Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 453
Filter
1.
Science ; 383(6684): 721-726, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38359125

ABSTRACT

We report the design conception, chemical synthesis, and microbiological evaluation of the bridged macrobicyclic antibiotic cresomycin (CRM), which overcomes evolutionarily diverse forms of antimicrobial resistance that render modern antibiotics ineffective. CRM exhibits in vitro and in vivo efficacy against both Gram-positive and Gram-negative bacteria, including multidrug-resistant strains of Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa. We show that CRM is highly preorganized for ribosomal binding by determining its density functional theory-calculated, solution-state, solid-state, and (wild-type) ribosome-bound structures, which all align identically within the macrobicyclic subunits. Lastly, we report two additional x-ray crystal structures of CRM in complex with bacterial ribosomes separately modified by the ribosomal RNA methylases, chloramphenicol-florfenicol resistance (Cfr) and erythromycin-resistance ribosomal RNA methylase (Erm), revealing concessive adjustments by the target and antibiotic that permit CRM to maintain binding where other antibiotics fail.


Subject(s)
Anti-Bacterial Agents , Bridged-Ring Compounds , Drug Resistance, Multiple, Bacterial , Lincosamides , Oxepins , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Erythromycin/chemistry , Erythromycin/pharmacology , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Bridged-Ring Compounds/chemical synthesis , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/pharmacology , Oxepins/chemical synthesis , Oxepins/chemistry , Oxepins/pharmacology , Lincosamides/chemical synthesis , Lincosamides/chemistry , Lincosamides/pharmacology , Animals , Mice , Drug Design , Ribosomes/chemistry
2.
Bioresour Technol ; 384: 129288, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37315621

ABSTRACT

Removing erythromycin from the environment is a major challenge. In this study, a dual microbial consortium (Delftia acidovorans ERY-6A and Chryseobacterium indologenes ERY-6B) capable of degrading erythromycin was isolated, and the erythromycin biodegradation products were studied. Coconut shell activated carbon was modified and its adsorption characteristics and erythromycin removal efficiency of the immobilized cells were studied. It was indicated that alkali-modified and water-modified coconut shell activated carbon and the dual bacterial system had excellent erythromycin removal ability. The dual bacterial system follows a new biodegradation pathway to degrade erythromycin. The immobilized cells removed 95% of erythromycin at a concentration of 100 mg L-1 within 24 h through pore adsorption, surface complexation, hydrogen bonding, and biodegradation. This study provides a new erythromycin removal agent and for the first time describes the genomic information of erythromycin-degrading bacteria, providing new clues regarding bacterial cooperation and efficient erythromycin removal.


Subject(s)
Charcoal , Erythromycin , Erythromycin/chemistry , Bacteria/genetics , Biodegradation, Environmental , Adsorption
3.
Article in English | MEDLINE | ID: mdl-36078780

ABSTRACT

Erythromycin is one of the most commonly used macrolide antibiotics. However, its pollution of the ecosystem is a significant risk to human health worldwide. Currently, there are no effective and environmentally friendly methods to resolve this issue. Although erythromycin esterase B (EreB) specifically degrades erythromycin, its non-recyclability and fragility limit the large-scale application of this enzyme. In this work, palygorskite was selected as a carrier for enzyme immobilization. The enzyme was attached to palygorskite via a crosslinking reaction to construct an effective erythromycin-degradation material (i.e., EreB@modified palygorskite), which was characterized using FT-IR, SEM, XRD, and Brunauer-Emmett-Teller techniques. The results suggested the successful modification of the material and the loading of the enzyme. The immobilized enzyme had a higher stability over varying temperatures (25-65 °C) and pH values (6.5-10.0) than the free enzyme, and the maximum rate of reaction (Vmax) and the turnover number (kcat) of the enzyme increased to 0.01 mM min-1 and 169 min-1, respectively, according to the enzyme-kinetics measurements. The EreB@modified palygorskite maintained about 45% of its activity after 10 cycles, and degraded erythromycin in polluted water to 20 mg L-1 within 300 min. These results indicate that EreB could serve as an effective immobilizing carrier for erythromycin degradation at the industrial scale.


Subject(s)
Carboxylic Ester Hydrolases , Enzymes, Immobilized , Erythromycin , Carboxylic Ester Hydrolases/chemistry , Ecosystem , Erythromycin/chemistry , Humans , Hydrogen-Ion Concentration , Magnesium Compounds/chemistry , Silicon Compounds/chemistry , Spectroscopy, Fourier Transform Infrared
4.
Nucleic Acids Res ; 49(16): 9560-9573, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34417608

ABSTRACT

Macrolides have been effective clinical antibiotics for over 70 years. They inhibit protein biosynthesis in bacterial pathogens by narrowing the nascent protein exit tunnel in the ribosome. The macrolide class of natural products consist of a macrolactone ring linked to one or more sugar molecules. Most of the macrolides used currently are semi-synthetic erythromycin derivatives, composed of a 14- or 15-membered macrolactone ring. Rapidly emerging resistance in bacterial pathogens is among the most urgent global health challenges, which render many antibiotics ineffective, including next-generation macrolides. To address this threat and advance a longer-term plan for developing new antibiotics, we demonstrate how 16-membered macrolides overcome erythromycin resistance in clinically isolated Staphylococcus aureus strains. By determining the structures of complexes of the large ribosomal subunit of Deinococcus radiodurans (D50S) with these 16-membered selected macrolides, and performing anti-microbial studies, we identified resistance mechanisms they may overcome. This new information provides important insights toward the rational design of therapeutics that are effective against drug resistant human pathogens.


Subject(s)
Macrolides/chemistry , Micromonospora/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Erythromycin/chemistry , Humans , Macrolides/pharmacology , Microbial Sensitivity Tests , Protein Synthesis Inhibitors/pharmacology , Ribosomes/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity
5.
Nat Commun ; 12(1): 4466, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294725

ABSTRACT

Macrolides and ketolides comprise a family of clinically important antibiotics that inhibit protein synthesis by binding within the exit tunnel of the bacterial ribosome. While these antibiotics are known to interrupt translation at specific sequence motifs, with ketolides predominantly stalling at Arg/Lys-X-Arg/Lys motifs and macrolides displaying a broader specificity, a structural basis for their context-specific action has been lacking. Here, we present structures of ribosomes arrested during the synthesis of an Arg-Leu-Arg sequence by the macrolide erythromycin (ERY) and the ketolide telithromycin (TEL). Together with deep mutagenesis and molecular dynamics simulations, the structures reveal how ERY and TEL interplay with the Arg-Leu-Arg motif to induce translational arrest and illuminate the basis for the less stringent sequence-specific action of ERY over TEL. Because programmed stalling at the Arg/Lys-X-Arg/Lys motifs is used to activate expression of antibiotic resistance genes, our study also provides important insights for future development of improved macrolide antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ketolides/pharmacology , Macrolides/pharmacology , Protein Synthesis Inhibitors/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Bacillus subtilis/drug effects , Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Binding Sites/genetics , Cryoelectron Microscopy , Drug Resistance, Microbial/genetics , Erythromycin/chemistry , Erythromycin/pharmacology , Genes, Bacterial , Ketolides/chemistry , Ketolides/pharmacokinetics , Macrolides/chemistry , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , Molecular Dynamics Simulation , Mutagenesis, Insertional , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , Ribosomes/drug effects
6.
Nat Commun ; 12(1): 1732, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741980

ABSTRACT

Macrolides are a class of antibiotics widely used in both medicine and agriculture. Unsurprisingly, as a consequence of their exensive usage a plethora of resistance mechanisms have been encountered in pathogenic bacteria. One of these resistance mechanisms entails the enzymatic cleavage of the macrolides' macrolactone ring by erythromycin esterases (Eres). The most frequently identified Ere enzyme is EreA, which confers resistance to the majority of clinically used macrolides. Despite the role Eres play in macrolide resistance, research into this family enzymes has been sparse. Here, we report the first three-dimensional structures of an erythromycin esterase, EreC. EreC is an extremely close homologue of EreA, displaying more than 90% sequence identity. Two structures of this enzyme, in conjunction with in silico flexible docking studies and previously reported mutagenesis data allowed for the proposal of a detailed catalytic mechanism for the Ere family of enzymes, labeling them as metal-independent hydrolases. Also presented are substrate spectrum assays for different members of the Ere family. The results from these assays together with an examination of residue conservation for the macrolide binding site in Eres, suggests two distinct active site archetypes within the Ere enzyme family.


Subject(s)
Anti-Bacterial Agents/chemistry , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Esterases/chemistry , Esterases/genetics , Macrolides/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/enzymology , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Erythromycin/chemistry , Genes, Bacterial , Macrolides/pharmacology , Molecular Docking Simulation , Protein Conformation , X-Ray Diffraction
7.
Org Biomol Chem ; 19(7): 1483-1487, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33521803

ABSTRACT

There is a significant need for new antibacterial agents as pathogenic bacteria continue to threaten human health through the acquisition of resistance and tolerance towards existing antibiotics. Over the last several years, our group has been developing a novel series of halogenated phenazines that demonstrate potent antibacterial and biofilm eradication activities against critical Gram-positive pathogens, including: Staphylococcus aureus, Staphylococcus epidermidis and Enterococcus faecium. Here, we report the design, chemical synthesis and initial biological assessment of a halogenated phenazine-erythromycin conjugate prodrug 5 aimed at enhancing the translational potential for halogenated phenazines as a treatment of bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Erythromycin/pharmacology , Phenazines/pharmacology , Prodrugs/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Enterococcus faecium/drug effects , Erythromycin/chemistry , Microbial Sensitivity Tests , Molecular Structure , Phenazines/chemistry , Prodrugs/chemical synthesis , Prodrugs/chemistry , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects
8.
FEMS Microbiol Lett ; 368(1)2021 01 26.
Article in English | MEDLINE | ID: mdl-33338238

ABSTRACT

Erythromycin pollution is an important risk to the ecosystem and human health worldwide. Thus, it is urgent to develop effective approaches to decontaminate erythromycin. In this study, we successfully isolated a novel erythromycin-degrading fungus from an erythromycin-contaminated site. The erythromycin biodegradation characteristics were investigated in mineral salt medium with erythromycin as the sole carbon and energy source. The metabolites of erythromycin degraded by fungus were identified and used to derive the degradation pathway. Based on morphological and phylogenetic analyses, the isolated strain was named Curvularia sp. RJJ-5 (MN759651). Optimal degradation conditions for strain RJJ-5 were 30°C, and pH 6.0 with 100 mg L-1 erythromycin substrate. The strain could degrade 75.69% erythromycin under this condition. The following metabolites were detected: 3-depyranosyloxy erythromycin A, 7,12-dyhydroxy-6-deoxyerythronolide B, 2,4,6,8,10,12-hexamethyl-3,5,6,11,12,13-hexahydroxy-9-ketopentadecanoic acid and cladinose. It was deduced that the erythromycin A was degraded to 3-depyranosyloxy erythromycin A by glycoside hydrolase in the initial reaction. These results imply that Curvularia sp. RJJ-5 is a novel erythromycin-degrading fungus that can hydrolyze erythromycin using a glycoside hydrolase and has great potential for removing erythromycin from mycelial dreg and the contaminated environment.


Subject(s)
Anti-Bacterial Agents/metabolism , Curvularia/metabolism , Erythromycin/metabolism , Anti-Bacterial Agents/chemistry , Biodegradation, Environmental , Curvularia/classification , Curvularia/genetics , Curvularia/isolation & purification , Erythromycin/chemistry , Phylogeny , Soil Microbiology
9.
Int J Biol Macromol ; 168: 464-473, 2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33316335

ABSTRACT

Herein, we fabricated the antibacterial nanofibrous mats composed of cellulose acetate (CA) nanofibers loaded with erythromycin-chitosan nanoparticles (Ery-CS NPs) intended for infected wound dressing. The Ery-loaded CS NPs were prepared by ionic gelation process and then incorporated into the CA electrospun nanofibers (NFs). Regarding physiochemical properties, the NPs and obtained mats were characterized using dynamic light scattering (DLS), scanning electron microscopy (SEM), attenuated total reflection fourier transform infrared (ATR-FTIR), and contact angle measurement. The antimicrobial activity and cell viability of fibroblast cells were also evaluated. The results indicated that Ery was loaded into CS NPs with high encapsulation efficiency (95%). The CA NFs (17% w/v) incorporated with the Ery-CS NPs (12 wt%) displayed smooth homogenous morphology with 141.7 ± 91.7 nm average diameter. The relevant analyses confirmed that the NPs incorporated into NFs and provided high water holding capacity with high porosity. Finally, Ery-CS NPs/CA mats were able to inhibit the growth of both Gram-positive and Gram-negative bacteria as well as showed no cytotoxic effect on the human dermal fibroblast cells. Overall, our findings concluded that the proposed system could be potentially applied as the proper antibacterial mats for infected wound dressing applications.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cellulose/analogs & derivatives , Chitosan/chemistry , Erythromycin/pharmacology , Wound Infection/drug therapy , Animals , Anti-Bacterial Agents/chemistry , Bandages , Cell Line , Cell Survival , Cellulose/chemistry , Erythromycin/chemistry , Fibroblasts/cytology , Fibroblasts/drug effects , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Humans , Nanofibers , Porosity
10.
Molecules ; 25(17)2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32872323

ABSTRACT

Clarithromycin and congeners are important antibacterial members of the erythromycin A 14-membered macrocyclic lactone family. The macrolide scaffold consists of a multifunctional core that carries both chemically reactive and non-reactive substituents and sites. Two main approaches are used in the preparation of the macrolides. In semisynthesis, the naturally occurring macrocycle serves as a substrate for structural modifications of peripheral substituents. This review is focused on substituents in non-activated positions. In the total synthesis approach, the macrolide antibiotics are constructed by a convergent assembly of building blocks from presynthesized substrates or substrates prepared by biogenetic engineering. The assembled block structures are linear chains that are cyclized by macrolactonization or by metal-promoted cross-coupling reactions to afford the 14-membered macrolactone. Pendant glycoside residues are introduced by stereoselective glycosylation with a donor complex. When available, a short summary of antibacterial MIC data is included in the presentations of the structural modifications discussed.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Erythromycin/chemistry , Erythromycin/pharmacology , Macrolides/chemistry , Macrolides/pharmacology , Anti-Bacterial Agents/chemical synthesis , Chemical Phenomena , Chemistry Techniques, Synthetic , Drug Design , Erythromycin/chemical synthesis , Macrolides/chemical synthesis , Molecular Structure
11.
J Photochem Photobiol B ; 210: 111978, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32771913

ABSTRACT

Common bacterial pathogens have become resistant to traditional antibiotics, representing an indispensable public health crisis. Photodynamic therapy (PDT), especially when common visible light sources are used as photodynamic power, is a promising bactericidal method. Based on the special photodynamic properties triggered by commonly available light emitting diode (LED) lamps, a kind of graphene quantum dots (GQDs) based composite system (termed GQDs@hMSN(EM)) was prepared through loading both GQDs and erythromycin (EM) into the hollow mesoporous silica nanoparticle (hMSN), aiming to achieve joint antimicrobial effect. Bacterial density experiments confirmed that GQDs@hMSN(EM) had combined antimicrobial effects from photodynamic effect and drug release on Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). In animal models, the healing degree of wounds infected by bacteria also confirmed that GQDs@hMSN(EM) group had the best therapeutic effect, with the significantly reduced inflammatory factors in blood. Different from traditional GQDs synthesized by solvothermal method, the as-prepared GQDs@hMSN can produce singlet oxygen (1O2) under light exposure to destroy the structure of bacteria, thus achieving highly efficient antimicrobial effect. The GQDs@hMSN(EM) in this work possesses good antimicrobial activity, sufficient drug loading, and controllable drug release ability, which provides a new opportunity for GQDs-based nanoplatform to enhance antimicrobial effect and reduce their drug resistance.


Subject(s)
Anti-Bacterial Agents/chemistry , Graphite/chemistry , Quantum Dots/chemistry , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/metabolism , Bacterial Infections/pathology , Disease Models, Animal , Drug Carriers/chemistry , Drug Liberation , Erythromycin/chemistry , Erythromycin/metabolism , Erythromycin/pharmacology , Erythromycin/therapeutic use , Escherichia coli/drug effects , Female , Light , Male , Mice , Nanoparticles/chemistry , Porosity , Quantum Dots/toxicity , Silicon Dioxide/chemistry , Singlet Oxygen/metabolism , Staphylococcus aureus/drug effects , Wound Healing/drug effects
12.
Chem Commun (Camb) ; 56(74): 10926-10929, 2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32807999

ABSTRACT

We report the application of VCD spectroscopy for the characterization of clarithromycin and erythromycin. We show that the VCD spectra of these large macrolides are distinctly different and that spectra calculations reproduce the experimentally observed VCD signatures. In addition, computed VCD spectra of different epimers indicate that they should also be distinguishable from the correct structure of clarithromycin.


Subject(s)
Anti-Bacterial Agents/chemistry , Clarithromycin/chemistry , Erythromycin/chemistry , Circular Dichroism , Density Functional Theory , Molecular Conformation , Stereoisomerism , Vibration
13.
Biomolecules ; 10(9)2020 08 25.
Article in English | MEDLINE | ID: mdl-32854353

ABSTRACT

Streptomyces rochei 7434AN4 produces two structurally unrelated polyketide antibiotics lankacidin and lankamycin, and their biosynthesis is tightly controlled by butenolide-type signaling molecules SRB1 and SRB2. SRBs are synthesized by SRB synthase SrrX, and induce lankacidin and lankamycin production at 40 nM concentration. We here investigated the role of a P450 monooxygenase gene srrO (orf84), which is located adjacent to srrX (orf85), in SRB biosynthesis. An srrO mutant KA54 accumulated lankacidin and lankamycin at a normal level when compared with the parent strain. To elucidate the chemical structures of the signaling molecules accumulated in KA54 (termed as KA54-SRBs), this mutant was cultured (30 L) and the active components were purified. Two active components (KA54-SRB1 and KA54-SRB2) were detected in ESI-MS and chiral HPLC analysis. The molecular formulae for KA54-SRB1 and KA54-SRB2 are C15H26O4 and C16H28O4, whose values are one oxygen smaller and two hydrogen larger when compared with those for SRB1 and SRB2, respectively. Based on extensive NMR analysis, the signaling molecules in KA54 were determined to be 6'-deoxo-SRB1 and 6'-deoxo-SRB2. Gel shift analysis indicated that a ligand affinity of 6'-deoxo-SRB1 to the specific receptor SrrA was 100-fold less than that of SRB1. We performed bioconversion of the synthetic 6'-deoxo-SRB1 in the Streptomyces lividans recombinant carrying SrrO-expression plasmid. Substrate 6'-deoxo-SRB1 was converted through 6'-deoxo-6'-hydroxy-SRB1 to SRB1 in a time-dependent manner. Thus, these results clearly indicated that SrrO catalyzes the C-6' oxidation at a final step in SRB biosynthesis.


Subject(s)
4-Butyrolactone/analogs & derivatives , Cytochrome P-450 Enzyme System/metabolism , Streptomyces/metabolism , 4-Butyrolactone/biosynthesis , 4-Butyrolactone/chemistry , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biosynthetic Pathways , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/genetics , Erythromycin/analogs & derivatives , Erythromycin/biosynthesis , Erythromycin/chemistry , Genes, Bacterial , Macrolides/chemistry , Macrolides/metabolism , Magnetic Resonance Spectroscopy , Molecular Structure , Mutation , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Streptomyces/genetics
14.
J Am Chem Soc ; 142(43): 18369-18377, 2020 10 28.
Article in English | MEDLINE | ID: mdl-32709196

ABSTRACT

Many microorganisms possess the capacity for producing multiple antibiotic secondary metabolites. In a few notable cases, combinations of secondary metabolites produced by the same organism are used in important combination therapies for treatment of drug-resistant bacterial infections. However, examples of conjoined roles of bioactive metabolites produced by the same organism remain uncommon. During our genetic functional analysis of oxidase-encoding genes in the everninomicin producer Micromonospora carbonacea var. aurantiaca, we discovered previously uncharacterized antibiotics everninomicin N and O, comprised of an everninomicin fragment conjugated to the macrolide rosamicin via a rare nitrone moiety. These metabolites were determined to be hydrolysis products of everninomicin P, a nitrone-linked conjugate likely the result of nonenzymatic condensation of the rosamicin aldehyde and the octasaccharide everninomicin F, possessing a hydroxylamino sugar moiety. Rosamicin binds the erythromycin macrolide binding site approximately 60 Å from the orthosomycin binding site of everninomicins. However, while individual ribosomal binding sites for each functional half of everninomicin P are too distant for bidentate binding, ligand displacement studies demonstrated that everninomicin P competes with rosamicin for ribosomal binding. Chemical protection studies and structural analysis of everninomicin P revealed that everninomicin P occupies both the macrolide- and orthosomycin-binding sites on the 70S ribosome. Moreover, resistance mutations within each binding site were overcome by the inhibition of the opposite functional antibiotic moiety binding site. These data together demonstrate a strategy for coupling orthogonal antibiotic pharmacophores, a surprising tolerance for substantial covalent modification of each antibiotic, and a potential beneficial strategy to combat antibiotic resistance.


Subject(s)
Nitrogen Oxides/chemistry , Ribosomes/metabolism , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Binding Sites , Cryoelectron Microscopy , Erythromycin/chemistry , Erythromycin/metabolism , Leucomycins/chemistry , Leucomycins/metabolism , Micromonospora/genetics , Multigene Family , Nitrogen Oxides/metabolism
15.
Physiol Rep ; 8(5): e14385, 2020 03.
Article in English | MEDLINE | ID: mdl-32147975

ABSTRACT

The macrolide antibiotic erythromycin has been associated with QT interval prolongation and inhibition of the hERG-encoded channels responsible for the rapid delayed rectifier K+ current I(Kr ). It has been suggested that low concentrations of erythromycin may have a protective effect against hERG block and associated drug-induced arrhythmia by reducing the affinity of the pore-binding site for high potency hERG inhibitors. This study aimed to explore further the notion of a potentially protective effect of erythromycin. Whole-cell patch-clamp experiments were performed in which hERG-expressing mammalian (Human Embryonic Kidney; HEK) cells were preincubated with low to moderate concentrations of erythromycin (3 or 30 µM) prior to whole-cell patch clamp recordings of hERG current (IhERG ) at 37°C. In contrast to a previous report, exposure to low concentrations of erythromycin did not reduce pharmacological sensitivity of hERG to the antipsychotic thioridazine and antihistamine terfenadine. The IC50 value for IhERG tail inhibition by terfenadine was decreased by ~32-fold in the presence of 3 µM erythromycin (p < .05 vs. no preincubation). Sensitivity to thioridazine remained unchanged (p > .05 vs. no preincubation). The effects of low concentrations of erythromycin were investigated for a series of pore blocking drugs, and the results obtained were consistent with additive and/or synergistic effects. Experiments with the externally acting blocker BeKm-1 on WT hERG and a pore mutant (F656V) were used to explore the location of the binding site for erythromycin. Our data are inconsistent with the use of erythromycin for the management of drug-induced QT prolongation.


Subject(s)
Antipsychotic Agents/administration & dosage , ERG1 Potassium Channel/antagonists & inhibitors , Erythromycin/administration & dosage , Histamine H1 Antagonists, Non-Sedating/administration & dosage , Terfenadine/administration & dosage , Thioridazine/administration & dosage , Binding Sites/drug effects , ERG1 Potassium Channel/physiology , Erythromycin/chemistry , HEK293 Cells , Humans , Inhibitory Concentration 50 , Macrolides/administration & dosage , Macrolides/chemistry , Patch-Clamp Techniques
16.
Rapid Commun Mass Spectrom ; 34(11): e8772, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32112476

ABSTRACT

RATIONALE: A simple and sensitive method was developed for the separation and characterization of four unknown impurities in azithromycin and erythromycin imino ether using two-dimensional liquid chromatography coupled to high-resolution quadrupole time-of-flight mass spectrometry (2D LC/QTOFMS) with positive and negative electrospray ionization. METHODS: The chromatographic separation in the first dimension was performed with a Waters Xbridge RP18 column in gradient mode using binary mobile phase: (A) phosphate buffer (pH 8.2)-acetonitrile (47:53, v/v) and (B) water-acetonitrile (90:10, v/v). In the second dimension, the chromatographic separation was performed using a Shimadzu Shim-pack GISS C18 column with volatile mobile phases: (A) ammonium formate solution (10 mM) and (B) methanol. RESULTS: The molecular formulae and structures of the four impurities were deduced based on the LC/MS/MS data, and further confirmed using 1 H NMR, 13 C NMR, 1 H-1 H COSY, HSQC and HMBC NMR spectra after semi-preparative isolation of impurities. In addition, the mechanism for the formation of the impurities was also proposed. CONCLUSIONS: The contradiction between the non-volatile salt mobile phase and mass spectrometry was solved by means of a multiple heart-cutting 2D LC approach and on-line desalination technology. Four impurities were separated and characterized. These results could further improve the method of official monographs in pharmacopoeias and guides to improve the process of reducing impurity content.


Subject(s)
Azithromycin/chemistry , Chromatography, Liquid/methods , Drug Contamination , Erythromycin/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Magnetic Resonance Spectroscopy/methods , Models, Molecular
17.
RNA ; 26(6): 715-723, 2020 06.
Article in English | MEDLINE | ID: mdl-32144191

ABSTRACT

Macrolides are one of the most successful and widely used classes of antibacterials, which kill or stop the growth of pathogenic bacteria by binding near the active site of the ribosome and interfering with protein synthesis. Dirithromycin is a derivative of the prototype macrolide erythromycin with additional hydrophobic side chain. In our recent study, we have discovered that the side chain of dirithromycin forms lone pair-π stacking interaction with the aromatic imidazole ring of the His69 residue in ribosomal protein uL4 of the Thermus thermophilus 70S ribosome. In the current work, we found that neither the presence of the side chain, nor the additional contact with the ribosome, improve the binding affinity of dirithromycin to the ribosome. Nevertheless, we found that dirithromycin is a more potent inhibitor of in vitro protein synthesis in comparison with its parent compound, erythromycin. Using high-resolution cryo-electron microscopy, we determined the structure of the dirithromycin bound to the translating Escherichia coli 70S ribosome, which suggests that the better inhibitory properties of the drug could be rationalized by the side chain of dirithromycin pointing into the lumen of the nascent peptide exit tunnel, where it can interfere with the normal passage of the growing polypeptide chain.


Subject(s)
Anti-Bacterial Agents/chemistry , Erythromycin/analogs & derivatives , Protein Synthesis Inhibitors/chemistry , Ribosomes/chemistry , Anti-Bacterial Agents/pharmacology , Cryoelectron Microscopy , Erythromycin/chemistry , Erythromycin/pharmacology , Escherichia coli/genetics , Models, Molecular , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Ribosomal, 23S/chemistry
18.
Biochim Biophys Acta Gen Subj ; 1864(6): 129546, 2020 06.
Article in English | MEDLINE | ID: mdl-32032658

ABSTRACT

Multidrug efflux is a major contributor to antibiotic resistance in Gram-negative bacterial pathogens. Inhibition of multidrug efflux pumps is a promising approach for reviving the efficacy of existing antibiotics. Previously, inhibitors targeting both the efflux transporter AcrB and the membrane fusion protein AcrA in the Escherichia coli AcrAB-TolC efflux pump were identified. Here we use existing physicochemical property guidelines to generate a filtered library of compounds for computational docking. We then experimentally test the top candidate coumpounds using in vitro binding assays and in vivo potentiation assays in bacterial strains with controllable permeability barriers. We thus identify a new class of inhibitors of E. coli AcrAB-TolC. Six molecules with a shared scaffold were found to potentiate the antimicrobial activity of erythromycin and novobiocin in hyperporinated E. coli cells. Importantly, these six molecules were also active in wild-type strains of both Acinetobacter baumannii and Klebsiella pneumoniae, potentiating the activity of erythromycin and novobiocin up to 8-fold.


Subject(s)
Anti-Infective Agents/pharmacology , Carrier Proteins/chemistry , Escherichia coli Proteins/chemistry , Gram-Negative Bacterial Infections/drug therapy , Lipoproteins/chemistry , Membrane Transport Proteins/chemistry , Multidrug Resistance-Associated Proteins/chemistry , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/pathogenicity , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/chemistry , Carrier Proteins/antagonists & inhibitors , Computational Biology/methods , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Drug Synergism , Erythromycin/chemistry , Erythromycin/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Humans , Klebsiella pneumoniae , Lipoproteins/antagonists & inhibitors , Molecular Docking Simulation , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Novobiocin/chemistry , Novobiocin/pharmacology
19.
Curr Pharm Biotechnol ; 21(10): 948-954, 2020.
Article in English | MEDLINE | ID: mdl-31994459

ABSTRACT

BACKGROUND: Staphylococcus aureus (S. aureus) is the most common infectious agent in the community and hospitals. Infections with S. aureus are now becoming difficult to be treated by using conventional antibiotics due to its emerging methicillin-resistant S. aureus (MRSA) strain. OBJECTIVE: In the present study, MRSA was isolated from clinical samples and evaluated for resistance against different antibiotics, TiO2 nanoparticles, and their combinations. METHODS: Clinical samples were collected from Ayub Medical Complex (AMC), Abbottabad, Pakistan, and identified by different biochemical tests and polymerase chain reactions (PCR). Kirby-Bauer disk diffusion method was performed to evaluate antimicrobial susceptibility. Minimum Inhibitory Concentration (MIC) of ampicillin, ciprofloxacin, erythromycin, and vancomycin was found out by agar dilution method while the broth dilution method was used for the MIC of TiO2 nanoparticles and their combinations with erythromycin. RESULTS: All 13/100 (13%) MRSA were successfully identified. All isolates were susceptible to quinupristin/ dalfopristin, teicoplanin, and vancomycin, while the highest resistance was seen with erythromycin, penicillin, and tetracycline. MIC showed high resistance against ampicillin (0.25-512 mg/L) and erythromycin (0.25-1024 mg/L). CONCLUSION: The MIC value of 2 mM TiO2 nanoparticles was found to be the most effective concentration after 12 h of incubation, while the combination of erythromycin with 3 mM TiO2 nanoparticles was found to be more potent which significantly lowered down the MIC of erythromycin to 2-16 mg/L.


Subject(s)
Anti-Bacterial Agents/pharmacology , Erythromycin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Nanoparticles/chemistry , Titanium/pharmacology , Anti-Bacterial Agents/chemistry , Drug Synergism , Erythromycin/chemistry , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Microbial Sensitivity Tests , Staphylococcal Infections/microbiology , Titanium/chemistry
20.
J Biomed Mater Res B Appl Biomater ; 108(2): 475-483, 2020 02.
Article in English | MEDLINE | ID: mdl-31070858

ABSTRACT

Calcium polyphosphate (CPP) hydrogel is used to load erythromycin (EM) and vancomycin (VCM) by means of two loading methods: they are either added directly to the formed CPP hydrogel (Gel Mixture method) or mixed with CPP powders, followed by the formation of CPP-antibiotic hydrogel (Powder Mixture method). The release of loaded antibiotics from CPP hydrogel is measured up to 48 hr. Compared to Powder Mixture method, Gel Mixture method significantly reduced the burst release of embedded antibiotics. A significant change in CPP hydrogel Raman characteristic peaks is observed only in Gel Mixture method, indicating a close interaction between embedded antibiotics with CPP hydrogel matrix. In contrast, a similarity between characteristic peaks of CPP hydrogel and Powder Mixture method shows that antibiotic incorporation does not interfere with CPP gel formation, resulting in no ionic interaction between antibiotic and polyphosphate chains. Rheometer analysis further confirms that the hydrophobic nature of EM impacts the viscoelastic properties of CPP hydrogel, whereas the hydrophilic VCM exhibits a higher loading efficiency. The potential application of CPP hydrogel as a ceramic matrix for sustained drug release warrants further investigation.


Subject(s)
Anti-Bacterial Agents/chemistry , Calcium Phosphates/chemistry , Drug Carriers/chemistry , Erythromycin/chemistry , Hydrogels/chemistry , Polymers/chemistry , Vancomycin/chemistry , Anti-Bacterial Agents/pharmacology , Drug Compounding , Drug Liberation , Drug Therapy, Combination , Erythromycin/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Rheology , Vancomycin/pharmacology , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL
...