Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
RNA ; 25(5): 600-606, 2019 05.
Article in English | MEDLINE | ID: mdl-30733327

ABSTRACT

The 70S ribosome is a major target for antibacterial drugs. Two of the classical antibiotics, chloramphenicol (CHL) and erythromycin (ERY), competitively bind to adjacent but separate sites on the bacterial ribosome: the catalytic peptidyl transferase center (PTC) and the nascent polypeptide exit tunnel (NPET), respectively. The previously reported competitive binding of CHL and ERY might be due either to a direct collision of the two drugs on the ribosome or due to a drug-induced allosteric effect. Because of the resolution limitations, the available structures of these antibiotics in complex with bacterial ribosomes do not allow us to discriminate between these two possible mechanisms. In this work, we have obtained two crystal structures of CHL and ERY in complex with the Thermus thermophilus 70S ribosome at a higher resolution (2.65 and 2.89 Å, respectively) allowing unambiguous placement of the drugs in the electron density maps. Our structures provide evidence of the direct collision of CHL and ERY on the ribosome, which rationalizes the observed competition between the two drugs.


Subject(s)
Anti-Bacterial Agents/chemistry , Chloramphenicol/chemistry , Erythromycin/chemistry , Ribosome Subunits/drug effects , Thermus thermophilus/drug effects , Anti-Bacterial Agents/pharmacology , Binding Sites , Binding, Competitive , Chloramphenicol/pharmacology , Crystallography, X-Ray , Erythromycin/pharmacology , Escherichia coli/chemistry , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Models, Molecular , Peptidyl Transferases/antagonists & inhibitors , Peptidyl Transferases/chemistry , Peptidyl Transferases/genetics , Peptidyl Transferases/metabolism , Protein Binding , Protein Biosynthesis , Protein Conformation , Ribosome Subunits/genetics , Ribosome Subunits/metabolism , Ribosome Subunits/ultrastructure , Thermus thermophilus/chemistry , Thermus thermophilus/genetics , Thermus thermophilus/metabolism
2.
Amino Acids ; 49(5): 995-1004, 2017 05.
Article in English | MEDLINE | ID: mdl-28283906

ABSTRACT

Chloramphenicol peptides were recently established as useful tools for probing nascent polypeptide chain interaction with the ribosome, either biochemically, or structurally. Here, we present a new 10mer chloramphenicol peptide, which exerts a dual inhibition effect on the ribosome function affecting two distinct areas of the ribosome, namely the peptidyl transferase center and the polypeptide exit tunnel. According to our data, the chloramphenicol peptide bound on the chloramphenicol binding site inhibits the formation of both acetyl-phenylalanine-puromycin and acetyl-lysine-puromycin, showing, however, a decreased peptidyl transferase inhibition compared to chloramphenicol-mediated inhibition per se. Additionally, we found that the same compound is a strong inhibitor of green fluorescent protein synthesis in a coupled in vitro transcription-translation assay as well as a potent inhibitor of lysine polymerization in a poly(A)-programmed ribosome, showing that an additional inhibitory effect may exist. Since chemical protection data supported the interaction of the antibiotic with bases A2058 and A2059 near the entrance of the tunnel, we concluded that the extra inhibition effect on the synthesis of longer peptides is coming from interactions of the peptide moiety of the drug with residues comprising the ribosomal tunnel, and by filling up the tunnel and blocking nascent chain progression through the restricted tunnel. Therefore, the dual interaction of the chloramphenicol peptide with the ribosome increases its inhibitory effect and opens a new window for improving the antimicrobial potency of classical antibiotics or designing new ones.


Subject(s)
Chloramphenicol/pharmacology , Fluorenes/chemistry , Peptides/pharmacology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Ribosomes/drug effects , Amino Acid Sequence , Binding Sites , Chloramphenicol/analogs & derivatives , Chloramphenicol/chemical synthesis , Escherichia coli K12/chemistry , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Green Fluorescent Proteins/antagonists & inhibitors , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Internal Ribosome Entry Sites/drug effects , Models, Molecular , Peptides/chemical synthesis , Peptidyl Transferases/antagonists & inhibitors , Peptidyl Transferases/genetics , Peptidyl Transferases/metabolism , Poly A/genetics , Poly A/metabolism , Protein Binding , Protein Synthesis Inhibitors/chemical synthesis , Puromycin/pharmacology , Ribosomes/genetics , Ribosomes/metabolism
3.
Nucleic Acids Res ; 42(13): 8621-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24939899

ABSTRACT

Chloramphenicol (CAM) is a broad-spectrum antibiotic, limited to occasional only use in developed countries because of its potential toxicity. To explore the influence of polyamines on the uptake and activity of CAM into cells, a series of polyamine-CAM conjugates were synthesized. Both polyamine architecture and the position of CAM-scaffold substitution were crucial in augmenting the antibacterial and anticancer potency of the synthesized conjugates. Compounds 4 and 5, prepared by replacement of dichloro-acetyl group of CAM with succinic acid attached to N4 and N1 positions of N(8),N(8)-dibenzylspermidine, respectively, exhibited higher activity than CAM in inhibiting the puromycin reaction in a bacterial cell-free system. Kinetic and footprinting analysis revealed that whereas the CAM-scaffold preserved its role in competing with the binding of aminoacyl-tRNA 3'-terminus to ribosomal A-site, the polyamine-tail could interfere with the rotatory motion of aminoacyl-tRNA 3'-terminus toward the P-site. Compared to CAM, compounds 4 and 5 exhibited comparable or improved antibacterial activity, particularly against CAM-resistant strains. Compound 4 also possessed enhanced toxicity against human cancer cells, and lower toxicity against healthy human cells. Thus, the designed conjugates proved to be suitable tools in investigating the ribosomal catalytic center plasticity and some of them exhibited greater efficacy than CAM itself.


Subject(s)
Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Chloramphenicol/pharmacology , Polyamines/chemistry , Protein Synthesis Inhibitors/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Binding Sites , Cell Line, Tumor , Chloramphenicol/chemistry , Chloramphenicol/toxicity , Escherichia coli/drug effects , Humans , Protein Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/toxicity , Ribosomes/drug effects
4.
J Enzyme Inhib Med Chem ; 31(2): 276-82, 2016.
Article in English | MEDLINE | ID: mdl-25807301

ABSTRACT

Ketolides belong to the latest generation of macrolides and are not only effective against macrolide susceptible bacterial strains but also against some macrolide resistant strains. Here we present data providing insights into the mechanism of action of K-1602, a novel alkyl-aryl-bearing fluoroketolide. According to our data, the K-1602 interacts with the ribosome as a one-step slow binding inhibitor, displaying an association rate constant equal to 0.28 × 10(4) M(-1) s(-1) and a dissociation rate constant equal to 0.0025 min(-1). Both constants contribute to produce an overall inhibition constant Ki equal to 1.49 × 10(-8) M, which correlates very well with the superior activity of this compound when compared with many other ketolides or fluoroketolides.


Subject(s)
Ketolides/chemistry , Ketolides/pharmacology , Ribosomes/drug effects , Binding, Competitive , Drug Resistance, Bacterial/drug effects , Escherichia coli/drug effects , Kinetics , Puromycin/chemistry , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Tylosin/chemistry , Tylosin/pharmacology
5.
Bioorg Med Chem ; 23(13): 3163-74, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26001343

ABSTRACT

A series of chloramphenicol (CAM) amides with polyamines (PAs), suitable for structure-activity relationship studies, were synthesized either by direct attachment of the PA chain on the 2-aminopropane-1,3-diol backbone of CAM, previously oxidized selectively at its primary hydroxyl group, or from chloramphenicol base (CLB) through acylation with succinic or phthalic anhydride and finally coupling with a PA. Conjugates 4 and 5, in which the CLB moiety was attached on N4 and N1 positions, respectively, of the N(8),N(8)-dibenzylated spermidine through the succinate linker, were the most potent antibacterial agents. Both conjugates were internalized into Escherichia coli cells by using the spermidine-preferential uptake system and caused decrease in protein and polyamine content of the cells. Noteworthy, conjugate 4 displayed comparable activity to CAM in MRSA or wild-type strains of Staphylococcus aureus and Escherichia coli, but superior activity in E. coli strains possessing ribosomal mutations or expressing the CAM acetyltransferase (cat) gene. Lead compounds, and in particular conjugate 4, have been therefore discovered during the course of the present work with clinical potential.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Chloramphenicol/chemistry , Escherichia coli/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Spermidine/chemistry , Acetyltransferases/genetics , Acetyltransferases/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enzyme Assays , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Gene Expression , Kinetics , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Mutation , Phthalic Anhydrides/chemistry , Succinic Anhydrides/chemistry
6.
Antimicrob Agents Chemother ; 58(1): 472-80, 2014.
Article in English | MEDLINE | ID: mdl-24189263

ABSTRACT

Ketolides, the third generation of expanded-spectrum macrolides, have in the last years become a successful weapon in the endless war against macrolide-resistant pathogens. Ketolides are semisynthetic derivatives of the naturally produced macrolide erythromycin, displaying not only improved activity against some erythromycin-resistant strains but also increased bactericidal activity as well as inhibitory effects at lower drug concentrations. In this study, we present a series of novel ketolides carrying alkyl-aryl side chains at the C-6 position of the lactone ring and, additionally, one or two fluorine atoms attached either directly to the lactone ring at the C-2 position or indirectly via the C-13 position. According to our genetic and biochemical studies, these novel ketolides occupy the known macrolide binding site at the entrance of the ribosomal tunnel and exhibit lower MIC values against wild-type or mutant strains than erythromycin. In most cases, the ketolides display activities comparable to or better than the clinically used ketolide telithromycin. Chemical protection experiments using Escherichia coli ribosomes bearing U2609C or U754A mutations in 23S rRNA suggest that the alkyl-aryl side chain establishes an interaction with the U2609-A752 base pair, analogous to that observed with telithromycin but unlike the interactions formed by cethromycin. These findings reemphasize the versatility of the alkyl-aryl side chains with respect to species specificity, which will be important for future design of improved antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Erythromycin/pharmacology , Escherichia coli/drug effects , Escherichia coli/metabolism , Ketolides/pharmacology , Microbial Sensitivity Tests , Protein Biosynthesis/drug effects , RNA, Ribosomal, 23S/genetics , Ribosomes/drug effects , Ribosomes/metabolism
7.
Nucleic Acids Res ; 40(11): 5078-87, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362747

ABSTRACT

Applying kinetics and footprinting analysis, we show that telithromycin, a ketolide antibiotic, binds to Escherichia coli ribosomes in a two-step process. During the first, rapidly equilibrated step, telithromycin binds to a low-affinity site (K(T) = 500 nM), in which the lactone ring is positioned at the upper portion of the peptide exit tunnel, while the alkyl-aryl side chain of the drug inserts a groove formed by nucleotides A789 and U790 of 23S rRNA. During the second step, telithromycin shifts slowly to a high-affinity site (K(T)* = 8.33 nM), in which the lactone ring remains essentially at the same position, while the side chain interacts with the base pair U2609:A752 and the extended loop of protein L22. Consistently, mutations perturbing either the base pair U2609:A752 or the L22-loop hinder shifting of telithromycin to the final position, without affecting the initial step of binding. In contrast, mutation Lys63Glu in protein L4 placed on the opposite side of the tunnel, exerts only a minor effect on telithromycin binding. Polyamines disfavor both sequential steps of binding. Our data correlate well with recent crystallographic data and rationalize the changes in the accessibility of ribosomes to telithromycin in response to ribosomal mutations and ionic changes.


Subject(s)
Anti-Bacterial Agents/chemistry , Escherichia coli/genetics , Ketolides/chemistry , Protein Synthesis Inhibitors/chemistry , Ribosomes/chemistry , Anti-Bacterial Agents/metabolism , Binding Sites , Ketolides/metabolism , Ligands , Models, Molecular , Protein Synthesis Inhibitors/metabolism , RNA, Ribosomal, 23S/chemistry , RNA, Ribosomal, 23S/metabolism , Ribosome Subunits, Large, Bacterial/chemistry , Ribosomes/metabolism
8.
Toxics ; 8(4)2020 Oct 18.
Article in English | MEDLINE | ID: mdl-33081042

ABSTRACT

The impact of metals bioaccumulation in marine organisms is a subject of intense investigation. This study was designed to determine the association between oxidative stress induced by seawater enriched with trace metals and protein synthesis using as a model the mussels Mytilus galloprovincialis. Mussels were exposed to 40 µg/L Cu, 30 µg/L Hg, or 100 µg/L Cd for 5 and 15 days, and the pollution effect was evaluated by measuring established oxidative biomarkers. The results showed damage on the protein synthesis machine integrity and specifically on translation factors and ribosomal proteins expression and modifications. The exposure of mussels to all metals caused oxidative damage that was milder in the cases of Cu and Hg and more pronounced for Cd. However, after prolonged exposure of mussels to Cd (15 days), the effects receded. These changes that perturb protein biosynthesis can serve as a great tool for elucidating the mechanisms of toxicity and could be integrated in biomonitoring programs.

9.
Antimicrob Agents Chemother ; 53(4): 1411-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19164155

ABSTRACT

Ketolides represent the latest generation of macrolide antibiotics, displaying improved activities against some erythromycin-resistant strains, while maintaining their activity against erythromycin-susceptible ones. In this study, we present a new ketolide, K-1325, that carries an alkyl-aryl side chain at C-13 of the lactone ring. According to our genetic and biochemical studies, K-1325 binds within the nascent polypeptide exit tunnel, at a site previously described as the primary attachment site of all macrolide antibiotics. Compared with telithromycin, K-1325 displays enhanced antimicrobial activity against wild-type Escherichia coli strains, as well as against strains bearing the U2609C mutation in 23S rRNA. Chemical protection experiments showed that the alkyl-aryl side chain of K-1325 interacts specifically with helix 35 of 23S rRNA, a fact leading to an increased affinity of U2609C mutant ribosomes for the drug and rationalizing the enhanced effectiveness of this new ketolide.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Ketolides/pharmacology , Erythromycin/metabolism , RNA, Ribosomal, 23S/chemistry , Structure-Activity Relationship
10.
Nucleic Acids Res ; 35(15): 5108-19, 2007.
Article in English | MEDLINE | ID: mdl-17652323

ABSTRACT

5S rRNA is an integral component of the large ribosomal subunit in virtually all living organisms. Polyamine binding to 5S rRNA was investigated by cross-linking of N1-azidobenzamidino (ABA)-spermine to naked 5S rRNA or 50S ribosomal subunits and whole ribosomes from Escherichia coli cells. ABA-spermine cross-linking sites were kinetically measured and their positions in 5S rRNA were localized by primer extension analysis. Helices III and V, and loops A, C, D and E in naked 5S rRNA were found to be preferred polyamine binding sites. When 50S ribosomal subunits or poly(U)-programmed 70S ribosomes bearing tRNA(Phe) at the E-site and AcPhe-tRNA at the P-site were targeted, the susceptibility of 5S rRNA to ABA-spermine was greatly reduced. Regardless of 5S rRNA assembly status, binding of spermine induced significant changes in the 5S rRNA conformation; loop A adopted an apparent 'loosening' of its structure, while loops C, D, E and helices III and V achieved a more compact folding. Poly(U)-programmed 70S ribosomes possessing 5S rRNA cross-linked with spermine were more efficient than control ribosomes in tRNA binding, peptidyl transferase activity and translocation. Our results support the notion that 5S rRNA serves as a signal transducer between regions of 23S rRNA responsible for principal ribosomal functions.


Subject(s)
RNA, Ribosomal, 5S/chemistry , Ribosomes/metabolism , Azides/chemistry , Base Sequence , Escherichia coli/genetics , Kinetics , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Peptidyl Transferases/metabolism , Photoaffinity Labels , Poly U/metabolism , RNA, Bacterial/chemistry , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/chemistry , Ribosomes/enzymology , Spermine/analogs & derivatives , Spermine/chemistry
11.
Antibiotics (Basel) ; 8(1)2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30699905

ABSTRACT

Over the last years, we have been focused on chloramphenicol conjugates that combine in their structure chloramphenicol base with natural polyamines, spermine, spermidine and putrescine, and their modifications. Conjugate 3, with spermidine (SPD) as a natural polyamine linked to chloramphenicol base, showed the best antibacterial and anticancer properties. Using 3 as a prototype, we here explored the influence of the antibacterial and anticancer activity of additional benzyl groups on N1 amino moiety together with modifications of the alkyl length of the aminobutyl fragment of SPD. Our data demonstrate that the novel modifications did not further improve the antibacterial activity of the prototype. However, one of the novel conjugates (4) showed anticancer activity without affecting bacterial growth, thus emerging as a promising anticancer agent, with no adverse effects on bacterial microflora when taken orally.

12.
Nucleic Acids Res ; 33(9): 2792-805, 2005.
Article in English | MEDLINE | ID: mdl-15897324

ABSTRACT

Polyamine binding to 23S rRNA was investigated, using a photoaffinity labeling approach. This was based on the covalent binding of a photoreactive analog of spermine, N1-azidobenzamidino (ABA)-spermine, to Escherichia coli ribosomes or naked 23S rRNA under mild irradiation conditions. The cross-linking sites of ABA-spermine in 23S rRNA were determined by RNase H digestion and primer-extension analysis. Domains I, II, IV and V in naked 23S rRNA were identified as discrete regions of preferred cross-linking. When 50S ribosomal subunits were targeted, the interaction of the photoprobe with the above 23S rRNA domains was elevated, except for helix H38 in domain II whose susceptibility to cross-linking was greatly reduced. In addition, cross-linking sites were identified in domains III and VI. Association of 30S with 50S subunits, poly(U), tRNA(Phe) and AcPhe-tRNA to form a post-translocation complex further altered the cross-linking, in particular to helices H11-H13, H21, H63, H80, H84, H90 and H97. Poly(U)-programmed 70S ribosomes, reconstituted from photolabeled 50S subunits and untreated 30S subunits, bound AcPhe-tRNA in a similar fashion to native ribosomes. However, they exhibited higher reactivity toward puromycin and enhanced tRNA-translocation efficiency. These results suggest an essential role for polyamines in the structural and functional integrity of the large ribosomal subunit.


Subject(s)
Azides/chemistry , Photoaffinity Labels , RNA, Ribosomal, 23S/chemistry , Ribosomes/chemistry , Spermine/analogs & derivatives , Spermine/chemistry , Azides/metabolism , Base Sequence , Binding Sites , Escherichia coli/genetics , Molecular Sequence Data , Peptidyl Transferases/metabolism , Poly U/metabolism , Protein Biosynthesis , RNA, Ribosomal, 23S/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/metabolism , Spermine/metabolism
13.
Br J Pharmacol ; 174(18): 2967-2983, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28664582

ABSTRACT

Macrolides represent a large family of protein synthesis inhibitors of great clinical interest due to their applicability to human medicine. Macrolides are composed of a macrocyclic lactone of different ring sizes, to which one or more deoxy-sugar or amino sugar residues are attached. Macrolides act as antibiotics by binding to bacterial 50S ribosomal subunit and interfering with protein synthesis. The high affinity of macrolides for bacterial ribosomes, together with the highly conserved structure of ribosomes across virtually all of the bacterial species, is consistent with their broad-spectrum activity. Since the discovery of the progenitor macrolide, erythromycin, in 1950, many derivatives have been synthesised, leading to compounds with better bioavailability and acid stability and improved pharmacokinetics. These efforts led to the second generation of macrolides, including well-known members such as azithromycin and clarithromycin. Subsequently, in order to address increasing antibiotic resistance, a third generation of macrolides displaying improved activity against many macrolide resistant strains was developed. However, these improvements were accompanied with serious side effects, leading to disappointment and causing many researchers to stop working on macrolide derivatives, assuming that this procedure had reached the end. In contrast, a recent published breakthrough introduced a new chemical platform for synthesis and discovery of a wide range of diverse macrolide antibiotics. This chemical synthesis revolution, in combination with reduction in the side effects, namely, 'Ketek effects', has led to a macrolide renaissance, increasing the hope for novel and safe therapeutic agents to combat serious human infectious diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Communicable Diseases/drug therapy , Communicable Diseases/microbiology , Macrolides/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Humans , Macrolides/chemical synthesis , Macrolides/chemistry
14.
Nucleic Acids Res ; 31(17): 5074-83, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12930958

ABSTRACT

Chloramphenicol is thought to interfere competitively with the binding of the aminoacyl-tRNA 3'-terminus to ribosomal A-site. However, noncompetitive or mixed-noncompetitive inhibition, often observed to be dependent on chloramphenicol concentration and ionic conditions, leaves some doubt about the precise mode of action. Here, we examine further the inhibition effect of chloramphenicol, using a model system derived from Escherichia coli in which a peptide bond is formed between puromycin and AcPhe-tRNA bound at the P-site of poly(U)-programmed ribosomes, under ionic conditions (6 mM Mg2+, 100 mM NH4+, 100 microM spermine) more closely resembling the physiological status. Kinetics reveal that chloramphenicol (I) reacts rapidly with AcPhe-tRNA.poly(U).70S ribosomal complex (C) to form the encounter complex CI which is then isomerized slowly to a more tight complex, C*I. A similar inhibition pattern is observed, if complex C modified by a photoreactive analogue of spermine, reacts in buffer free of spermine. Spermine, either reversibly interacting with or covalently attached to ribosomes, enhances the peptidyltransferase activity and increases the chloramphenicol potency, without affecting the isomerization step. As indicated by photoaffinity labeling, the peptidyltransferase center at which chloramphenicol binds, is one of the preferred cross-linking sites for polyamines. This fact may explain the effect of spermine on chloramphenicol binding to ribosomes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Peptidyl Transferases/metabolism , Polyamines/pharmacology , Spermine/analogs & derivatives , Anti-Bacterial Agents/metabolism , Azides/metabolism , Azides/pharmacology , Base Sequence , Binding Sites , Binding, Competitive/drug effects , Chloramphenicol/metabolism , Chloramphenicol/pharmacology , Dose-Response Relationship, Drug , Escherichia coli/genetics , Escherichia coli/metabolism , Kinetics , Magnesium/pharmacology , Molecular Sequence Data , Nucleic Acid Conformation , Peptide Biosynthesis/drug effects , Peptidyl Transferases/antagonists & inhibitors , Polyamines/metabolism , Puromycin/metabolism , RNA, Ribosomal, 23S/chemistry , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 23S/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosomes/metabolism , Spermine/metabolism , Spermine/pharmacology
15.
Antibiotics (Basel) ; 5(2)2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27271676

ABSTRACT

Chloramphenicol (CAM) is the D-threo isomer of a small molecule, consisting of a p-nitrobenzene ring connected to a dichloroacetyl tail through a 2-amino-1,3-propanediol moiety. CAM displays a broad-spectrum bacteriostatic activity by specifically inhibiting the bacterial protein synthesis. In certain but important cases, it also exhibits bactericidal activity, namely against the three most common causes of meningitis, Haemophilus influenzae, Streptococcus pneumoniae and Neisseria meningitidis. Resistance to CAM has been frequently reported and ascribed to a variety of mechanisms. However, the most important concerns that limit its clinical utility relate to side effects such as neurotoxicity and hematologic disorders. In this review, we present previous and current efforts to synthesize CAM derivatives with improved pharmacological properties. In addition, we highlight potentially broader roles of these derivatives in investigating the plasticity of the ribosomal catalytic center, the main target of CAM.

16.
PLoS One ; 10(8): e0134526, 2015.
Article in English | MEDLINE | ID: mdl-26267355

ABSTRACT

As fight against antibiotic resistance must be strengthened, improving old drugs that have fallen in reduced clinical use because of toxic side effects and/or frequently reported resistance, like chloramphenicol (CAM), is of special interest. Chloramphenicol (CAM), a prototypical wide-spectrum antibiotic has been shown to obstruct protein synthesis via binding to the bacterial ribosome. In this study we sought to identify features intensifying the bacteriostatic action of CAM. Accordingly, we synthesized a series of CAM-dimers with various linker lengths and functionalities and compared their efficiency in inhibiting peptide-bond formation in an Escherichia coli cell-free system. Several CAM-dimers exhibited higher activity, when compared to CAM. The most potent of them, compound 5, containing two CAM bases conjugated via a dicarboxyl aromatic linker of six successive carbon-bonds, was found to simultaneously bind both the ribosomal catalytic center and the exit-tunnel, thus revealing a second, kinetically cryptic binding site for CAM. Compared to CAM, compound 5 exhibited comparable antibacterial activity against MRSA or wild-type strains of Staphylococcus aureus, Enterococcus faecium and E. coli, but intriguingly superior activity against some CAM-resistant E. coli and Pseudomonas aeruginosa strains. Furthermore, it was almost twice as active in inhibiting the growth of T-leukemic cells, without affecting the viability of normal human lymphocytes. The observed effects were rationalized by footprinting tests, crosslinking analysis, and MD-simulations.


Subject(s)
Anti-Infective Agents/chemistry , Chloramphenicol/chemistry , Dimerization , Drug Resistance, Microbial/genetics , Anti-Infective Agents/therapeutic use , Cell-Free System/drug effects , Chloramphenicol/therapeutic use , Escherichia coli/drug effects , Escherichia coli/pathogenicity , Humans , Lymphocytes/drug effects , Molecular Dynamics Simulation , Protein Biosynthesis/genetics , Pseudomonas aeruginosa/drug effects , Ribosomes/genetics , Staphylococcus aureus/drug effects
17.
Biochimie ; 95(9): 1765-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23770443

ABSTRACT

The ribosomal exit tunnel had recently become the centre of many functional and structural studies. Accumulated evidence indicates that the tunnel is not simply a passive conduit for the nascent chain, but a rather functionally important compartment where nascent peptide sequences can interact with the ribosome to signal translation to slow down or even stop. To explore further this interaction, we have synthesized short peptides attached to the amino group of a chloramphenicol (CAM) base, such that when bound to the ribosome these compounds mimic a nascent peptidyl-tRNA chain bound to the A-site of the peptidyltransferase center (PTC). Here we show that these CAM-peptides interact with the PTC of the ribosome while their effectiveness can be modulated by the sequence of the peptide, suggesting a direct interaction of the peptide with the ribosomal tunnel. Indeed, chemical footprinting in the presence of CAM-P2, one of the tested CAM-peptides, reveals protection of 23S rRNA nucleotides located deep within the tunnel, indicating a potential interaction with specific components of the ribosomal tunnel. Collectively, our findings suggest that the CAM-based peptide derivatives will be useful tools for targeting polypeptide chain mimics to the ribosomal tunnel, allowing their conformation and interaction with the ribosomal tunnel to be explored using further biochemical and structural methods.


Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Chloramphenicol/metabolism , Chloramphenicol/pharmacology , Oligopeptides/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Amino Acid Sequence , Binding Sites , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Models, Molecular , Oligopeptides/chemistry , Peptidyl Transferases/chemistry , Peptidyl Transferases/metabolism , Protein Binding , Protein Biosynthesis/drug effects , Protein Conformation , Ribosomes/genetics
18.
Int J Antimicrob Agents ; 35(3): 235-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20045632

ABSTRACT

Ketolides are effective not only against macrolide-sensitive bacteria but also against some macrolide-resistant strains. Here we present data regarding a new ketolide with an alkyl-aryl side chain at C-13 of its lactone ring. It behaves as a strong inhibitor of protein synthesis in a model coupled transcription/translation system, although it does not affect the accuracy of translation. In addition, detailed kinetic analysis shows that it slowly forms a very tight, slowly reversible complex with prokaryotic ribosomes, a property that could be correlated with its superior activity compared with erythromycin against Escherichia coli both in vivo and in vitro.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Ketolides/chemistry , Ketolides/pharmacology , Ribosomes/drug effects , Humans , Kinetics , Molecular Structure , Protein Biosynthesis/drug effects
19.
J Mol Biol ; 385(4): 1179-92, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19071138

ABSTRACT

Azithromycin is a semisynthetic derivative of erythromycin that inhibits bacterial protein synthesis by binding within the peptide exit tunnel of the 50S ribosomal subunit. Nevertheless, there is still debate over what localization is primarily responsible for azithromycin binding and as to how many molecules of the drug actually bind per ribosome. In the present study, kinetic methods and footprinting analysis are coupled together to provide time-resolved details of the azithromycin binding process. It is shown that azithromycin binds to Escherichia coli ribosomes in a two-step process: The first-step involves recognition of azithromycin by the ribosomal machinery and places the drug in a low-affinity site located in the upper part of the exit tunnel. The second step corresponds to the slow formation of a final complex that is both much tighter and more potent in hindering the progression of the nascent peptide through the exit tunnel. Substitution of uracil by cytosine at nucleoside 2609 of 23S rRNA, a base implicated in the high-affinity site, facilitates the shift of azithromycin to this site. In contrast, mutation U754A hardly affects the binding process. Binding of azithromycin to both sites is hindered by high concentrations of Mg(2+) ions. Unlike Mg(2+) ions, polyamines do not significantly affect drug binding to the low-affinity site but attenuate the formation of the final complex. The low- and high-affinity sites of azithromycin binding are mutually exclusive, which means that one molecule of the drug binds per E. coli ribosome at a time. In contrast, kinetic and binding data indicate that in Deinococcus radiodurans, two molecules of azithromycin bind cooperatively to the ribosome. This finding confirms previous crystallographic results and supports the notion that species-specific structural differences may primarily account for the apparent discrepancies between the antibiotic binding modes obtained for different organisms.


Subject(s)
Azithromycin/metabolism , Escherichia coli/metabolism , Ribosomes/metabolism , Anti-Bacterial Agents/pharmacology , Azithromycin/chemistry , Binding Sites , Deinococcus/drug effects , Deinococcus/metabolism , Escherichia coli/drug effects , Kinetics , Ligands , Nucleosides/metabolism , Puromycin/chemical synthesis , RNA, Ribosomal/metabolism , Ribosome Subunits/chemistry , Ribosomes/drug effects , Time Factors , Tylosin/chemistry , Tylosin/pharmacology
20.
J Biol Chem ; 283(8): 4756-65, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18079110

ABSTRACT

Erythromycin and tylosin are 14- and 16-membered lactone ring macrolides, respectively. The current work shows by means of kinetic and chemical footprinting analysis that both antibiotics bind to Escherichia coli ribosomes in a two-step process. The first step established rapidly, involves a low-affinity binding site placed at the entrance of the exit tunnel in the large ribosomal subunit, where macrolides bind primarily through their hydrophobic portions. Subsequently, slow conformational changes mediated by the antibiotic hydrophilic portion push the drugs deeper into the tunnel, in a high-affinity site. Compared with erythromycin, tylosin shifts to the high-affinity site more rapidly, due to the interaction of the mycinose sugar of the drug with the loop of H35 in domain II of 23 S rRNA. Consistently, mutations of nucleosides U2609 and U754 implicated in the high-affinity site reduce the shift of tylosin to this site and destabilize, respectively, the final drug-ribosome complex. The weak interaction between tylosin and the ribosome is Mg2+ independent, unlike the tight binding. In contrast, both interactions between erythromycin and the ribosome are reduced by increasing concentrations of Mg2+ ions. Polyamines attenuate erythromycin affinity for the ribosome at both sequential steps of binding. In contrast, polyamines facilitate the initial binding of tylosin, but exert a detrimental, more pronounced, effect on the drug accommodation at its final position. Our results emphasize the role of the particular interactions that side chains of tylosin and erythromycin establish with 23 S rRNA, which govern the exact binding process of each drug and its response to the ionic environment.


Subject(s)
Anti-Bacterial Agents/chemistry , Erythromycin/chemistry , Escherichia coli/chemistry , RNA, Bacterial/chemistry , RNA, Ribosomal, 23S/chemistry , Ribosomes/chemistry , Tylosin/chemistry , Escherichia coli/genetics , Hydrophobic and Hydrophilic Interactions , Kinetics , Magnesium/chemistry , Mutation , Polyamines/chemistry , RNA, Bacterial/genetics , RNA, Ribosomal, 23S/genetics , Ribosomes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL