Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 399
Filter
1.
Am J Ther ; 29(6): e625-e631, 2022.
Article in English | MEDLINE | ID: mdl-36301538

ABSTRACT

BACKGROUND: Advances in drug therapy for pulmonary tuberculosis have had an extraordinary impact on the incidence of tuberculosis in the United States in the past century, which has decreased from 113/100,000 persons in 1920 to 2.2/100,000 in 2020. Modern treatments have contributed to a remarkable decrease in hospitalizations and mortality and have had a significant impact on the duration and severity of illness, quality of life, and work potential of affected persons. STUDY QUESTION: What are the milestones of the changes in the expert approach to the pharmacological management of pulmonary tuberculosis in the past century? STUDY DESIGN: To determine the changes in the experts' approach to the management of pulmonary tuberculosis, as presented in a widely used textbook in the United States. DATA SOURCES: The chapters describing the management of pulmonary tuberculosis in the 26 editions of Cecil Textbook of Medicine published from 1927 through 2020. RESULTS: In the preantibiotic era (1927-1943), the Cecil authors emphasized rest, good food, and fresh air as the treatment pillars for pulmonary tuberculosis. The modern era (1947-1971) recorded the discovery of all the drugs that are still used for the initial treatment, in the following order: streptomycin, para-aminosalicylic acid, isoniazid, pyrazinamide, ethambutol, cycloserine, kanamycin, ethionamide, capreomycin, and rifampin. In the postmodern era (1975-2020), therapeutic advances continued with trials of many drug combinations aimed at ameliorating the duration of treatment, drug resistance adverse effects, and poor the recent addition of fluoroquinolones, bedaquiline, and clofazimine. CONCLUSIONS: The pharmacological management of tuberculosis has remained archaic until the middle of the 20th century. Fundamental progress occurred in a very short period (1947-1971) and was because of the recognition of the antituberculous effect of many antibiotics and chemotherapy agents. The challenges created by mycobacterial infections resistant to multiple drugs remain and have prompted the addition of new drugs in the past decade.


Subject(s)
Tuberculosis, Pulmonary , Tuberculosis , Viomycin , Humans , Expert Testimony , Quality of Life , Aminosalicylic Acids , Drug Resistance , Drug Resistance, Microbial , Tuberculosis, Pulmonary/drug therapy , Streptomycin , Pyrazinamide , Isoniazid , Antitubercular Agents/therapeutic use
2.
RNA ; 27(9): 981-990, 2021 09.
Article in English | MEDLINE | ID: mdl-34117118

ABSTRACT

Many antibiotics that bind to the ribosome inhibit translation by blocking the movement of tRNAs and mRNA or interfering with ribosome dynamics, which impairs the formation of essential translocation intermediates. Here we show how translocation inhibitors viomycin (Vio), neomycin (Neo), paromomycin (Par), kanamycin (Kan), spectinomycin (Spc), hygromycin B (HygB), and streptomycin (Str, an antibiotic that does not inhibit tRNA movement), affect principal motions of the small ribosomal subunits (SSU) during EF-G-promoted translocation. Using ensemble kinetics, we studied the SSU body domain rotation and SSU head domain swiveling in real time. We show that although antibiotics binding to the ribosome can favor a particular ribosome conformation in the absence of EF-G, their kinetic effect on the EF-G-induced transition to the rotated/swiveled state of the SSU is moderate. The antibiotics mostly inhibit backward movements of the SSU body and/or the head domains. Vio, Spc, and high concentrations of Neo completely inhibit the backward movements of the SSU body and head domain. Kan, Par, HygB, and low concentrations of Neo slow down both movements, but their sequence and coordination are retained. Finally, Str has very little effect on the backward rotation of the SSU body domain, but retards the SSU head movement. The data underscore the importance of ribosome dynamics for tRNA-mRNA translocation and provide new insights into the mechanism of antibiotic action.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Ribosome Subunits/drug effects , Biological Transport , Cinnamates/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Hygromycin B/analogs & derivatives , Hygromycin B/pharmacology , Kanamycin/pharmacology , Kinetics , Neomycin/pharmacology , Paromomycin/pharmacology , Peptide Elongation Factor G/genetics , Peptide Elongation Factor G/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Transfer/antagonists & inhibitors , RNA, Transfer/chemistry , RNA, Transfer/genetics , Ribosome Subunits/genetics , Ribosome Subunits/metabolism , Ribosome Subunits/ultrastructure , Spectinomycin/pharmacology , Streptomycin/pharmacology , Viomycin/pharmacology
3.
J Phys Chem A ; 125(8): 1720-1737, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33620220

ABSTRACT

The viomycin biosynthesis enzyme VioC is a nonheme iron and α-ketoglutarate-dependent dioxygenase involved in the selective hydroxylation of l-arginine at the C3-position for antibiotics biosynthesis. Interestingly, experimental studies showed that using the substrate analogue, namely, l-homo-arginine, a mixture of products was obtained originating from C3-hydroxylation, C4-hydroxylation, and C3-C4-desaturation. To understand how the addition of one CH2 group to a substrate can lead to such a dramatic change in selectivity and activity, we decided to perform a computational study using quantum mechanical (QM) cluster models. We set up a large active-site cluster model of 245 atoms that includes the oxidant with its first- and second-coordination sphere influences as well as the substrate binding pocket. The model was validated against experimental work from the literature on related enzymes and previous computational studies. Thereafter, possible pathways leading to products and byproducts were investigated for a model containing l-Arg and one for l-homo-Arg as substrate. The calculated free energies of activation predict product distributions that match the experimental observation and give a low-energy C3-hydroxylation pathway for l-Arg, while for l-homo-Arg, several barriers are found to be close in energy leading to a mixture of products. We then analyzed the origins of the differences in product distributions using thermochemical, valence bond, and electrostatic models. Our studies show that the C3-H and C4-H bond strengths of l-Arg and l-homo-Arg are similar; however, external perturbations from an induced electric field of the protein affect the relative C-H bond strengths of l-Arg dramatically and make the C3-H bond the weakest and guide the reaction to a selective C3-hydroxylation channel. Therefore, the charge distribution in the protein and the induced electric dipole field of the active site of VioC guides the l-Arg substrate activation to C3-hydroxylation and disfavors the C4-hydroxylation pathway, while this does not occur for l-homo-Arg. Tight substrate positioning and electrostatic perturbations from the second-coordination sphere residues in VioC also result in a slower overall reaction for l-Arg; however, they enable a high substrate selectivity. Our studies highlight the importance of the second-coordination sphere in proteins that position the substrate and oxidant, perturb charge distributions, and enable substrate selectivity.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Iron/metabolism , Nonheme Iron Proteins/chemistry , Nonheme Iron Proteins/metabolism , Oxygenases/chemistry , Oxygenases/metabolism , Static Electricity , Viomycin/biosynthesis , Catalytic Domain , Hydroxylation , Models, Molecular
4.
J Biomol Struct Dyn ; 39(10): 3802-3811, 2021 07.
Article in English | MEDLINE | ID: mdl-32406317

ABSTRACT

The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) which was first reported in Wuhan province of China, has become a deadly pandemic causing alarmingly high morbidity and mortality. In the absence of new targeted drugs and vaccines against SARS-CoV-2 at present, the choices for effective treatments are limited. Therefore, considering the exigency of the situation, we focused on identifying the available approved drugs as potential inhibitor against the promising Coronavirus drug target, the Main Protease, using computer-aided methods. We created a library of U. S. Food and Drug Administration approved anti-microbial drugs and virtually screened it against the available crystal structures of Main Protease of the virus. The study revealed that Viomycin showed the highest -CDocker energy after docking at the active site of SARS-CoV-2 Main Protease. It is noteworthy that Viomycin showed higher -CDocker energy as compared to the drugs currently under clinical trial for SARS-CoV-2 treatment viz. Ritonavir and Lopinavir. Additionally, Viomycin formed higher number of H-bonds with SARS-CoV-2 Main Protease than its co-crystallised inhibitor compound N3. Molecular dynamics simulation further showed that Viomycin embedded deeply inside the binding pocket and formed robust binding with SARS-CoV-2 Main Protease. Therefore, we propose that Viomycin may act as a potential inhibitor of the Main Protease of SARS-CoV-2. Further optimisations with the drug may support the much-needed rapid response to mitigate the pandemic.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases/antagonists & inhibitors , Protease Inhibitors , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Drug Repositioning , Molecular Docking Simulation , Protease Inhibitors/pharmacology , Viomycin/pharmacology
5.
Proc Natl Acad Sci U S A ; 117(19): 10271-10277, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32341159

ABSTRACT

Viomycin, an antibiotic that has been used to fight tuberculosis infections, is believed to block the translocation step of protein synthesis by inhibiting ribosomal subunit dissociation and trapping the ribosome in an intermediate state of intersubunit rotation. The mechanism by which viomycin stabilizes this state remains unexplained. To address this, we have determined cryo-EM and X-ray crystal structures of Escherichia coli 70S ribosome complexes trapped in a rotated state by viomycin. The 3.8-Å resolution cryo-EM structure reveals a ribosome trapped in the hybrid state with 8.6° intersubunit rotation and 5.3° rotation of the 30S subunit head domain, bearing a single P/E state transfer RNA (tRNA). We identify five different binding sites for viomycin, four of which have not been previously described. To resolve the details of their binding interactions, we solved the 3.1-Å crystal structure of a viomycin-bound ribosome complex, revealing that all five viomycins bind to ribosomal RNA. One of these (Vio1) corresponds to the single viomycin that was previously identified in a complex with a nonrotated classical-state ribosome. Three of the newly observed binding sites (Vio3, Vio4, and Vio5) are clustered at intersubunit bridges, consistent with the ability of viomycin to inhibit subunit dissociation. We propose that one or more of these same three viomycins induce intersubunit rotation by selectively binding the rotated state of the ribosome at dynamic elements of 16S and 23S rRNA, thus, blocking conformational changes associated with molecular movements that are required for translocation.


Subject(s)
Escherichia coli/metabolism , Protein Biosynthesis , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Viomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , Escherichia coli/drug effects , Escherichia coli/growth & development , Models, Molecular , Molecular Conformation , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal/genetics , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomal Proteins/metabolism , Ribosomes/chemistry
6.
Elife ; 82019 06 07.
Article in English | MEDLINE | ID: mdl-31172942

ABSTRACT

Applying pre-steady state kinetics to an Escherichia-coli-based reconstituted translation system, we have studied how the antibiotic viomycin affects the accuracy of genetic code reading. We find that viomycin binds to translating ribosomes associated with a ternary complex (TC) consisting of elongation factor Tu (EF-Tu), aminoacyl tRNA and GTP, and locks the otherwise dynamically flipping monitoring bases A1492 and A1493 into their active conformation. This effectively prevents dissociation of near- and non-cognate TCs from the ribosome, thereby enhancing errors in initial selection. Moreover, viomycin shuts down proofreading-based error correction. Our results imply a mechanism in which the accuracy of initial selection is achieved by larger backward rate constants toward TC dissociation rather than by a smaller rate constant for GTP hydrolysis for near- and non-cognate TCs. Additionally, our results demonstrate that translocation inhibition, rather than error induction, is the major cause of cell growth inhibition by viomycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Viomycin/pharmacology , Cell-Free System
7.
Nucleic Acids Res ; 45(17): 10168-10177, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973468

ABSTRACT

Bacterial ribosome recycling requires breakdown of the post-termination complex (PoTC), comprising a messenger RNA (mRNA) and an uncharged transfer RNA (tRNA) cognate to the terminal mRNA codon bound to the 70S ribosome. The translation factors, elongation factor G and ribosome recycling factor, are known to be required for recycling, but there is controversy concerning whether these factors act primarily to effect the release of mRNA and tRNA from the ribosome, with the splitting of the ribosome into subunits being somewhat dispensable, or whether their main function is to catalyze the splitting reaction, which necessarily precedes mRNA and tRNA release. Here, we utilize three assays directly measuring the rates of mRNA and tRNA release and of ribosome splitting in several model PoTCs. Our results largely reconcile these previously held views. We demonstrate that, in the absence of an upstream Shine-Dalgarno (SD) sequence, PoTC breakdown proceeds in the order: mRNA release followed by tRNA release and then by 70S splitting. By contrast, in the presence of an SD sequence all three processes proceed with identical apparent rates, with the splitting step likely being rate-determining. Our results are consistent with ribosome profiling results demonstrating the influence of upstream SD-like sequences on ribosome occupancy at or just before the mRNA stop codon.


Subject(s)
Escherichia coli/genetics , Models, Biological , Ribosomes/metabolism , Bacterial Proteins/metabolism , Codon, Terminator , Escherichia coli/metabolism , Fluorescence Polarization , Fusidic Acid/pharmacology , Guanosine Triphosphate/metabolism , Kinetics , Peptide Elongation Factor G/metabolism , Prokaryotic Initiation Factor-3/metabolism , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , RNA, Transfer/metabolism , Ribosome Subunits/metabolism , Ribosomes/drug effects , Thiostrepton/pharmacology , Viomycin/pharmacology
8.
Dalton Trans ; 45(20): 8645-58, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27143296

ABSTRACT

Viomycin is a basic peptide antibiotic, which is among the most effective agents against multidrug-resistant tuberculosis. In this paper we provide the characteristics of its acid base properties, coordination preferences towards the Cu(ii) ions, as well as the reactivity of the resulting complexes against plasmid DNA and HDV ribozyme. Careful coordination studies throughout the wide pH range allow for the characterisation of all the Cu(ii)-viomycin complex species. The assignment of proton chemical shifts was achieved by NMR experiments, while the DTF level of theory was applied to support molecular structures of the studied complexes. The experiments with the plasmid DNA reveal that at the physiological levels of hydrogen peroxide the Cu(ii)-viomycin complex is more aggressive against DNA than uncomplexed metal ions. Moreover, the degradation of DNA by viomycin can be carried out without the presence of transition metal ions. In the studies of antigenomic delta ribozyme catalytic activity, viomycin and its complex are shown to modulate the ribozyme functioning. The molecular modelling approach allows the indication of two different locations of viomycin binding sites to the ribozyme.


Subject(s)
Antitubercular Agents/chemistry , Coordination Complexes/chemistry , Copper/chemistry , RNA, Catalytic/metabolism , Viomycin/chemistry , Antitubercular Agents/pharmacology , Binding Sites , Circular Dichroism , Coordination Complexes/pharmacology , DNA Fragmentation/drug effects , Electron Spin Resonance Spectroscopy , Hydrogen Bonding , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Molecular Conformation , Nucleic Acid Conformation , Potentiometry , RNA, Catalytic/chemistry , Viomycin/pharmacology
9.
Proc Natl Acad Sci U S A ; 113(4): 978-83, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26755601

ABSTRACT

Viomycin is a tuberactinomycin antibiotic essential for treating multidrug-resistant tuberculosis. It inhibits bacterial protein synthesis by blocking elongation factor G (EF-G) catalyzed translocation of messenger RNA on the ribosome. Here we have clarified the molecular aspects of viomycin inhibition of the elongating ribosome using pre-steady-state kinetics. We found that the probability of ribosome inhibition by viomycin depends on competition between viomycin and EF-G for binding to the pretranslocation ribosome, and that stable viomycin binding requires an A-site bound tRNA. Once bound, viomycin stalls the ribosome in a pretranslocation state for a minimum of ∼ 45 s. This stalling time increases linearly with viomycin concentration. Viomycin inhibition also promotes futile cycles of GTP hydrolysis by EF-G. Finally, we have constructed a kinetic model for viomycin inhibition of EF-G catalyzed translocation, allowing for testable predictions of tuberactinomycin action in vivo and facilitating in-depth understanding of resistance development against this important class of antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Peptide Elongation Factor G/antagonists & inhibitors , Protein Biosynthesis/drug effects , Viomycin/pharmacology , Bacteria/metabolism , Dose-Response Relationship, Drug , Guanosine Triphosphate/chemistry , Probability , Ribosomes/drug effects , Ribosomes/metabolism , Viomycin/metabolism
10.
RNA ; 22(1): 49-60, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26554029

ABSTRACT

Here we report that the specificity of peptide release in the ribosome on a nonstop mRNA by ArfA and RF2 is achieved by an induced-fit mechanism. Using RF2 that is methylated on the glutamine of its GGQ motif (RF2(m)), we show that methylation substantially increases the rate of ArfA/RF2-catalyzed peptide release on a nonstop mRNA that does not occupy the ribosomal A site, but has only a modest effect on k(cat) by the same proteins on longer nonstop mRNAs occupying the A site of the mRNA channel in the ribosome. Our data suggest that enhancement in the kcat of peptide release by ArfA and RF2 under the cognate decoding condition is the result of favorable conformational changes in the nonstop complex. We demonstrate a shared mechanism between canonical and nonstop termination, supported by similarities in the kinetic mechanisms in antibiotic inhibition and methylation-correlated enhancement in the rate of peptide release. Despite these similarities, our data suggest that nonstop termination differs from canonical pathway in the downstream event of recycling.


Subject(s)
Escherichia coli Proteins/metabolism , Peptide Termination Factors/metabolism , Peptides/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Biocatalysis , Escherichia coli/metabolism , Methylation , Paromomycin/pharmacology , Peptide Chain Termination, Translational , Ribosomes/metabolism , Viomycin/pharmacology
11.
Proc Natl Acad Sci U S A ; 111(42): 15060-5, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25288752

ABSTRACT

Translocation of mRNA and tRNAs through the ribosome is catalyzed by a universally conserved elongation factor (EF-G in prokaryotes and EF-2 in eukaryotes). Previous studies have suggested that ribosome-bound EF-G undergoes significant structural rearrangements. Here, we follow the movement of domain IV of EF-G, which is critical for the catalysis of translocation, relative to protein S12 of the small ribosomal subunit using single-molecule FRET. We show that ribosome-bound EF-G adopts distinct conformations corresponding to the pre- and posttranslocation states of the ribosome. Our results suggest that, upon ribosomal translocation, domain IV of EF-G moves toward the A site of the small ribosomal subunit and facilitates the movement of peptidyl-tRNA from the A to the P site. We found no evidence of direct coupling between the observed movement of domain IV of EF-G and GTP hydrolysis. In addition, our results suggest that the pretranslocation conformation of the EF-G-ribosome complex is significantly less stable than the posttranslocation conformation. Hence, the structural rearrangement of EF-G makes a considerable energetic contribution to promoting tRNA translocation.


Subject(s)
Peptide Elongation Factor G/metabolism , Ribosomes/metabolism , Biological Transport , Catalysis , Fluorescence Resonance Energy Transfer , Guanosine Triphosphate/chemistry , Microscopy , Protein Binding , Protein Structure, Tertiary , Protein Synthesis Inhibitors/chemistry , Protein Transport , RNA, Messenger/metabolism , RNA, Transfer/chemistry , Ribosomes/chemistry , Viomycin/chemistry
12.
Nat Commun ; 5: 4891, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25205267

ABSTRACT

The antibiotic streptomycin is widely used in the treatment of microbial infections. The primary mechanism of action is inhibition of translation by binding to the ribosome, but how it enters the bacterial cell is unclear. Early in the study of this antibiotic, a mysterious streptomycin-induced potassium efflux preceding any decrease in viability was observed; it was speculated that this changed the electrochemical gradient such that streptomycin better accessed the cytoplasm. Here we use a high-throughput screen to search for compounds targeting the mechanosensitive channel of large conductance (MscL) and find dihydrostreptomycin among the 'hits'. Furthermore, we find that MscL is not only necessary for the previously described streptomycin-induced potassium efflux, but also directly increases MscL activity in electrophysiological studies. The data suggest that gating MscL is a novel mode of action of dihydrostreptomycin, and that MscL's large pore may provide a mechanism for cell entry.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dihydrostreptomycin Sulfate/pharmacology , Escherichia coli Proteins/drug effects , Escherichia coli/drug effects , Ion Channels/drug effects , Potassium/metabolism , Dihydrostreptomycin Sulfate/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , High-Throughput Screening Assays , Ion Channels/metabolism , Patch-Clamp Techniques , Spectinomycin/pharmacology , Streptomycin/metabolism , Streptomycin/pharmacology , Viomycin/pharmacology
13.
Article in English | MEDLINE | ID: mdl-23545648

ABSTRACT

Nonribosomal peptide synthetases (NRPSs) are large multimodular enzymes that synthesize important secondary metabolites such as antibiotics. NRPSs follow a modular synthetic logic whereby each successive amino-acid monomer is added to the peptide chain by successive multi-domain modules. The condensation domain catalyzes the central chemical event in the synthetic cycle, peptide-bond formation, and is present in every elongation module of the NRPS. Viomycin is an antituberculosis nonribosomal peptide that is synthesized by a series of four NRPS proteins and then modified by tailoring proteins. In order to study the mechanisms of peptide-bond formation in viomycin and in NRPSs in general, a structural study of the first condensation domain of the viomycin synthetase protein VioA (VioA-C1) was initiated. The gene for VioA-C1 was cloned from genomic DNA of Streptomyces vinaceus, expressed as an octahistidine-tagged construct and purified by column chromatography. VioA-C1 was crystallized using the sitting-drop vapor-diffusion method. X-ray diffraction data were collected on a rotating-anode source to 2.9 Å resolution. The data could be indexed in the orthorhombic space group P212121, with unit-cell parameters a = 46.165, b = 68.335, c = 146.423 Å. There is likely to be one monomer in the asymmetric unit, giving a solvent content of 49.2% and a Matthews coefficient (VM) of 2.42 Å(3) Da(-1). Structural determination is in progress.


Subject(s)
Peptide Synthases/chemistry , Streptomyces/enzymology , Crystallization , Crystallography, X-Ray , Peptide Synthases/metabolism , Viomycin/biosynthesis
14.
Mol Microbiol ; 85(6): 1194-203, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22779429

ABSTRACT

The binding site of the cyclic peptide antibiotics capreomycin and viomycin is located on the ribosomal subunit interface close to nucleotides C1409 in 16S rRNA and C1920 in 23S rRNA. In Mycobacterium tuberculosis, the 2'-hydroxyls of both nucleotides are methylated by the enzyme TlyA. Loss of these methylations through inactivation of TlyA confers resistance to capreomycin and viomycin. We report here that TlyA orthologues occur in diverse bacteria and fall into two distinct groups. One group, now termed TlyA(I) , has shorter N- and C-termini and methylates only C1920; the second group (now TlyA(II) ) includes the mycobacterial enzyme, and these longer orthologues methylate at both C1409 and C1920. Ribosomal subunits are the preferred substrates for both groups of orthologues. Amino acid substitutions at the N-terminus of TlyA(II) reduce its ability to methylate these substrates. Growing pairs of recombinant TlyA(II) Escherichia coli strains in competition shows that even subtle changes in the level of rRNA methylation lead to significant differences in susceptibility to sub-inhibitory concentrations of capreomycin. The findings reveal that 2'-O-methyls at both C1409 and C1920 play a role in facilitating the inhibitory effects of capreomycin and viomycin on the bacterial ribosome.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/enzymology , Bacterial Proteins/metabolism , Capreomycin/pharmacology , RNA, Ribosomal/metabolism , tRNA Methyltransferases/metabolism , Bacterial Proteins/genetics , Methylation , Microbial Sensitivity Tests , Models, Molecular , Nucleic Acid Conformation , Ribosome Subunits/metabolism , Viomycin/pharmacology , tRNA Methyltransferases/genetics
15.
RNA ; 18(2): 230-40, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22187675

ABSTRACT

The class II release factor RF3 is a GTPase related to elongation factor EF-G, which catalyzes release of class I release factors RF1 and RF2 from the ribosome after termination of protein synthesis. The 3.3 Å crystal structure of the RF3·GDPNP·ribosome complex provides a high-resolution description of interactions and structural rearrangements that occur when binding of this translational GTPase induces large-scale rotational movements in the ribosome. RF3 induces a 7° rotation of the body and 14° rotation of the head of the 30S ribosomal subunit, and itself undergoes inter- and intradomain conformational rearrangements. We suggest that ordering of critical elements of switch loop I and the P loop, which help to form the GTPase catalytic site, are caused by interactions between the G domain of RF3 and the sarcin-ricin loop of 23S rRNA. The rotational movements in the ribosome induced by RF3, and its distinctly different binding orientation to the sarcin-ricin loop of 23S rRNA, raise interesting implications for the mechanism of action of EF-G in translocation.


Subject(s)
Escherichia coli Proteins/chemistry , Guanosine Triphosphate/chemistry , Peptide Termination Factors/chemistry , Ribosomes/chemistry , Catalytic Domain , Crystallography, X-Ray , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Guanosine Triphosphate/metabolism , Models, Molecular , Peptide Elongation Factor G/metabolism , Peptide Termination Factors/metabolism , Protein Binding/drug effects , Protein Biosynthesis/drug effects , Protein Structure, Tertiary/drug effects , RNA, Ribosomal, 23S/metabolism , Ribosomes/metabolism , Translocation, Genetic/drug effects , Translocation, Genetic/genetics , Viomycin/pharmacology
17.
Antimicrob Agents Chemother ; 55(10): 4712-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21768509

ABSTRACT

Capreomycin and the structurally similar compound viomycin are cyclic peptide antibiotics which are particularly active against Mycobacterium tuberculosis, including multidrug resistant strains. Both antibiotics bind across the ribosomal interface involving 23S rRNA helix 69 (H69) and 16S rRNA helix 44 (h44). The binding site of tuberactinomycins in h44 partially overlaps with that of aminoglycosides, and they share with these drugs the side effect of irreversible hearing loss. Here we studied the drug target interaction on ribosomes modified by site-directed mutagenesis. We identified rRNA residues in h44 as the main determinants of phylogenetic selectivity, predict compensatory evolution to impact future resistance development, and propose mechanisms involved in tuberactinomycin ototoxicity, which may enable the development of improved, less-toxic derivatives.


Subject(s)
Antitubercular Agents/pharmacology , Capreomycin/pharmacology , Mycobacterium tuberculosis/drug effects , Ribosomes/drug effects , Viomycin/pharmacology , Aminoglycosides/pharmacology , Antitubercular Agents/metabolism , Antitubercular Agents/toxicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Capreomycin/metabolism , Capreomycin/toxicity , Drug Resistance, Multiple, Bacterial/genetics , Enviomycin/analogs & derivatives , Enviomycin/pharmacology , Enviomycin/toxicity , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , RNA, Ribosomal, 16S/metabolism , RNA, Ribosomal, 23S/metabolism , Viomycin/metabolism , Viomycin/toxicity
18.
Biochemistry ; 49(45): 9732-8, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-20886842

ABSTRACT

Viomycin belongs to the tuberactinomycin family of antibiotics against tuberculosis. However, its inhibition mechanism remains elusive. Although it is clear that viomycin inhibits the ribosome intersubunit ratcheting, there are contradictory reports about whether the antibiotic viomycin stabilizes the tRNA hybrid or classical state. By using a single-molecule FRET method to directly observe the tRNA dynamics relative to ribosomal protein L27, we have found that viomycin trapped the hybrid state within certain ribosome subgroups but did not significantly suppress the tRNA dynamics. The persistent fluctuation of tRNA implied that tRNA motions were decoupled from the ribosome intersubunit ratcheting. Viomycin also promoted peptidyl-tRNA fluctuation in the posttranslocation complex, implying that, in addition to acylated P-site tRNA, the decoding center also played an important role of ribosome locking after translocation. Therefore, viomycin inhibits translocation by trapping the hybrid state in the pretranslocation complex and disturbing the stability of posttranslocation complex. Our results imply that ribosome translocation is possibly a synergistic process of multiple decoupled local dynamics.


Subject(s)
Ribosomes/drug effects , Viomycin/pharmacology , Biological Transport/drug effects , Fluorescence Resonance Energy Transfer/methods , Oligopeptides/biosynthesis , Oligopeptides/metabolism , Peptide Elongation Factor G/genetics , Peptide Elongation Factor G/metabolism , Protein Biosynthesis/drug effects , Protein Transport , RNA, Messenger/genetics , RNA, Transfer/drug effects , RNA, Transfer/genetics , Ribosomes/genetics , Ribosomes/metabolism , Translocation, Genetic/drug effects
19.
Biochemistry ; 49(41): 8815-7, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20845982

ABSTRACT

The biosynthesis of many natural products of clinical interest involves large, multidomain enzymes called nonribosomal peptide synthetases (NRPSs). In bacteria, many of the gene clusters coding for NRPSs also code for a member of the MbtH-like protein superfamily, which are small proteins of unknown function. Using MbtH-like proteins from three separate NRPS systems, we show that these proteins copurify with the NRPSs and influence amino acid activation. As a consequence, MbtH-like proteins are integral components of NRPSs.


Subject(s)
Bacteria/enzymology , Bacterial Proteins/metabolism , Capreomycin/biosynthesis , Multigene Family/physiology , Peptide Synthases/metabolism , Viomycin/biosynthesis , Bacteria/genetics , Bacterial Proteins/genetics , Peptide Synthases/genetics
20.
Nat Struct Mol Biol ; 17(3): 289-93, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20154709

ABSTRACT

Viomycin and capreomycin belong to the tuberactinomycin family of antibiotics, which are among the most effective antibiotics against multidrug-resistant tuberculosis. Here we present two crystal structures of the 70S ribosome in complex with three tRNAs and bound to either viomycin or capreomycin at 3.3- and 3.5-A resolution, respectively. Both antibiotics bind to the same site on the ribosome, which lies at the interface between helix 44 of the small ribosomal subunit and helix 69 of the large ribosomal subunit. The structures of these complexes suggest that the tuberactinomycins inhibit translocation by stabilizing the tRNA in the A site in the pretranslocation state. In addition, these structures show that the tuberactinomycins bind adjacent to the binding sites for the paromomycin and hygromycin B antibiotics, which may enable the development of new derivatives of tuberactinomycins that are effective against drug-resistant strains.


Subject(s)
Capreomycin/chemistry , Capreomycin/metabolism , Ribosomes/metabolism , Thermus thermophilus/metabolism , Viomycin/chemistry , Viomycin/metabolism , Antitubercular Agents/chemistry , Antitubercular Agents/metabolism , Crystallography, X-Ray , Molecular Sequence Data , Molecular Structure , Protein Binding , Protein Structure, Secondary , RNA, Transfer/genetics , Ribosomes/chemistry , Ribosomes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...