Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Microbiol Spectr ; 10(5): e0270022, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36214681

ABSTRACT

In the current scenario of growing antibiotic resistance, understanding the interplay between resistance mechanisms and biological costs is crucial for designing therapeutic strategies. In this regard, intrinsic AmpC ß-lactamase hyperproduction is probably the most important resistance mechanism of Pseudomonas aeruginosa, proven to entail important biological burdens that attenuate virulence mostly under peptidoglycan recycling alterations. P. aeruginosa can acquire resistance to new ß-lactam-ß-lactamase inhibitor combinations (ceftazidime-avibactam and ceftolozane-tazobactam) through mutations affecting ampC and its regulatory genes, but the impact of these mutations on the associated biological cost and the role that ß-lactamase activity plays per se in contributing to the above-mentioned virulence attenuation are unknown. The same questions remain unsolved for plasmid-encoded AmpC-type ß-lactamases such as FOX enzymes, some of which also provide resistance to new ß-lactam-ß-lactamase inhibitor combinations. Here, we assessed from different perspectives the effects of changes in the active center and, thus, in the hydrolytic spectrum resistance to inhibitors of AmpC-type ß-lactamases on the fitness and virulence of P. aeruginosa, using site-directed mutagenesis; the previously described AmpC variants T96I, G183D, and ΔG229-E247; and, finally, blaFOX-4 versus blaFOX-8. Our results indicate the essential role of AmpC activity per se in causing the reported full virulence attenuation (in terms of growth, motility, cytotoxicity, and Galleria mellonella larvae killing), although the biological cost of the above-mentioned AmpC-type variants was similar to that of the wild-type enzymes. This suggests that there is not an important biological burden that may limit the selection/spread of these variants, which could progressively compromise the future effectiveness of the above-mentioned drug combinations. IMPORTANCE The growing antibiotic resistance of the top nosocomial pathogen Pseudomonas aeruginosa pushes research to explore new therapeutic strategies, for which the resistance-versus-virulence balance is a promising source of targets. While resistance often entails significant biological costs, little is known about the bases of the virulence attenuations associated with a resistance mechanism as extraordinarily relevant as ß-lactamase production. We demonstrate that besides potential energy and cell wall alterations, the enzymatic activity of the P. aeruginosa cephalosporinase AmpC is essential for causing the full attenuation associated with its hyperproduction by affecting different features related to pathogenesis, a fact exploitable from the antivirulence perspective. Less encouraging, we also show that the production of different chromosomal/plasmid-encoded AmpC derivatives conferring resistance to some of the newest antibiotic combinations causes no significantly increased biological burdens, which suggests a free way for the selection/spread of these types of variants, potentially compromising the future effectiveness of these antipseudomonal therapies.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , Cephalosporinase/metabolism , Cephalosporinase/pharmacology , Cephalosporinase/therapeutic use , Peptidoglycan/metabolism , Microbial Sensitivity Tests , beta-Lactamases/genetics , Tazobactam/metabolism , Tazobactam/pharmacology , Tazobactam/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cephalosporins/pharmacology , Cephalosporins/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Drug Combinations , Pseudomonas Infections/drug therapy
2.
J Proteomics ; 268: 104715, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36058541

ABSTRACT

Carbapenem-resistant Klebsiella pneumoniae (CRKP) is an urgent threat to human health. Major outer membrane proteins (OMPs) porin mutation is one important resistance mechanism of CRKP, and may also affect the inhibition activity of ß-lactam and ß-lactamase inhibitor combinations. The ertapenem-resistant K. pneumoniae strain 2018B120 with major porin mutations was isolated from a clinical patient. Genomic and time-series proteomic analyses were conducted to retrieve the ertapenem-challenged response of 2018B120. The abundance changing of proteins from PTS systems,  ABC transporters, the autoinducer 2 (AI-2) quorum sensing system, and antioxidant systems can be observed. Overexpression of alternative porins was also noticed to balance major porins' defection. These findings added a detailed regulation network in bacterial resistance mechanisms and gave new insights into bypass adaptation mechanisms the porin deficient bacteria adopted under carbapenem antibiotics pressure. SIGNIFICANCE: Outer membrane porins deficiency is an important mechanism of carbapenem resistance in K. pneumoniae. Comprehensive genomic and proteomic profiling of an ertapenem-resistant K. pneumoniae strain 2018B120 gives a detailed systematic regulation network in bacterial resistance mechanisms. Overexpression of alternative porins to balance major porins' defection was noticed, giving new insights into bypass adaptation mechanisms of porin deficient bacteria.


Subject(s)
Klebsiella pneumoniae , Porins , beta-Lactam Resistance , ATP-Binding Cassette Transporters/metabolism , Anti-Bacterial Agents/pharmacology , Antioxidants/metabolism , Bacterial Proteins/metabolism , Carbapenems/metabolism , Carbapenems/pharmacology , Ertapenem/metabolism , Ertapenem/pharmacology , Humans , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Microbial Sensitivity Tests , Porins/genetics , Porins/metabolism , Proteomics/methods , beta-Lactam Resistance/genetics , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/genetics , beta-Lactamases/metabolism , beta-Lactams/metabolism , beta-Lactams/pharmacology
3.
Eur J Med Chem ; 240: 114571, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-35792385

ABSTRACT

Inhibition of ß-lactamases is a promising strategy to overcome antimicrobial resistance to commonly used ß-lactam antibiotics. Boronic acid derivatives have proven to be effective inhibitors of ß-lactamases due to their direct interaction with the catalytic site of these enzymes. We synthesized a series of phenylboronic acid derivatives and evaluated their structure-activity relationships as Klebsiella pneumoniae carbapenemase (KPC-2) inhibitors. We identified potent KPC-2 inhibitors 2e & 6c (Ki = 0.032 µM and 0.038 µM, respectively) that enhance the activity of cefotaxime in KPC-2 expressing Escherichia coli. The measured acid dissociation constants (pKa) of selected triazole-containing phenylboronic acids was broad (5.98-10.0), suggesting that this is an additional property of the compounds that could be tuned to optimize the target interaction and/or the physicochemical properties of the compounds. These findings will help to guide the future development of boronic acid compounds as inhibitors of KPC-2 and other target proteins.


Subject(s)
Klebsiella pneumoniae , Triazoles , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Boronic Acids/chemistry , Drug Resistance, Microbial , Escherichia coli , Microbial Sensitivity Tests , Triazoles/pharmacology , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism
4.
Nature ; 602(7896): 343-348, 2022 02.
Article in English | MEDLINE | ID: mdl-35110734

ABSTRACT

Carbapenems are antibiotics of last resort in the clinic. Owing to their potency and broad-spectrum activity, they are an important part of the antibiotic arsenal. The vital role of carbapenems is exemplified by the approval acquired by Merck from the US Food and Drug Administration (FDA) for the use of an imipenem combination therapy to treat the increased levels of hospital-acquired and ventilator-associated bacterial pneumonia that have occurred during the COVID-19 pandemic1. The C6 hydroxyethyl side chain distinguishes the clinically used carbapenems from the other classes of ß-lactam antibiotics and is responsible for their low susceptibility to inactivation by occluding water from the ß-lactamase active site2. The construction of the C6 hydroxyethyl side chain is mediated by cobalamin- or B12-dependent radical S-adenosylmethionine (SAM) enzymes3. These radical SAM methylases (RSMTs) assemble the alkyl backbone by sequential methylation reactions, and thereby underlie the therapeutic usefulness of clinically used carbapenems. Here we present X-ray crystal structures of TokK, a B12-dependent RSMT that catalyses three-sequential methylations during the biosynthesis of asparenomycin A. These structures, which contain the two metallocofactors of the enzyme and were determined in the presence and absence of a carbapenam substrate, provide a visualization of a B12-dependent RSMT that uses the radical mechanism that is shared by most of these enzymes. The structures provide insight into the stereochemistry of initial C6 methylation and suggest that substrate positioning governs the rate of each methylation event.


Subject(s)
Carbapenems/biosynthesis , Methyltransferases/chemistry , Methyltransferases/metabolism , S-Adenosylmethionine/metabolism , Streptomyces/enzymology , Thienamycins/biosynthesis , Vitamin B 12/metabolism , Binding Sites , Biocatalysis , Coenzymes/metabolism , Crystallography, X-Ray , Kinetics , Methylation , Models, Molecular , Protein Binding , Protein Domains , Streptomyces/metabolism , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/chemistry , beta-Lactamases/metabolism
5.
Chembiochem ; 23(3): e202100504, 2022 02 04.
Article in English | MEDLINE | ID: mdl-34821011

ABSTRACT

Disrupting protein-protein interactions is difficult due to the large and flat interaction surfaces of the binding partners. The BLIP and BLIP-II proteins are unrelated in sequence and structure and yet each potently inhibit ß-lactamases. High-throughput oligonucleotide synthesis was used to construct a 12,470-member library containing overlapping linear and cyclic peptides ranging in size from 6 to 21 amino acids that scan through the sequences of BLIP and BLIP-II. Phage display affinity selections and deep sequencing revealed that, despite the differences in interaction surfaces with ß-lactamases, rapid enrichment of consensus peptide regions originating from both BLIP and BLIP-II contact residues in the binding interface occurred. BLIP and BLIP-II peptides that were enriched by affinity selection were shown to bind ß-lactamases and disrupt the BLIP/ß-lactamase interaction. The results suggest that peptides that bind at and disrupt PPI interfaces can be identified through systematic peptide library construction, affinity selection, and deep sequencing.


Subject(s)
Bacterial Proteins/metabolism , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/metabolism , Bacterial Proteins/chemistry , Models, Molecular , Peptide Library , Protein Binding , Streptomyces/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry
6.
Nat Chem ; 14(1): 15-24, 2022 01.
Article in English | MEDLINE | ID: mdl-34903857

ABSTRACT

Carbapenems are vital antibiotics, but their efficacy is increasingly compromised by metallo-ß-lactamases (MBLs). Here we report the discovery and optimization of potent broad-spectrum MBL inhibitors. A high-throughput screen for NDM-1 inhibitors identified indole-2-carboxylates (InCs) as potential ß-lactamase stable ß-lactam mimics. Subsequent structure-activity relationship studies revealed InCs as a new class of potent MBL inhibitor, active against all MBL classes of major clinical relevance. Crystallographic studies revealed a binding mode of the InCs to MBLs that, in some regards, mimics that predicted for intact carbapenems, including with respect to maintenance of the Zn(II)-bound hydroxyl, and in other regards mimics binding observed in MBL-carbapenem product complexes. InCs restore carbapenem activity against multiple drug-resistant Gram-negative bacteria and have a low frequency of resistance. InCs also have a good in vivo safety profile, and when combined with meropenem show a strong in vivo efficacy in peritonitis and thigh mouse infection models.


Subject(s)
beta-Lactamase Inhibitors/pharmacology , beta-Lactams/metabolism , Animals , Gram-Negative Bacteria/drug effects , Humans , Mice , Microbial Sensitivity Tests , Protein Binding , Structure-Activity Relationship , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/metabolism
7.
J Am Chem Soc ; 143(50): 21127-21142, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34860516

ABSTRACT

The rising prevalence of multidrug-resistant bacteria is an urgent health crisis that can only be countered through renewed investment in the discovery and development of antibiotics. There is no panacea for the antibacterial resistance crisis; instead, a multifaceted approach is called for. In this Perspective we make the case that, in the face of evolving clinical needs and enabling technologies, numerous validated antibacterial targets and associated lead molecules deserve a second look. At the same time, many worthy targets lack good leads despite harboring druggable active sites. Creative and inspired techniques buoy discovery efforts; while soil screening efforts frequently lead to antibiotic rediscovery, researchers have found success searching for new antibiotic leads by studying underexplored ecological niches or by leveraging the abundance of available data from genome mining efforts. The judicious use of "polypharmacology" (i.e., the ability of a drug to alter the activities of multiple targets) can also provide new opportunities, as can the continued search for inhibitors of resistance enzymes with the capacity to breathe new life into old antibiotics. We conclude by highlighting available pharmacoeconomic models for antibacterial discovery and development while making the case for new ones.


Subject(s)
Anti-Bacterial Agents/chemistry , Drug Discovery , Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/metabolism , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Drug Resistance, Multiple, Bacterial/drug effects , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/metabolism , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/chemistry , beta-Lactamases/metabolism
8.
Eur J Med Chem ; 223: 113667, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34225181

ABSTRACT

Bacteria carrying New Delhi metallo-ß-lactamase-1 (New Delhi metallo-ß-lactamase, NDM-1) resistance gene is a new type of "superbug", which can hydrolyze almost all ß-lactam antibiotics, rapidly spread among the same species and even spread among different species. NDM-1 belongs to the class B1 broad-spectrum enzyme of ß-lactamase. The two positively charged zinc ions in the active center have electrostatic interaction with the hydroxyl ions in them to seize the hydrogen atom near the water molecule to form a bridging ring water molecule, which strengthens its nucleophilicity and attacks the carbonyl group on the lactam ring; thus, catalyzing the hydrolysis of ß-lactam antibiotics. Since NDM-1 has an open active site and unique electrostatic structure, it essentially provides a wider range of substrate specificity. Due to its flexible hydrolysis mechanism and more and more variants also aggravate the threat of drug-resistant bacteria infection, there is still no effective inhibitor in clinic, which is a serious threat to human health and public health safety. The electron-rich substituents of NDM-1 inhibitors coordinate with two positively charged zinc ions in the active center of the enzyme through ion-dipole interaction to produce NDM-1 inhibitory activity. In this review, the research progress of NDM-1 enzyme and its inhibitors in the past 5 years was reviewed. The crystal structure, active center structure, surrounding important amino acid residues, newly discovered inhibitors and their action mechanism are classified and summarized in detail, which can be used as a reference for the development of effective drugs against drug-resistant bacteria targeting NDM-1.


Subject(s)
Anti-Bacterial Agents/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/enzymology , Binding Sites , Catalytic Domain , Drug Resistance, Bacterial/drug effects , Molecular Docking Simulation , Picolinic Acids/chemistry , Picolinic Acids/metabolism , Picolinic Acids/pharmacology , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism
9.
J Biol Chem ; 297(2): 100980, 2021 08.
Article in English | MEDLINE | ID: mdl-34302811

ABSTRACT

ß-Lactamase inhibitory protein (BLIP) consists of a tandem repeat of αß domains conjugated by an interdomain loop and can effectively bind and inactivate class A ß-lactamases, which are responsible for resistance of bacteria to ß-lactam antibiotics. The varied ability of BLIP to bind different ß-lactamases and the structural determinants for significant enhancement of BLIP variants with a point mutation are poorly understood. Here, we investigated the conformational dynamics of BLIP upon binding to three clinically prevalent class A ß-lactamases (TEM1, SHV1, and PC1) with dissociation constants between subnanomolar and micromolar. Hydrogen deuterium exchange mass spectrometry revealed that the flexibility of the interdomain region was significantly suppressed upon strong binding to TEM1, but was not significantly changed upon weak binding to SHV1 or PC1. E73M and K74G mutations in the interdomain region improved binding affinity toward SHV1 and PC1, respectively, showing significantly increased flexibility of the interdomain region compared to the wild-type and favorable conformational changes upon binding. In contrast, more rigidity of the interfacial loop 135-145 was observed in these BLIP mutants in both free and bound states. Consistently, molecular dynamics simulations of BLIP exhibited drastic changes in the flexibility of the loop 135-145 in all complexes. Our results indicated for the first time that higher flexibility of the interdomain linker, as well as more rigidity of the interfacial loop 135-145, could be desirable determinants for enhancing inhibition of BLIP to class A ß-lactamases. Together, these findings provide unique insights into the design of enhanced inhibitors.


Subject(s)
Bacteria/enzymology , Bacterial Proteins/metabolism , Drug Resistance, Bacterial , Molecular Dynamics Simulation , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/metabolism , Amino Acid Sequence , Bacteria/chemistry , Bacteria/drug effects , Bacterial Proteins/chemistry , Protein Binding , Protein Domains , Protein Structural Elements , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry
10.
Chem Biol Drug Des ; 98(4): 481-492, 2021 10.
Article in English | MEDLINE | ID: mdl-34148302

ABSTRACT

Metallo-ß-lactamases (MBLs) are zinc-containing carbapenemases that inactivate a broad range of ß-lactam antibiotics. There is a lack of ß-lactamase inhibitors for restoring existing ß-lactam antibiotics arsenals against common bacterial infections. Fragment-based screening of a non-specific metal chelator library demonstrates 8-hydroxyquinoline as a broad-spectrum nanomolar inhibitor against VIM-2 and NDM-1. A hit-based substructure search provided an early structure-activity relationship of 8-hydroxyquinolines and identified 8-hydroxyquinoline-7-carboxylic acid as a low-cytotoxic ß-lactamase inhibitor that can restore ß-lactam activity against VIM-2-expressing E. coli. Molecular modeling further shed structural insight into its potential mode of binding within the dinuclear zinc active site. 8-Hydroxyquinoline-7-carboxylic acid is highly stable in human plasma and human liver microsomal study, making it an ideal lead candidate for further development.


Subject(s)
Hydroxyquinolines/chemistry , Small Molecule Libraries/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/metabolism , Bacterial Proteins/metabolism , Binding Sites , Escherichia coli/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Hydroxyquinolines/metabolism , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Protein Binding , Small Molecule Libraries/metabolism , Structure-Activity Relationship , Zinc/chemistry , beta-Lactamase Inhibitors/metabolism
11.
Bioorg Chem ; 113: 105024, 2021 08.
Article in English | MEDLINE | ID: mdl-34116340

ABSTRACT

In Gram-negative bacteria, the major mechanism of resistance to ß-lactam antibiotics is the production of one or several ß-lactamases (BLs), including the highly worrying carbapenemases. Whereas inhibitors of these enzymes were recently marketed, they only target serine-carbapenemases (e.g. KPC-type), and no clinically useful inhibitor is available yet to neutralize the class of metallo-ß-lactamases (MBLs). We are developing compounds based on the 1,2,4-triazole-3-thione scaffold, which binds to the di-zinc catalytic site of MBLs in an original fashion, and we previously reported its promising potential to yield broad-spectrum inhibitors. However, up to now only moderate antibiotic potentiation could be observed in microbiological assays and further exploration was needed to improve outer membrane penetration. Here, we synthesized and characterized a series of compounds possessing a diversely functionalized alkyl chain at the 4-position of the heterocycle. We found that the presence of a carboxylic group at the extremity of an alkyl chain yielded potent inhibitors of VIM-type enzymes with Ki values in the µM to sub-µM range, and that this alkyl chain had to be longer or equal to a propyl chain. This result confirmed the importance of a carboxylic function on the 4-substituent of 1,2,4-triazole-3-thione heterocycle. As observed in previous series, active compounds also preferentially contained phenyl, 2-hydroxy-5-methoxyphenyl, naphth-2-yl or m-biphenyl at position 5. However, none efficiently inhibited NDM-1 or IMP-1. Microbiological study on VIM-2-producing E. coli strains and on VIM-1/VIM-4-producing multidrug-resistant K. pneumoniae clinical isolates gave promising results, suggesting that the 1,2,4-triazole-3-thione scaffold worth continuing exploration to further improve penetration. Finally, docking experiments were performed to study the binding mode of alkanoic analogues in the active site of VIM-2.


Subject(s)
Thiones/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Binding Sites , Cell Survival/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/enzymology , HeLa Cells , Humans , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Microbial Sensitivity Tests , Molecular Docking Simulation , Protein Binding , Structure-Activity Relationship , Thiones/metabolism , Triazoles/chemistry , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/metabolism
12.
BMC Infect Dis ; 21(1): 444, 2021 May 17.
Article in English | MEDLINE | ID: mdl-34001022

ABSTRACT

BACKGROUND: To investigate the trends and correlation between antibacterial consumption and carbapenem resistance in Gram-negative bacteria from 2012 to 2019 in a tertiary-care teaching hospital in southern China. METHODS: This retrospective study included data from hospital-wide inpatients collected between January 2012 and December 2019. Data on antibacterial consumption were expressed as defined daily doses (DDDs)/1000 patient-days. Antibacterials were classified according to the Anatomical Therapeutic Chemical (ATC) classification system. The trends in antimicrobial usage and resistance were analyzed by linear regression, while Pearson correlation analysis was used for assessing correlations. RESULTS: An increasing trend in the annual consumption of tetracyclines, ß-lactam/ß-lactamase inhibitor (BL/BLI) combinations, and carbapenems was observed (P < 0.05). Carbapenem resistance in Acinetobacter baumannii (A. baumannii) significantly increased (P < 0.05) from 18% in 2012 to 60% in 2019. Moreover, significant positive correlations were found between resistance to carbapenems in A. baumannii (P < 0.05) and Escherichia coli (E. coli; P < 0.05) and consumption of carbapenems, while the resistance rate of A. baumannii to carbapenems was positively correlated with cephalosporin/ß-lactamase inhibitor (C/BLI) combinations (P < 0.01) and tetracyclines usage (P < 0.05). We also found that use of quinolones was positively correlated with the resistance rate of Burkholderia cepacia (B. cepacia) to carbapenems (P < 0.05), and increasing uses of carbapenems (P < 0.01) and penicillin/ß-Lactamase inhibitor (P/BLI) combinations (P < 0.01) were significantly correlated with reduced resistance of Enterobacter cloacae (E. cloacae) to carbapenems. CONCLUSION: These results revealed significant correlations between consumption of antibiotics and carbapenem resistance rates in Gram-negative bacteria. Implementing proper management strategies and reducing the unreasonable use of antibacterial drugs may be an effective measure to reduce the spread of carbapenem-resistant Gram-negative bacteria (CRGN), which should be confirmed by further studies.


Subject(s)
Drug Resistance, Bacterial , Gram-Negative Bacteria/metabolism , Gram-Negative Bacterial Infections/diagnosis , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/isolation & purification , Acinetobacter baumannii/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Burkholderia cepacia/drug effects , Burkholderia cepacia/isolation & purification , Burkholderia cepacia/metabolism , Carbapenems/pharmacology , Carbapenems/therapeutic use , Cephalosporins/metabolism , China , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Humans , Linear Models , Microbial Sensitivity Tests , Retrospective Studies , Tertiary Care Centers , Tetracyclines/metabolism , beta-Lactamase Inhibitors/metabolism
13.
Anal Bioanal Chem ; 413(13): 3561-3571, 2021 May.
Article in English | MEDLINE | ID: mdl-33851228

ABSTRACT

Clavulanic acid is a molecule with antimicrobial effect used in several livestock species treatment. Its inclusion in the treatment of infectious diseases of broilers requires determination of pharmacokinetic and pharmacodynamic parameters in order to determine the appropriate dosage for broilers and ensure safety of chicken products for human health. The present study describes the optimisation of analytical LC-MS/MS method for identification and quantification of clavulanic acid in broiler chicken plasma and meat. The limit of detection and the limit of quantification for the developed method were 3.09 µg·L-1 and 10.21 µg·L-1 for plasma and 2.57 µg·kg-1 and 8.47 µg·kg-1 for meat. The recoveries of the developed plasma and tissue extraction procedure were > 105.7% and > 95.6%, respectively. The achieved coefficient of variation of within-run precision ranged from 2.8 to 10.9% for plasma and from 6.5 to 8.5% for meat. The pharmacokinetic experiment was performed in 112 Ross broiler chickens assigned into time interval groups ranging from 10 min to 24 h in accredited animal facilities. Administered dose of clavulanic acid was 2.5 mg·kg-1 according to the manufacturer's recommendations. The pharmacokinetic parameters obtained from the experiment are as follows: Cmax = 1.82 ± 0.91 mg·L-1, Tmax = 0.25 h, T1/2 = 0.87 h, Kel = 0.80 ± 0.04 h-1, AUC0-∞ = 2.17 mg·h ·L-1.


Subject(s)
Clavulanic Acid/metabolism , Mass Spectrometry/methods , beta-Lactamase Inhibitors/metabolism , Animals , Chickens , Chromatography, High Pressure Liquid/methods , Clavulanic Acid/blood , Clavulanic Acid/pharmacokinetics , Limit of Detection , Reference Standards , Reproducibility of Results , beta-Lactamase Inhibitors/blood , beta-Lactamase Inhibitors/pharmacokinetics
14.
Eur J Med Chem ; 219: 113418, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33862516

ABSTRACT

The occurrence of resistances in Gram negative bacteria is steadily increasing to reach extremely worrying levels and one of the main causes of resistance is the massive spread of very efficient ß-lactamases which render most ß-lactam antibiotics useless. Herein, we report the development of a series of imino-analogues of ß-lactams (namely azetidinimines) as efficient non-covalent inhibitors of ß-lactamases. Despite the structural and mechanistic differences between serine-ß-lactamases KPC-2 and OXA-48 and metallo-ß-lactamase NDM-1, all three enzymes can be inhibited at a submicromolar level by compound 7dfm, which can also repotentiate imipenem against a resistant strain of Escherichia coli expressing NDM-1. We show that 7dfm can efficiently inhibit not only the three main clinically-relevant carbapenemases of Ambler classes A (KPC-2), B (NDM-1) and D (OXA-48) with Ki's below 0.3 µM, but also the cephalosporinase CMY-2 (class C, 86% inhibition at 10 µM). Our results pave the way for the development of a new structurally original family of non-covalent broad-spectrum inhibitors of ß-lactamases.


Subject(s)
Anti-Bacterial Agents/chemistry , Azetidines/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Azetidines/metabolism , Binding Sites , Catalytic Domain , Cell Line , Cell Proliferation/drug effects , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gram-Negative Bacteria/drug effects , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Docking Simulation , Structure-Activity Relationship , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/genetics , beta-Lactamases/metabolism
15.
Eur J Med Chem ; 215: 113257, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33618159

ABSTRACT

Penems have demonstrated potential as antibacterials and ß-lactamase inhibitors; however, their clinical use has been limited, especially in comparison with the structurally related carbapenems. Faropenem is an orally active antibiotic with a C-2 tetrahydrofuran (THF) ring, which is resistant to hydrolysis by some ß-lactamases. We report studies on the reactions of faropenem with carbapenem-hydrolysing ß-lactamases, focusing on the class A serine ß-lactamase KPC-2 and the metallo ß-lactamases (MBLs) VIM-2 (a subclass B1 MBL) and L1 (a B3 MBL). Kinetic studies show that faropenem is a substrate for all three ß-lactamases, though it is less efficiently hydrolysed by KPC-2. Crystallographic analyses on faropenem-derived complexes reveal opening of the ß-lactam ring with formation of an imine with KPC-2, VIM-2, and L1. In the cases of the KPC-2 and VIM-2 structures, the THF ring is opened to give an alkene, but with L1 the THF ring remains intact. Solution state studies, employing NMR, were performed on L1, KPC-2, VIM-2, VIM-1, NDM-1, OXA-23, OXA-10, and OXA-48. The solution results reveal, in all cases, formation of imine products in which the THF ring is opened; formation of a THF ring-closed imine product was only observed with VIM-1 and VIM-2. An enamine product with a closed THF ring was also observed in all cases, at varying levels. Combined with previous reports, the results exemplify the potential for different outcomes in the reactions of penems with MBLs and SBLs and imply further structure-activity relationship studies are worthwhile to optimise the interactions of penems with ß-lactamases. They also exemplify how crystal structures of ß-lactamase substrate/inhibitor complexes do not always reflect reaction outcomes in solution.


Subject(s)
Anti-Bacterial Agents/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry , beta-Lactams/chemistry , Anti-Bacterial Agents/metabolism , Catalytic Domain , Crystallography, X-Ray , Hydrolysis , Klebsiella pneumoniae/enzymology , Protein Binding , Pseudomonas aeruginosa/enzymology , Stenotrophomonas maltophilia/enzymology , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/metabolism , beta-Lactams/metabolism
16.
Bioorg Med Chem ; 29: 115902, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33302045

ABSTRACT

ß-lactam antibiotics have long been the mainstay for the treatment of bacterial infections. New Delhi metallo-ß-lactamase 1 (NDM-1) is able to hydrolyze nearly all ß-lactam antibiotics and even clinically used serine-ß-lactamase inhibitors. The wide and rapid spreading of NDM-1 gene among pathogenic bacteria has attracted extensive attention, therefore high potency NDM-1 inhibitors are urgently needed. Here we report a series of structure-guided design of D-captopril derivatives that can inhibit the activity of NDM-1 in vitro and at cellular levels. Structural comparison indicates the mechanisms of inhibition enhancement and provides insights for further inhibitor optimization.


Subject(s)
Anti-Bacterial Agents/chemistry , Captopril/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/metabolism , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Binding Sites , Captopril/metabolism , Captopril/pharmacology , Crystallography, X-Ray , Drug Discovery , Drug Resistance, Microbial/drug effects , Humans , Hydrolysis/drug effects , Models, Molecular , Protein Binding , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/pharmacology
17.
J Med Chem ; 63(24): 15802-15820, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33306385

ABSTRACT

The diazabicyclooctanes (DBOs) are a class of serine ß-lactamase (SBL) inhibitors that use a strained urea moiety as the warhead to react with the active serine residue in the active site of SBLs. The first in-class drug, avibactam, as well as several other recently approved DBOs (e.g., relebactam) or those in clinical development (e.g., nacubactam and zidebactam) potentiate activity of ß-lactam antibiotics, to various extents, against carbapenem-resistant Enterobacterales (CRE) carrying class A, C, and D SBLs; however, none of these are able to rescue the activity of ß-lactam antibiotics against carbapenem-resistant Acinetobacter baumannii (CRAB), a WHO "critical priority pathogen" producing class D OXA-type SBLs. Herein, we describe the chemical optimization and resulting structure-activity relationship, leading to the discovery of a novel DBO, ANT3310, which uniquely has a fluorine atom replacing the carboxamide and stands apart from the current DBOs in restoring carbapenem activity against OXA-CRAB as well as SBL-carrying CRE pathogens.


Subject(s)
Acinetobacter/drug effects , Anti-Bacterial Agents/pharmacology , Enterobacteriaceae/drug effects , Octanes/chemistry , beta-Lactamases/chemistry , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Binding Sites , Carbapenems/pharmacology , Drug Evaluation, Preclinical , Drug Resistance, Bacterial/drug effects , Half-Life , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Octanes/metabolism , Octanes/pharmacology , Stereoisomerism , Structure-Activity Relationship , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism
18.
Molecules ; 25(23)2020 Dec 05.
Article in English | MEDLINE | ID: mdl-33291474

ABSTRACT

Modern medicine relies upon antibiotics, but we have arrived to the point where our inability to come up with new effective molecules against resistant pathogens, together with the declining private investment, is resulting in the number of untreatable infections increasing worldwide at worrying pace. Among other pathogens, widely recognized institutions have indicated Gram-negative bacteria as particularly challenging, due to the presence of the outer membrane. The very first step in the action of every antibiotic or adjuvant is the permeation through this membrane, with small hydrophilic drugs usually crossing through protein channels. Thus, a detailed understanding of their properties at a molecular level is crucial. By making use of Molecular Dynamics simulations, we compared the two main porins of four members of the Enterobacteriaceae family, and, in this paper, we show their shared geometrical and electrostatic characteristics. Then, we used metadynamics simulations to reconstruct the free energy for permeation of selected diazobicyclooctans through OmpF. We demonstrate how porins features are coupled to those of the translocating species, modulating their passive permeation. In particular, we show that the minimal projection area of a molecule is a better descriptor than its molecular mass or the volume. Together with the magnitude and orientation of the electric dipole moment, these are the crucial parameters to gain an efficient compensation between the entropic and enthalpic contributions to the free energy barrier required for permeation. Our results confirm the possibility to predict the permeability of molecules through porins by using a few molecular parameters and bolster the general model according to which the free energy increase is mostly due to the decrease of conformational entropy, and this can be compensated by a favorable alignment of the electric dipole with respect to the channel intrinsic electric field.


Subject(s)
Cell Membrane Permeability/physiology , Gram-Negative Bacteria/metabolism , Porins/metabolism , beta-Lactamase Inhibitors/metabolism , Anti-Bacterial Agents/metabolism , Enterobacteriaceae/metabolism , Molecular Dynamics Simulation , Static Electricity
19.
Eur J Med Chem ; 208: 112829, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33002736

ABSTRACT

ß-Lactam antibiotics are one of the most relevant drug classes of antibacterial agents worldwide. The discovery and the market of first ß-lactam antibiotic (Penicillin G) is a symbolic landmark of modern chemotherapy. Since then, several other ß-lactam antibiotics have been introduced in the therapy, revolutionizing the treatment of bacterial infections. Their antibacterial efficacy has been kept in check by the emergence of bacterial resistance. Among the resistance mechanisms, the expression of ß-lactamase enzymes is one of the most studied and prevalent. The combined use of beta-lactamase inhibitors with broad spectrum activity ß-lactam antibiotics has been an effective strategy to circumvent the resistance issue. This review discusses, with a focus on structural aspects, the different classes of beta-lactam antibiotics (penicillins, cephalosporins, carbapenems, monobactams and penems) in light of their stability, sensitivity to ß-lactamases, mechanism of action and spectrum of antimicrobial activity. ß-Lactamase inhibitors (structurally correlated and non-correlated to the ß-lactam system) and their proposed inhibition mechanisms are also discussed.


Subject(s)
Anti-Bacterial Agents/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactams/chemistry , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Bacteria/drug effects , Bacterial Infections/drug therapy , Chemistry, Pharmaceutical , Humans , Hydrolysis , Models, Chemical , Molecular Structure , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamases/metabolism , beta-Lactams/metabolism , beta-Lactams/therapeutic use
20.
Eur J Med Chem ; 208: 112720, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32937203

ABSTRACT

Resistance to ß-lactam antibiotics in Gram-negatives producing metallo-ß-lactamases (MBLs) represents a major medical threat and there is an extremely urgent need to develop clinically useful inhibitors. We previously reported the original binding mode of 5-substituted-4-amino/H-1,2,4-triazole-3-thione compounds in the catalytic site of an MBL. Moreover, we showed that, although moderately potent, they represented a promising basis for the development of broad-spectrum MBL inhibitors. Here, we synthesized and characterized a large number of 4-amino-1,2,4-triazole-3-thione-derived Schiff bases. Compared to the previous series, the presence of an aryl moiety at position 4 afforded an average 10-fold increase in potency. Among 90 synthetic compounds, more than half inhibited at least one of the six tested MBLs (L1, VIM-4, VIM-2, NDM-1, IMP-1, CphA) with Ki values in the µM to sub-µM range. Several were broad-spectrum inhibitors, also inhibiting the most clinically relevant VIM-2 and NDM-1. Active compounds generally contained halogenated, bicyclic aryl or phenolic moieties at position 5, and one substituent among o-benzoic, 2,4-dihydroxyphenyl, p-benzyloxyphenyl or 3-(m-benzoyl)-phenyl at position 4. The crystallographic structure of VIM-2 in complex with an inhibitor showed the expected binding between the triazole-thione moiety and the dinuclear centre and also revealed a network of interactions involving Phe61, Tyr67, Trp87 and the conserved Asn233. Microbiological analysis suggested that the potentiation activity of the compounds was limited by poor outer membrane penetration or efflux. This was supported by the ability of one compound to restore the susceptibility of an NDM-1-producing E. coli clinical strain toward several ß-lactams in the presence only of a sub-inhibitory concentration of colistin, a permeabilizing agent. Finally, some compounds were tested against the structurally similar di-zinc human glyoxalase II and found weaker inhibitors of the latter enzyme, thus showing a promising selectivity towards MBLs.


Subject(s)
Schiff Bases/pharmacology , Thiones/pharmacology , Triazoles/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism , Crystallography, X-Ray , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Microbial Sensitivity Tests , Protein Binding , Pseudomonas aeruginosa/chemistry , Schiff Bases/chemical synthesis , Schiff Bases/metabolism , Thiones/chemical synthesis , Thiones/metabolism , Triazoles/chemical synthesis , Triazoles/metabolism , beta-Lactamase Inhibitors/chemical synthesis , beta-Lactamase Inhibitors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...