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










Database
Language
Publication year range
1.
J Antibiot (Tokyo) ; 74(9): 574-579, 2021 09.
Article in English | MEDLINE | ID: mdl-34234284

ABSTRACT

The superbug infection caused by metallo-ß-lactamases (MßLs) carrying drug-resistant bacteria, specifically, New Delhi metallo-ß-lactamase (NDM-1) has become an emerging threat. In an effort to develop novel inhibitors of NDM-1, thirteen thiosemicarbazones (1a-1m) were synthesized and assayed. The obtained molecules specifically inhibited NDM-1, with an IC50 in the range of 0.88-20.2 µM, and 1a and 1f were found to be the potent inhibitors (IC50 = 1.79 and 0.88 µM) using cefazolin as substrate. ITC and kinetic assays indicated that 1a irreversibly and non-competitively inhibited NDM-1 in vitro. Importantly, MIC assays revealed that these molecules by themselves can sterilize NDM-producing clinical isolates EC01 and EC08, exhibited 78-312-fold stronger activities than the cefazolin. MIC assays suggest that 1a (16 µg ml-1) has synergistic antimicrobial effect with ampicillin, cefazolin and meropenem on E. coli producing NDM-1, resulting in MICs of 4-32-, 4-32-, and 4-8-fold decrease, respectively. These studies indicate that the thiosemicarbazide is a valuable scaffold for the development of inhibitors of NDM-1 and NDM-1 carrying drug-resistant bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Thiosemicarbazones/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cefazolin/pharmacology , Drug Resistance, Bacterial , Drug Synergism , Escherichia coli/drug effects , Humans , Inhibitory Concentration 50 , Meropenem/pharmacology , Microbial Sensitivity Tests , Structure-Activity Relationship , Thiosemicarbazones/chemical synthesis , Thiosemicarbazones/chemistry , beta-Lactamase Inhibitors/chemical synthesis , beta-Lactamase Inhibitors/chemistry
2.
Bioorg Chem ; 114: 105138, 2021 09.
Article in English | MEDLINE | ID: mdl-34229201

ABSTRACT

The expression of ß-lactamases, especially metallo-ß-lactamases (MßLs) in bacteria is one of the main causes of drug resistance. In this work, an effective N-acylhydrazone scaffold as MßL inhibitor was constructed and characterized. The biological activity assays indicated that the synthesized N-acylhydrazones 1-11 preferentially inhibited MßL NDM-1, and 1 was found to be the most effective inhibitor with an IC50 of 1.2 µM. Analysis of IC50 data revealed a structure-activity relationship, which is that the pyridine and hydroxylbenzene substituents at 2-position improved inhibition of the compounds on NDM-1. ITC and enzyme kinetics assays suggested that it reversibly and competitively inhibited NDM-1 (Ki = 0.29 ± 0.05 µM). The synthesized N-acylhydrazones showed synergistic antibacterial activities with meropenem, reduced 4-16-fold MIC of meropenem on NDM-1- producing E. coli BL21 (DE3), while 1 restored 4-fold activity of meropenem on K. pneumonia expressing NDM-1 (NDM-K. pneumoniae). The mice experiments suggested that 1 combined meropenem to fight against NDM-K. pneumoniae infection in the spleen and liver. Cytotoxicity assays showed that 1 and 2 have low cytotoxicity. This study offered a new framework for the development of NDM-1 inhibitors.


Subject(s)
Hydrazones/pharmacology , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases/metabolism , Animals , Chlorocebus aethiops , Dose-Response Relationship, Drug , Hydrazones/chemical synthesis , Hydrazones/chemistry , Mice , Mice, Inbred Strains , Molecular Structure , Structure-Activity Relationship , Vero Cells , beta-Lactamase Inhibitors/chemical synthesis , beta-Lactamase Inhibitors/chemistry
3.
ACS Infect Dis ; 6(5): 975-985, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32119777

ABSTRACT

The emergence and prevalence of carbapenem-resistant bacterial infection have seriously threatened the clinical use of almost all ß-lactam antibacterials. The development of effective metallo-ß-lactamase (MßL) inhibitors to restore the existing antibiotics efficacy is an ideal alternative. Although several types of serine-ß-lactamase inhibitors have been successfully developed and used in clinical settings, MßL inhibitors are not clinically available to date. Herein, we identified that cisplatin and Pd(II) complexes are potent broad-spectrum inhibitors of the B1 and B2 subclasses of MßLs and effectively revived Meropenem efficacy against MßL-expressing bacteria in vitro. Enzyme kinetics, thermodynamics, inductively coupled plasma atomic emission spectrometry (ICP-AES), matrix-assisted laser desorption/ionization-time of flight-mass spectrometry (MALDI-TOF-MS), and site-directed mutation assays revealed that these metal complexes irreversibly inhibited NDM-1 through a novel inhibition mode involving binding to Cys208 and displacing one Zn(II) ion of the enzyme with one Pt(II) containing two NH3's or one Pd(II) ion. Importantly, the combination therapy of Meropenem and metal complexes significantly suppressed the development of higher-level resistance in bacteria producing NDM-1, also effectively reduced the bacterial burden in liver and spleen of mice infected by carbapenem-resistant Enterobacteriaceae producing NDM-1. These findings will offer potential lead compounds for the further development of clinically useful inhibitors targeting MßLs.


Subject(s)
Carbapenem-Resistant Enterobacteriaceae/drug effects , Cisplatin/pharmacology , Enterobacteriaceae Infections/drug therapy , Palladium/pharmacology , beta-Lactamase Inhibitors , Animals , Mice , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases
4.
Eur J Pharm Sci ; 142: 105161, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31751778

ABSTRACT

The bacteria, harboring metallo-ß-lactamases (MßLs), become resistant on most ß-lactam antibiotics, specifically New Delhi metallo-ß-lactamase-1 (NDM-1), which hydrolyzes almost all ß-lactam antibiotics leading to bacterial multiple-drug resistance. It is highly desirable to develop effective NDM-1 inhibitors in reviving the efficacy of existing antibiotics. Here, we report a potent covalently reversible scaffold, 3-Bromopyruvate (3BP) to target the NDM-1 in vitro and in vivo. Enzymatic kinetic studies revealed that 3BP is capable of inhibiting the B1 and B2 MßLs and exhibited the best inhibition on NDM-1 with an IC50 of 2.57 µM, also, it was found to be a dose- and time-dependent inhibitor. The study of inhibition mechanism suggested that 3BP reversibly inactivate NDM-1, and may form a dynamic reversible covalent bond with cysteine at active site of the enzyme. Besides, 3BP effectively restored the activity of five ß-lactam antibiotics on three clinical strains expressing NDM-1, resulting in 2-8-fold reduction in MIC. Moreover, the toxicity evaluation of 3BP against L929 mouse fibroblastic cells indicated that 3BP had low cytotoxicity, implying it may be used as lead molecule for future drug candidate.


Subject(s)
Anti-Bacterial Agents/pharmacology , Pyruvates/pharmacology , beta-Lactamases/metabolism , Animals , Bacteria/drug effects , Catalytic Domain/drug effects , Cell Line , Cysteine/metabolism , Drug Resistance, Multiple, Bacterial/drug effects , Fibroblasts/microbiology , Kinetics , Mice
5.
Bioconjug Chem ; 31(1): 51-63, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31830418

ABSTRACT

The traditional antibiotics have specific intracellular targets and disinfect in chemical ways, and the drug-resistance induced by the antibiotics has grown into an emerging threat. It is urgent to call for novel strategies and antibacterial materials to control this situation. Herein, we report a class of silver-decorated nanocomposite AgNPs@PCL-b-AMPs as potent nanoantibiotic, constructed by ring-opening polymerization of the monomers ε-caprolactone, Z-Lys-N-carboxyanhydrides (NCAs), and Phe-NCAs, then decorated with AgNPs, and characterized by SEM, TEM, and DLS. The biological assays revealed that the nanocomposite possessed strong antibacterial efficacy against both Gram-positive and Gram-negative bacteria including clinical isolated bacteria MRSA, VRE, P. aeruginosa, and K. pneumonia, exhibiting a MIC value range in 2-8 µg/mL. Importantly, the S. aureus and P. aeruginosa treated with the nanocomposite did not show drug-resistance even after 21 passages. Also, in vivo anti-infective assays showed that the nanocomposite was able to effectively kill bacteria in the infected viscera of mice. The study of the sterilization mechanism showed that the nanocomposite exhibited a multimodal antimicrobial mechanism, including irreversibly damaging the membrane structure, making the leakage of intracellular ions and subsequently inducing generation of the reactive oxygen species (ROS), ultimately sterilizing the bacteria. The nanocomposite exhibits effective broad-spectrum antibacterial properties and shows low toxicity to the mammalian cells/animal. Overall, the AgNPs@PCL-b-AMPs gained in this work show great potential as a highly promising antibacterial material for biomedical applications including drug-resistant bacterial infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Metal Nanoparticles , Polyesters/pharmacology , Silver/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Polyesters/chemistry , Polyesters/therapeutic use , Silver/chemistry , Silver/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...