-specific mecA and nuc gene are isothermally amplified using loop-mediated isothermal amplification (LAMP) technique, followed by Argonaute-based detection enabled by its programmable, guided, sequence-specific recognition and cleavage. With the strategy, the targeted nucleic acid signals gene are dexterously converted into fluorescent signals. STAR is capable of detecting the nuc gene and mecA gene simultaneously in a single reaction. The limit of detection is 10 CFU/mL with a dynamic range from 10 to 107 CFU/mL. The sample-to-result time is <65 min. This method is successfully adapted to detect clinical samples, contaminated foods, and MRSA-infected animals. This work broadens the reach of Argonaute-based biosensing and presents a novel bacterial point-of-need (PON) detection platform.


Subject(s)
Biosensing Techniques , Methicillin-Resistant Staphylococcus aureus , Nucleic Acid Amplification Techniques , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Biosensing Techniques/methods , Nucleic Acid Amplification Techniques/methods , Argonaute Proteins/metabolism , Argonaute Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Penicillin-Binding Proteins/genetics , Animals , Micrococcal Nuclease/metabolism , Micrococcal Nuclease/genetics
7.
J Clin Microbiol ; 62(2): e0012023, 2024 02 14.
Article in English | MEDLINE | ID: mdl-38284761

ABSTRACT

Rapid phenotypic detection assays, including Carba NP and its variants, are widely applied for clinical diagnosis of carbapenemase-producing Enterobacterales (CPE). However, these tests are based on the acidification of the pH indicator during carbapenem hydrolysis, which limits test sensitivity and speed, especially for the detection of CPE producing low-activity carbapenem (e.g., OXA-48 variants). Herein, we developed a novel rapid and sensitive CPE detection method (Carba PBP) that could measure substrate (meropenem) consumption based on penicillin-binding protein (PBP). Meropenem-specific PBP was used to develop a competitive lateral flow assay (LFA) for meropenem identification. For the detection of carbapenemase activity, meropenem concentration was optimized using a checkerboard assay. The performance of Carba PBP was evaluated and compared with that of Carba NP using a panel of 94 clinical strains characterized by whole-genome sequencing and carbapenem susceptibility test. The limit of detection of PBP-based LFA for meropenem identification was 7 ng mL-1. Using 10 ng mL-1 meropenem as the substrate, Carba PBP and Carba NP could detect 10 ng mL-1 carbapenemase within 25 min and 1,280 ng mL-1 CPE in 2 h, respectively. The sensitivity and specificity were 100% (75/75) and 100% (19/19) for Carba PBP and 85.3% (64/75) and 100% (19/19) for Carba NP, respectively. When compared with Carba NP, Carba PBP showed superior performance in detecting all the tested CPE strains (including OXA-48-like variants) within 25 min and presented two orders of magnitude higher analytical sensitivity, demonstrating potential for clinical diagnosis of CPE. IMPORTANCE This study successfully achieved the goal of carbapenemase activity detection with both high sensitivity and convenience, offering a convenient lateral flow assay for clinical diagnosis of carbapenemase-producing Enterobacterales.


Subject(s)
Bacterial Proteins , beta-Lactamases , Humans , Penicillin-Binding Proteins/genetics , Meropenem/pharmacology , Microbial Sensitivity Tests , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , beta-Lactamases/metabolism , Carbapenems/pharmacology , Sensitivity and Specificity
8.
J Antimicrob Chemother ; 79(2): 403-411, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38153239

ABSTRACT

BACKGROUND: Streptococcus suis is an important pig pathogen and an emerging zoonotic agent. In a previous study, we described a high proportion of penicillin-resistant serotype 9 S. suis (SS9) isolates on pig farms in Italy. OBJECTIVES: We hypothesized that resistance to penicillin emerged in some SS9 lineages characterized by substitutions at the PBPs, contributing to the successful spread of these lineages in the last 20 years. METHODS: Sixty-six SS9 isolates from cases of streptococcosis in pigs were investigated for susceptibility to penicillin, ceftiofur and ampicillin. The isolates were characterized for ST, virulence profile, and antimicrobial resistance genes through WGS. Multiple linear regression models were employed to investigate the associations between STs, year of isolation, substitutions at the PBPs and an increase in MIC values to ß-lactams. RESULTS: MIC values to penicillin increased by 4% each year in the study period. Higher MIC values for penicillin were also positively associated with ST123, ST1540 and ST1953 compared with ST16. The PBP sequences presented a mosaic organization of blocks. Within the same ST, substitutions at the PBPs were generally more frequent in recent isolates. Resistance to penicillin was driven by substitutions at PBP2b, including K479T, D512E and K513E, and PBP2x, including T551S, while reduced susceptibility to ceftiofur and ampicillin were largely dependent on substitutions at PBP2x. CONCLUSIONS: Here, we identify the STs and substitutions at the PBPs responsible for increased resistance of SS9 to penicillin on Italian pig farms. Our data highlight the need for monitoring the evolution of S. suis in the coming years.


Subject(s)
Aminoacyltransferases , Cephalosporins , Streptococcus suis , Animals , Swine , Penicillins/pharmacology , Penicillin-Binding Proteins/genetics , Streptococcus suis/genetics , Bacterial Proteins/genetics , Streptococcus pneumoniae/genetics , Serogroup , Aminoacyltransferases/genetics , Microbial Sensitivity Tests , Penicillin Resistance/genetics , Genomics , Ampicillin , Clone Cells , Anti-Bacterial Agents/pharmacology
9.
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
10.
PLoS Pathog ; 18(7): e1010727, 2022 07.
Article in English | MEDLINE | ID: mdl-35877768

ABSTRACT

Understanding how antimicrobial resistance spreads is critical for optimal application of new treatments. In the naturally competent human pathogen Streptococcus pneumoniae, resistance to ß-lactam antibiotics is mediated by recombination events in genes encoding the target proteins, resulting in reduced drug binding affinity. However, for the front-line antibiotic amoxicillin, the exact mechanism of resistance still needs to be elucidated. Through successive rounds of transformation with genomic DNA from a clinically resistant isolate, we followed amoxicillin resistance development. Using whole genome sequencing, we showed that multiple recombination events occurred at different loci during one round of transformation. We found examples of non-contiguous recombination, and demonstrated that this could occur either through multiple D-loop formation from one donor DNA molecule, or by the integration of multiple DNA fragments. We also show that the final minimum inhibitory concentration (MIC) differs depending on recipient genome, explained by differences in the extent of recombination at key loci. Finally, through back transformations of mutant alleles and fluorescently labelled penicillin (bocillin-FL) binding assays, we confirm that pbp1a, pbp2b, pbp2x, and murM are the main resistance determinants for amoxicillin resistance, and that the order of allele uptake is important for successful resistance evolution. We conclude that recombination events are complex, and that this complexity contributes to the highly diverse genotypes of amoxicillin-resistant pneumococcal isolates.


Subject(s)
Amoxicillin , Streptococcus pneumoniae , Amoxicillin/metabolism , Amoxicillin/pharmacology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Transfer, Horizontal , Humans , Microbial Sensitivity Tests , Penicillin Resistance/genetics , Penicillin-Binding Proteins/genetics , Streptococcus pneumoniae/metabolism
11.
Mol Biol Rep ; 51(1): 761, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874884

ABSTRACT

BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA) poses a great health threat to humans. Looking for compounds that could reduce the resistance of S. aureus towards methicillin is an effective way to alleviate the antimicrobial resistance crisis. METHODS AND RESULTS: Minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), Time-killing growth curve, staphyloxanthin and penicillin-binding protein 2a (PBP2a) were detected. A quantitative polymerase chain reaction was used to measure the effect of BBH on the gene transcription profiles of MRSA. The MIC of MRSA-ST59-t437 towards oxacillin was 8 µg/ml, and MBC was 128 µg/ml. After adding a sub-inhibitory concentration of BBH, the MIC and MBC of MRSA-ST59-t478 towards oxacillin went down to 0.125 and 32 µg/ml respectively. The amount of PBP2a and staphyloxanthin were reduced after treatment with BBH. Moreover, the transcription levels of sarA, mecA and fni genes were downregulated. CONCLUSIONS: It is for the first time reported that BBH could inhibit staphyloxanthin synthesis by inhibiting fni gene. Moreover, fni might be the target gene of sarA, and there might be another regulatory pathway to inhibit staphyloxanthin biosynthesis. BBH could effectively reduce the methicillin resistance of MRSA-ST59-t437 by downregulating fni, sarA and mecA genes.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Berberine , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Xanthophylls , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Xanthophylls/pharmacology , Berberine/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Oxacillin/pharmacology
12.
Mol Biol Rep ; 51(1): 665, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777940

ABSTRACT

BACKGROUND: Staphylococcus aureus (S. aureus) associated with COVID-19 has not been well documented. This cross-sectional study evaluated the association between nasal S. aureus carriage and COVID-19. METHODS AND RESULTS: Nasopharyngeal samples were collected from 391 participants presenting for COVID-19 test in Lagos, Nigeria, and S. aureus was isolated from the samples. Antimicrobial susceptibility test was done by disc diffusion method. All S. aureus isolates were screened for the presence of mecA, panton-valentine leucocidin (PVL) and toxic shock syndrome toxin (TSST) virulence genes by polymerase chain reaction. Staphylococcal protein A (spa) typing was conducted for all the isolates. Participants with COVID-19 had double the prevalence of S. aureus (42.86%) compared to those who tested negative (20.54%). A significant association was seen between S. aureus nasal carriage and COVID-19 (p = 0.004). Antimicrobial sensitivity results showed resistance to oxacillin (100%), cefoxitin (53%), and vancomycin (98.7%). However, only 41% of the isolates harbored the mecA gene, with SCCmecV being the most common SCCmec type. There was no association between the carriage of virulence genes and COVID-19. A total of 23 Spa types were detected, with t13249 and t095 being the two most common spa types. CONCLUSION: This study examined the association between nasal S. aureus carriage and SARS-COV-2 infection. Further research is required to fully explore the implications of S. aureus co-infection with COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Staphylococcal Infections , Staphylococcus aureus , Humans , COVID-19/microbiology , COVID-19/epidemiology , COVID-19/virology , Staphylococcal Infections/epidemiology , Staphylococcal Infections/microbiology , Cross-Sectional Studies , Male , Female , Staphylococcus aureus/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/isolation & purification , Adult , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Middle Aged , Bacterial Toxins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Methicillin-Resistant Staphylococcus aureus/drug effects , Comorbidity , Bacterial Proteins/genetics , Virulence/genetics , Nigeria/epidemiology , Drug Resistance, Multiple, Bacterial/genetics , Anti-Bacterial Agents/pharmacology , Carrier State/epidemiology , Carrier State/microbiology , Microbial Sensitivity Tests , Penicillin-Binding Proteins/genetics , Leukocidins/genetics , Exotoxins/genetics , Virulence Factors/genetics , Young Adult
13.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34429361

ABSTRACT

A cell wall made of the heteropolymer peptidoglycan (PG) surrounds most bacterial cells. This essential surface layer is required to prevent lysis from internal osmotic pressure. The class A penicillin-binding proteins (aPBPs) play key roles in building the PG network. These bifunctional enzymes possess both PG glycosyltransferase (PGT) and transpeptidase (TP) activity to polymerize the wall glycans and cross-link them, respectively. In Escherichia coli and other gram-negative bacteria, aPBP function is dependent on outer membrane lipoproteins. The lipoprotein LpoA activates PBP1a and LpoB promotes PBP1b activity. In a purified system, the major effect of LpoA on PBP1a is TP stimulation. However, the relevance of this activation to the cellular function of LpoA has remained unclear. To better understand why PBP1a requires LpoA for its activity in cells, we identified variants of PBP1a from E. coli and Pseudomonas aeruginosa that function in the absence of the lipoprotein. The changes resulting in LpoA bypass map to the PGT domain and the linker region between the two catalytic domains. Purification of the E. coli variants showed that they are hyperactivated for PGT but not TP activity. Furthermore, in vivo analysis found that LpoA is necessary for the glycan synthesis activity of PBP1a in cells. Thus, our results reveal that LpoA exerts a much greater control over the cellular activity of PBP1a than previously appreciated. It not only modulates PG cross-linking but is also required for its cognate synthase to make PG glycans in the first place.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Cell Wall/enzymology , Cross-Linking Reagents/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Lipoproteins/metabolism , Penicillin-Binding Proteins/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Peptidoglycan/metabolism , Bacterial Outer Membrane Proteins/genetics , Cross-Linking Reagents/metabolism , Escherichia coli Proteins/genetics , Lipoproteins/genetics , Penicillin-Binding Proteins/genetics , Peptidoglycan Glycosyltransferase/genetics
14.
PLoS Genet ; 17(4): e1009366, 2021 04.
Article in English | MEDLINE | ID: mdl-33857142

ABSTRACT

SEDS family peptidoglycan (PG) glycosyltransferases, RodA and FtsW, require their cognate transpeptidases PBP2 and FtsI (class B penicillin binding proteins) to synthesize PG along the cell cylinder and at the septum, respectively. The activities of these SEDS-bPBPs complexes are tightly regulated to ensure proper cell elongation and division. In Escherichia coli FtsN switches FtsA and FtsQLB to the active forms that synergize to stimulate FtsWI, but the exact mechanism is not well understood. Previously, we isolated an activation mutation in ftsW (M269I) that allows cell division with reduced FtsN function. To try to understand the basis for activation we isolated additional substitutions at this position and found that only the original substitution produced an active mutant whereas drastic changes resulted in an inactive mutant. In another approach we isolated suppressors of an inactive FtsL mutant and obtained FtsWE289G and FtsIK211I and found they bypassed FtsN. Epistatic analysis of these mutations and others confirmed that the FtsN-triggered activation signal goes from FtsQLB to FtsI to FtsW. Mapping these mutations, as well as others affecting the activity of FtsWI, on the RodA-PBP2 structure revealed they are located at the interaction interface between the extracellular loop 4 (ECL4) of FtsW and the pedestal domain of FtsI (PBP3). This supports a model in which the interaction between the ECL4 of SEDS proteins and the pedestal domain of their cognate bPBPs plays a critical role in the activation mechanism.


Subject(s)
Bacterial Proteins/ultrastructure , Escherichia coli Proteins/ultrastructure , Membrane Proteins/ultrastructure , Multiprotein Complexes/ultrastructure , Penicillin-Binding Proteins/ultrastructure , Peptidoglycan Glycosyltransferase/ultrastructure , Protein Conformation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/genetics , Peptidoglycan/chemistry , Peptidoglycan/genetics , Peptidoglycan/ultrastructure , Peptidoglycan Glycosyltransferase/chemistry , Peptidoglycan Glycosyltransferase/genetics , Peptidyl Transferases/chemistry , Peptidyl Transferases/genetics , Peptidyl Transferases/ultrastructure
15.
J Korean Med Sci ; 39(15): e136, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38651222

ABSTRACT

BACKGROUND: Haemophilus influenzae is a frequently encountered pathogen responsible for respiratory tract infections in children. Following the detection of ceftriaxone-resistant H. influenzae at our institution, we aimed to investigate the resistance mechanisms of ceftriaxone in H. influenzae, with a particular focus on alterations in penicillin-binding protein 3 (PBP3) and ß-lactamase production. METHODS: Among H. influenzae isolates collected at Asan Medical Center Children's Hospital from March 2014 to April 2019, ceftriaxone-resistant strains by the disk-diffusion test were included. Ceftriaxone minimum inhibitory concentrations (MICs) were determined using the E-test according to the European Committee on Antimicrobial Susceptibility Testing (EUCAST) guidelines. The presence of ß-lactamase was assessed through cefinase test and TEM-1/ROB-1 polymerase chain reaction (PCR). PBP3 alterations were explored via ftsI gene sequencing. RESULTS: Out of the 68 collected strains, 21 exhibited resistance to ceftriaxone in disk diffusion tests. Two strains were excluded due to failed subculture. Among 19 ceftriaxone-resistant H. influenzae isolates, eighteen were non-typeable H. influenzae, and twelve were positive for TEM-1 PCR. Isolates were classified into groups II (harboring only N526K, n = 3), III (N526K+S385T, n = 2), III+ (S385T+L389F+N526K, n = 11), and III-like+ (S385T+L389F+R517H, n = 3) according to the PBP3 alteration pattern. With a median ceftriaxone MIC of 0.190 mg/L (range, 0.008-0.750), the median ceftriaxone MIC was the highest in group III-like+ (0.250 mg/L), followed by groups III+ (0.190 mg/L), III (0.158 mg/L), and II (0.012 mg/L). All three strains belonging to group II, which did not harbor the S385T substitution, had ceftriaxone MICs of ≤ 0.125 mg/L. CONCLUSION: The emergence of ceftriaxone-resistant H. influenzae with ceftriaxone MIC values of up to 0.75 mg/L was observed even in children in South Korea, with most associated with S385T and L389F substitutions. The N526K mutation alone does not significantly impact ceftriaxone resistance. Further large-scale studies are essential to investigate changes in antibiotic resistance patterns and factors influencing antibiotic resistance in H. influenzae isolated from pediatric patients in Korea.


Subject(s)
Anti-Bacterial Agents , Ceftriaxone , Haemophilus Infections , Haemophilus influenzae , Microbial Sensitivity Tests , beta-Lactamases , Ceftriaxone/pharmacology , Haemophilus influenzae/drug effects , Haemophilus influenzae/isolation & purification , Haemophilus influenzae/genetics , Humans , Anti-Bacterial Agents/pharmacology , Republic of Korea , beta-Lactamases/genetics , beta-Lactamases/metabolism , Child , Haemophilus Infections/microbiology , Haemophilus Infections/drug therapy , Penicillin-Binding Proteins/genetics , Child, Preschool , Drug Resistance, Bacterial , Infant , Female , Male , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
16.
Int J Mol Sci ; 25(2)2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38256057

ABSTRACT

Mycobacterium tuberculosis, a major cause of mortality from a single infectious agent, possesses a remarkable mycobacterial cell envelope. Penicillin-Binding Proteins (PBPs) are a family of bacterial enzymes involved in the biosynthesis of peptidoglycan. PBP4 (DacB) from M. tuberculosis (MtbPBP4) has been known to function as a carboxypeptidase, and the role and significance of carboxypeptidases as targets for anti-tuberculosis drugs or antibiotics have been extensively investigated over the past decade. However, their precise involvement remains incompletely understood. In this study, we employed predictive modeling and analyzed the three-dimensional structure of MtbPBP4. Interestingly, MtbPBP4 displayed a distinct domain structure compared to its homologs. Docking studies with meropenem verified the presence of active site residues conserved in PBPs. These findings establish a structural foundation for comprehending the molecular function of MtbPBP4 and offer a platform for the exploration of novel antibiotics.


Subject(s)
Mycobacterium tuberculosis , Penicillin-Binding Proteins/genetics , Antitubercular Agents , Cell Membrane , Cell Wall
17.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39125830

ABSTRACT

The increase in the resistance of mutant strains of Neisseria gonorrhoeae to the antibiotic ceftriaxone is pronounced in the decrease in the second-order acylation rate constant, k2/KS, by penicillin-binding protein 2 (PBP2). These changes can be caused by both the decrease in the acylation rate constant, k2, and the weakening of the binding affinity, i.e., an increase in the substrate constant, KS. A501X mutations in PBP2 affect second-order acylation rate constants. The PBP2A501V variant exhibits a higher k2/KS value, whereas for PBP2A501R and PBP2A501P variants, these values are lower. We performed molecular dynamic simulations with both classical and QM/MM potentials to model both acylation energy profiles and conformational dynamics of four PBP2 variants to explain the origin of k2/KS changes. The acylation reaction occurs in two elementary steps, specifically, a nucleophilic attack by the oxygen atom of the Ser310 residue and C-N bond cleavage in the ß-lactam ring accompanied by the elimination of the leaving group of ceftriaxone. The energy barrier of the first step increases for PBP2 variants with a decrease in the observed k2/KS value. Submicrosecond classic molecular dynamic trajectories with subsequent cluster analysis reveal that the conformation of the ß3-ß4 loop switches from open to closed and its flexibility decreases for PBP2 variants with a lower k2/KS value. Thus, the experimentally observed decrease in the k2/KS in A501X variants of PBP2 occurs due to both the decrease in the acylation rate constant, k2, and the increase in KS.


Subject(s)
Ceftriaxone , Molecular Dynamics Simulation , Neisseria gonorrhoeae , Penicillin-Binding Proteins , Ceftriaxone/pharmacology , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/drug effects , Neisseria gonorrhoeae/metabolism , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Mutation , Drug Resistance, Bacterial/genetics , Acylation , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Serine-Type D-Ala-D-Ala Carboxypeptidase
18.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731965

ABSTRACT

Antimicrobial resistance has recently been considered an emerging catastrophe globally. The public health and environmental threats were aggravated by the injudicious use of antibiotics in animal farming, aquaculture, and croup fields, etc. Consequently, failure of antibiotic therapies is common because of the emergence of multidrug-resistant (MDR) bacteria in the environment. Thus, the reduction in antibiotic spillage in the environment could be an important step for overcoming this situation. Bear in mind, this research was focused on the green synthesis of chitosan nanoparticles (ChiNPs) using Citrus lemon (Assam lemon) extract as a cross-linker and application in controlling MDR bacteria to reduce the antibiotic spillage in that sector. For evaluating antibacterial activity, Staphylococcus aureus and Escherichia coli were isolated from environmental specimens, and their multidrug-resistant pattern were identified both phenotypically by disk diffusion and genotypically by detecting methicillin- (mecA), penicillin- (blaZ), and streptomycin (aadA1)-resistance encoding genes. The inhibitory zone's diameter was employed as a parameter for determining the antibacterial effect against MDR bacteria revealing 30 ± 0.4 mm, 34 ± 0.2 mm, and 36 ± 0.8 mm zones of inhibition against methicillin- (mecA) and penicillin (blaZ)-resistant S. aureus, and streptomycin (aadA1)-resistant E. coli, respectively. The minimum inhibitory concentration at 0.31 mg/mL and minimum bactericidal concentration at 0.62 mg/mL of yielded ChiNPs were used as the broad-spectrum application against MDR bacteria. Finally, the biocompatibility of ChiNPs was confirmed by showing a negligible decrease in BHK-21 cell viability at doses less than 2 MIC, suggesting their potential for future application in antibiotic-free farming practices.


Subject(s)
Anti-Bacterial Agents , Chitosan , Drug Resistance, Multiple, Bacterial , Escherichia coli , Nanoparticles , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Chitosan/pharmacology , Chitosan/chemistry , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Green Chemistry Technology , Microbial Sensitivity Tests , Nanoparticles/chemistry , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Penicillin-Binding Proteins/antagonists & inhibitors , Staphylococcus aureus/drug effects
19.
Antimicrob Agents Chemother ; 67(1): e0103322, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36475717

ABSTRACT

Limited therapeutic options dictate the need for new classes of antimicrobials active against carbapenem-resistant Acinetobacter baumannii. Presented data confirm and extend penicillin binding protein 7/8 (PBP 7/8) as a high-value target in the CR A. baumannii strain HUMC1. PBP 7/8 was essential for optimal growth/survival of HUMC1 in ex vivo human ascites and in a rat subcutaneous abscess model; in a mouse pneumonia model, the absence of PBP 7/8 decreased lethality 11-fold. The loss of PBP 7/8 resulted in increased permeability, sensitivity to complement, and lysozyme-mediated bactericidal activity. These changes did not appear to be due to alterations in the cellular fatty acid composition or capsule production. However, a decrease in lipid A and an increase in coccoidal cells and cell aggregation were noted. The compromise of the stringent permeability barrier in the PBP 7/8 mutant was reflected by an increased susceptibility to several antimicrobials. Importantly, expression of ampC was not significantly affected by the loss of PBP 7/8 and serial passage of the mutant strain in human ascites over 7 days did not yield revertants possessing a wild-type phenotype. In summary, these data and other features support PBP 7/8 as a high-value drug target for extensively drug-resistant and CR A. baumannii. Our results guide next-stage studies; the determination that the inactivation of PBP 7/8 results in an increased sensitivity to lysozyme enables the design of a high-throughput screening assay to identify small molecule compounds that can specifically inhibit PBP 7/8 activity.


Subject(s)
Acinetobacter baumannii , Mice , Animals , Humans , Rats , Penicillin-Binding Proteins/genetics , Acinetobacter baumannii/genetics , Acinetobacter baumannii/metabolism , Muramidase/metabolism , Ascites , Microbial Sensitivity Tests , Carbapenems/pharmacology , Carbapenems/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism
20.
Antimicrob Agents Chemother ; 67(10): e0071623, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37655923

ABSTRACT

Acquisition of PBP2a (encoded by the mec gene) is the key resistance mechanism to ß-lactams in Staphylococcus aureus. The mec gene can be easily detected by PCR assays; however, these tools will miss mec-independent oxacillin resistance. This phenotype is mediated by mutations in cell wall metabolism genes that can be acquired during persistent infections under prolonged antibiotic exposure. The complex case presented by Hess et al. (Antimicrob Agents Chemother 67:e00437-23, 2023, https://doi.org/10.1128/aac.00437-23) highlights the diagnostic and therapeutic challenges in the management of mec-independent oxacillin resistance.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Humans , Oxacillin/pharmacology , Oxacillin/therapeutic use , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Methicillin-Resistant Staphylococcus aureus/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Staphylococcal Infections/drug therapy , Microbial Sensitivity Tests , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL