Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
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
2.
Drug Des Devel Ther ; 14: 4579-4591, 2020.
Article in English | MEDLINE | ID: mdl-33149555

ABSTRACT

PURPOSE: Steroids are known to inhibit osteogenic differentiation and subsequent bone formation in bone mesenchymal stem cells (BMSCs). However, little is known about the role of BMSC exosomes (Exos) and tRNA-derived small RNAs (tsRNAs) in steroid-induced osteonecrosis of the femoral head (SONFH). The objective of this study was to characterize the tsRNA expression profiles of plasma Exos collected from SONFH patients and healthy individuals using small RNA sequencing and further explore the effect of BMSC Exos carrying specific tsRNAs on osteogenic differentiation. MATERIALS AND METHODS: Based on insights from small RNA sequencing, five differentially expressed (DE) tsRNAs were selected for quantitative real-time polymerase chain reaction (qRT-PCR). The regulatory networks associated with interactions of the tsRNAs-mRNA-pathways were reconstructed. The osteogenesis and adipogenesis in BMSCs were detected via ALP and oil red O staining methods, respectively. RESULTS: A total of 345 DE small RNAs were screened, including 223 DE tsRNAs. The DE tsRNAs were enriched in Wnt signaling pathway and osteogenic differentiation. We identified five DE tsRNAs, among which tsRNA-10277 was significantly downregulated in plasma Exos of SONFH patients compared to that in healthy individuals. Dexamethasone-induced BMSCs were associated with an increased fraction of lipid droplets and decreased osteogenic differentiation, whereas BMSC Exos restored the osteogenic differentiation of that. After treatment of tsRNA-10277-loaded BMSC Exos, the lipid droplets and osteogenic differentiation ability were found to be decreased and enhanced in dexamethasone-induced BMSCs, respectively. CONCLUSION: An altered tsRNA profile might be involved in the pathophysiology of SONFH. tsRNA-10277-loaded BMSC Exos enhanced osteogenic differentiation ability of dexamethasone-induced BMSCs. Our results provide novel insights into the osteogenic effect of BMSC Exos carrying specific tsRNAs on SONFH.


Subject(s)
Dexamethasone/adverse effects , Exosomes/drug effects , Femur Head/drug effects , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , RNA, Transfer/antagonists & inhibitors , Adult , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Exosomes/metabolism , Female , Femur Head/metabolism , Humans , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Molecular Structure , RNA, Transfer/genetics , RNA, Transfer/metabolism , Sequence Analysis, RNA , Structure-Activity Relationship
3.
J Comput Aided Mol Des ; 34(3): 281-291, 2020 03.
Article in English | MEDLINE | ID: mdl-31832846

ABSTRACT

Linezolid, an antibiotic of oxazolidinone family, is a translation inhibitor. The mechanism of its action that consists in preventing the binding of aminoacyl-tRNA to the A-site of the large subunit of a ribosome was embraced on the basis of the X-ray structural analysis of the linezolid complexes with vacant bacterial ribosomes. However, the known structures of the linezolid complexes with bacterial ribosomes poorly explain the linezolid selectivity in suppression of protein biosynthesis, depending on the amino acid sequence of the nascent peptide. In the present study the most probable structure of the linezolid complex with a E. coli ribosome in the A,A/P,P-state that is in line with the results of biochemical studies of linezolid action has been obtained by molecular dynamics simulation methods.


Subject(s)
Anti-Bacterial Agents/chemistry , Linezolid/chemistry , Protein Biosynthesis/drug effects , RNA, Transfer/chemistry , Amino Acid Sequence/genetics , Binding Sites/drug effects , Crystallography, X-Ray , Escherichia coli/drug effects , Molecular Dynamics Simulation , Protein Binding/drug effects , RNA, Transfer/antagonists & inhibitors , RNA, Transfer/genetics , Ribosomes/chemistry , Ribosomes/drug effects , Ribosomes/genetics
4.
Mol Nutr Food Res ; 62(9): e1700992, 2018 05.
Article in English | MEDLINE | ID: mdl-29573169

ABSTRACT

SCOPE: This study was undertaken to expand our insights into the mechanisms involved in the tolerance to resveratrol (RSV) that operate at system-level in gut microorganisms and advance knowledge on new RSV-responsive gene circuits. METHODS AND RESULTS: Whole genome transcriptional profiling was used to characterize the molecular response of Lactobacillus plantarum WCFS1 to RSV. DNA repair mechanisms were induced by RSV and responses were triggered to decrease the load of copper, a metal required for RSV-mediated DNA cleavage, and H2 S, a genotoxic gas. To counter the effects of RSV, L. plantarum strongly up- or downregulated efflux systems and ABC transporters pointing to transport control of RSV across the membrane as a key mechanism for RSV tolerance. L. plantarum also downregulated tRNAs, induced chaperones, and reprogrammed its transcriptome to tightly control ammonia levels. RSV induced a probiotic effector gene and a likely deoxycholate transporter, two functions that improve the host health status. CONCLUSION: Our data identify novel protective mechanisms involved in RSV tolerance operating at system level in a gut microbe. These insights could influence the way RSV is used for a better management of gut microbial ecosystems to obtain associated health benefits.


Subject(s)
Antioxidants/metabolism , Bacterial Proteins/metabolism , DNA Repair , Gene Expression Regulation, Bacterial , Lactobacillus plantarum/metabolism , Models, Biological , Resveratrol/metabolism , Animals , Bacterial Proteins/agonists , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Biological Transport , Copper/chemistry , Copper/metabolism , Coumaric Acids , Dietary Supplements , Gene Expression Profiling , Gene Ontology , Humans , Hydrogen Sulfide/antagonists & inhibitors , Hydrogen Sulfide/metabolism , Lactobacillus plantarum/growth & development , Lactobacillus plantarum/isolation & purification , Probiotics , Propionates/metabolism , RNA, Bacterial/antagonists & inhibitors , RNA, Bacterial/metabolism , RNA, Transfer/antagonists & inhibitors , RNA, Transfer/metabolism , Saliva/microbiology
5.
Nucleic Acids Res ; 45(12): 7507-7514, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28505372

ABSTRACT

The emergence of multi-drug resistant bacteria is limiting the effectiveness of commonly used antibiotics, which spurs a renewed interest in revisiting older and poorly studied drugs. Streptogramins A is a class of protein synthesis inhibitors that target the peptidyl transferase center (PTC) on the large subunit of the ribosome. In this work, we have revealed the mode of action of the PTC inhibitor madumycin II, an alanine-containing streptogramin A antibiotic, in the context of a functional 70S ribosome containing tRNA substrates. Madumycin II inhibits the ribosome prior to the first cycle of peptide bond formation. It allows binding of the tRNAs to the ribosomal A and P sites, but prevents correct positioning of their CCA-ends into the PTC thus making peptide bond formation impossible. We also revealed a previously unseen drug-induced rearrangement of nucleotides U2506 and U2585 of the 23S rRNA resulting in the formation of the U2506•G2583 wobble pair that was attributed to a catalytically inactive state of the PTC. The structural and biochemical data reported here expand our knowledge on the fundamental mechanisms by which peptidyl transferase inhibitors modulate the catalytic activity of the ribosome.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Peptidyl Transferases/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , RNA, Transfer/antagonists & inhibitors , Ribosomes/drug effects , Streptogramins/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Models, Molecular , Nucleic Acid Conformation , Peptidyl Transferases/chemistry , Peptidyl Transferases/genetics , Peptidyl Transferases/metabolism , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , RNA, Ribosomal, 23S/antagonists & inhibitors , RNA, Ribosomal, 23S/chemistry , RNA, Ribosomal, 23S/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Streptogramins/chemistry , Thermus thermophilus/drug effects , Thermus thermophilus/enzymology , Thermus thermophilus/genetics
6.
Ukr Biokhim Zh (1999) ; 84(5): 68-75, 2012.
Article in Russian | MEDLINE | ID: mdl-23342637

ABSTRACT

The influence of the penetrating cryoprotector and Mg2+ ions on the protein-synthesizing activity of postmitochondrial supernantant of the rat liver as well as on aminoacylation processes has been investigated. The addition of the penetrating cryoprotectors--ethylene glycol and DMSO--resulted in the concentration-dependant reversible inhibition of the protein biosynthesis and aminoacylation reaction in the cell-free system. These cryoprotectors at low concentrations intensified the stimulating effect of Mg2+ on the cumulative protein synthesis in the cell-free system.


Subject(s)
Dimethyl Sulfoxide/pharmacology , Ethylene Glycol/pharmacology , Magnesium/pharmacology , Protein Biosynthesis/drug effects , RNA, Transfer/antagonists & inhibitors , Amino Acids/metabolism , Aminoacylation/drug effects , Animals , Cell-Free System , Centrifugation , Chromatography, Gel , Dose-Response Relationship, Drug , Liver/drug effects , Liver/metabolism , Magnesium/metabolism , RNA, Transfer/metabolism , Rats
7.
Wiley Interdiscip Rev RNA ; 2(2): 209-32, 2011.
Article in English | MEDLINE | ID: mdl-21957007

ABSTRACT

The key role of the bacterial ribosome makes it an important target for antibacterial agents. Indeed, a large number of clinically useful antibiotics target this complex translational ribonucleoprotein machinery. The majority of these compounds, mostly of natural origin, bind to one of the three key ribosomal sites: the decoding (or A-site) on the 30S, the peptidyl transferase center (PTC) on the 50S, and the peptide exit tunnel on the 50S. Antibiotics that bind the A-site, such as the aminoglycosides, interfere with codon recognition and translocation. Peptide bond formation is inhibited when small molecules like oxazolidinones bind at the PTC. Finally, macrolides tend to block the growth of the amino acid chain at the peptide exit tunnel. In this article, the major classes of antibiotics that target the bacterial ribosome are discussed and classified according to their respective target. Notably, most antibiotics solely interact with the RNA components of the bacterial ribosome. The surge seen in the appearance of resistant bacteria has not been met by a parallel development of effective and broad-spectrum new antibiotics, as evident by the introduction of only two novel classes of antibiotics, the oxazolidinones and lipopeptides, in the past decades. Nevertheless, this significant health threat has revitalized the search for new antibacterial agents and novel targets. High resolution structural data of many ribosome-bound antibiotics provide unprecedented insight into their molecular contacts and mode of action and inspire the design and synthesis of new candidate drugs that target this fascinating molecular machine.


Subject(s)
Anti-Bacterial Agents/pharmacology , Protein Biosynthesis/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Base Sequence , Drug Delivery Systems/methods , Humans , Models, Biological , Models, Molecular , Molecular Targeted Therapy/methods , RNA, Ribosomal, 16S/antagonists & inhibitors , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/metabolism , RNA, Transfer/antagonists & inhibitors , RNA, Transfer/chemistry , RNA, Transfer/metabolism
8.
ACS Chem Biol ; 4(4): 299-307, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19278238

ABSTRACT

Peptoids that inhibit the group I intron RNA from Candida albicans, an opportunistic pathogen that kills immunocompromised hosts, have been identified using microarrays. The arrayed peptoid library was constructed using submonomers with moieties similar to ones found in small molecules known to bind RNA. Library members that passed quality control analysis were spotted onto a microarray and screened for binding to the C. albicans group I intron ribozyme. Each ligand binder identified from microarray-based screening inhibited self-splicing in the presence of 1 mM nucleotide concentration of bulk yeast tRNA with IC(50)'s between 150 and 2200 microM. The binding signals and the corresponding IC(50)'s were used to identify features in the peptoids that predispose them for RNA binding. After statistical analysis of the peptoids' structures that bind, a second generation of inhibitors was constructed using these important features; all second generation inhibitors have improved potencies with IC(50)'s of <100 microM. The most potent inhibitor is composed of one phenylguanidine and three tryptamine submonomers and has an IC(50) of 31 microM. This compound is 6-fold more potent than pentamidine, a clinically used drug that inhibits self-splicing. These results show that (i) modulators of RNA function can be identified by designing RNA-focused chemical libraries and screening them via microarray; (ii) statistical analysis of ligand binders can identify features in leads that predispose them for binding to their targets; and (iii) features can then be programmed into second generation inhibitors to design ligands with improved potencies.


Subject(s)
Candida albicans/drug effects , Introns/genetics , Oligonucleotide Array Sequence Analysis/methods , Peptoids/pharmacology , RNA, Catalytic/antagonists & inhibitors , RNA, Fungal/antagonists & inhibitors , RNA, Transfer/antagonists & inhibitors , Animals , Binding Sites , Candida albicans/enzymology , Candida albicans/pathogenicity , Dose-Response Relationship, Drug , Ligands , Molecular Conformation , Peptide Library , Peptoids/analogs & derivatives , Peptoids/chemistry , Pneumocystis carinii/drug effects , Pneumocystis carinii/genetics , RNA Splicing/drug effects , RNA, Catalytic/chemistry , RNA, Catalytic/genetics , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Transfer/chemistry , RNA, Transfer/genetics , Tetrahymena thermophila/drug effects , Tetrahymena thermophila/genetics
9.
Curr Pharm Biotechnol ; 9(3): 215-25, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18673287

ABSTRACT

Rana pipiens oocytes contain two homologues of pancreatic ribonuclease A that are cytostatic and cytotoxic to human cancer cells. Extensively studied Onconase is in advanced Phase IIIb clinical trials against malignant mesothelioma, while Amphinase is a novel enzyme in pre-clinical development. Onconase is the smallest (104 amino acid residues) member of the ribonuclease A superfamily while Amphinase (114 residues) is the largest among amphibian ribonucleases. Both enzymes share the characteristic frog ribonucleases C-terminal disulfide bond but another signature of this group, the N-terminal pyroglutamate, an integral part of Onconase active site is not conserved in Amphinase. Although Onconase and Amphinase are weak catalysts their enzymatic activities are required for cytostatic and cytotoxic activity. While it was postulated that tRNA is the primary substrate of Onconase in vivo there is also extensive indirect evidence that suggests other RNA species, in particular micro RNAs, may actually be the critical target of these ribonucleases. The cytostatic effects of Onconase and Amphinase are manifested as cell arrest in the G(1) cell cycle phase. Apoptosis then follows involving activation of endonucleases(s), caspases, serine proteases and transglutaminase. Onconase was shown to be strongly synergistic when combined with numerous other antitumor modalities. Onconase and Amphinase are highly cationic molecules and their preferential toxicity towards cancer cells (having distinctly higher negative charge compared to normal cells) may depend on increased binding efficiency to the cell surface by electrostatic interactions.


Subject(s)
Antineoplastic Agents/pharmacology , Oocytes/enzymology , Rana pipiens , Ribonucleases/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Clinical Trials as Topic , Enzyme Stability , Humans , Models, Molecular , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , RNA, Transfer/antagonists & inhibitors , Ribonucleases/chemistry , Ribonucleases/isolation & purification , Sequence Alignment
10.
Arch Virol ; 152(4): 655-68, 2007.
Article in English | MEDLINE | ID: mdl-17219019

ABSTRACT

Nucleic-acid-induced polymerisation of prion protein, when monitored by anilino naphthalene sulfonic acid dye, shows, successively, an immediate fluorescence increase of the dye upon mixing of the reactants, followed by a lag period in which the dye fluorescence remains unchanged, and then a phase in which dye fluorescence increases with time. The biological polyamines spermine and spermidine reduce the extent of the initial fluorescence increase, increase the lag period, and reduce both the rate and the extent of increase in fluorescence intensity of the dye in the final phase of the reaction. Spermidine is less effective than spermine in all of these processes. A nearly fivefold lower concentration of spermine can inhibit polymerisation of prion protein by tRNAs compared to the same process induced by double-stranded nucleic acid. The change in the secondary structure of the globular domain of the protein induced by nucleic acid is reversed by the addition of spermine, and it prevents structural destabilization of this domain induced by nucleic acids. It is suggested that physiological event(s) that would reduce the concentrations of intracellular biological amines may make nucleic acid available to induce oligomerization and polymerisation of cellular prion protein related to prion disease.


Subject(s)
Biogenic Polyamines/pharmacology , DNA/metabolism , Prions/metabolism , RNA, Transfer/metabolism , Spermidine/pharmacology , Spermine/pharmacology , Anilino Naphthalenesulfonates/metabolism , Biopolymers , Circular Dichroism , DNA/antagonists & inhibitors , Fluorescent Dyes/metabolism , Prions/chemistry , Protein Folding , Protein Structure, Secondary , RNA, Transfer/antagonists & inhibitors , Temperature
11.
Biochimie ; 87(9-10): 885-8, 2005.
Article in English | MEDLINE | ID: mdl-16164995

ABSTRACT

Human tRNA3(Lys) is used by HIV virus as a primer for the reverse transcription of its genome. The 18 nucleotides at the 3'-end of the tRNA3(Lys) are hybridized to a complementary sequence of the viral RNA called the primer-binding site. A screen against the human tRNA3(Lys) over a peptide library designed to target RNA has been performed. Of the 175 hexapeptides tested, three were found to bind to the d-stem of tRNA3(Lys). Alanine-scanning was used to define the determinants of the interaction between the peptides and tRNA3(Lys). They also bind to two other tested tRNAs, also at the level of the d-stem and loop, although the nucleotide sequence of the stem differs in one of them. These short peptides thus recognize specific structural features within the d-stem and loop of tRNAs. Associated with other pharmacophores, they could be useful to design optimized ligands targeting specific tRNAs such as retroviral replication primers.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Peptides/metabolism , RNA, Transfer/metabolism , Binding Sites , DNA Primers , HIV-1/genetics , Humans , Ligands , Nucleic Acid Conformation , Peptide Library , RNA, Transfer/antagonists & inhibitors , RNA, Transfer/chemistry , Reverse Transcription
12.
Article in English | MEDLINE | ID: mdl-12784065

ABSTRACT

The ever-growing resistance of pathogens to antibiotics and the lack of potent antibacterial drugs constitute major problems in the treatment of infectious diseases. Thus, the better understanding of the mode of action of antibiotics at the molecular level is of essential importance. Accumulating evidence points towards RNA as being a crucial target of antibacterial and antiviral drugs. Interestingly, aminoglycosides, one of the most important families of antibiotics, apart from their inhibitory effect on ribosome function, reportedly interfere with various RNA molecules and in vitro suppress the proliferation of human keratinocytes. In this study we investigated the effect of the aminoglycosides neomycin B, paromomycin, tobramycin and gentamycin on ribonuclease P activity from normal human epidermal keratinocytes. All aminoglycosides tested revealed a dose-dependent inhibition of tRNA maturation, which was reduced by increasing Mg(2+) ion concentrations, indicating competition of the cationic aminoglycosides with magnesium ions required for catalysis. Our in vitro findings suggest that the inhibitory effects of aminoglycosides on tRNA processing may be implicated in the mechanisms of their antiproliferative action on human epidermal keratinocytes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Epidermis/drug effects , Keratinocytes/drug effects , RNA, Transfer/antagonists & inhibitors , Anti-Bacterial Agents/administration & dosage , Carbohydrate Sequence , Cell-Free System , Cells, Cultured , Dose-Response Relationship, Drug , Endoribonucleases/antagonists & inhibitors , Epidermal Cells , Epidermis/metabolism , Gentamicins/chemistry , Gentamicins/pharmacology , Humans , Keratinocytes/metabolism , Magnesium/chemistry , Magnesium/pharmacology , Molecular Sequence Data , Neomycin/chemistry , Neomycin/pharmacology , Paromomycin/chemistry , Paromomycin/pharmacology , RNA, Catalytic/antagonists & inhibitors , RNA, Transfer/metabolism , Ribonuclease P , Structure-Activity Relationship , Tobramycin/chemistry , Tobramycin/pharmacology
13.
Biochemistry ; 40(3): 603-8, 2001 Jan 23.
Article in English | MEDLINE | ID: mdl-11170376

ABSTRACT

2,2'-p-Phenylene bis[6-(4-methyl-1-piperazinyl)]benzimidazole, 2,2'-bis(3,5-dihydroxyphenyl)-6,6'-bis benzimidazole, and 2,2'-bis(4-hydroxyphenyl)-6,6'-bis benzimidazole are shown by UV-visible and fluorescence spectrophotometry to be strong ligands for tRNA, giving simple, hyperbolic binding isotherms with apparent dissociation constants in the micromolar range. Hydroxyl radical footprinting indicates that they may bind in the D and T loops. On the basis of this tRNA recognition as a rationale, they were tested as inhibitors of the processing of precursor tRNAs by the RNA subunit of Escherichia coli RNase P (M1 RNA). Preliminary studies show that inhibition of the processing of Drosophila tRNA precursor molecules by phosphodiester bond cleavage, releasing the extraneous 5'-portion of RNA and the mature tRNA molecule, was dependent on both the structure of the inhibitor and the structure of the particular tRNA precursor substrate for tRNA(Ala), tRNA(Val), and tRNA(His). In more detailed followup using the tRNA(His) precursor as the substrate, experiments to determine the concentration dependence of the reaction showed that inhibition took time to reach its maximum extent. I(50) values (concentrations for 50% inhibition) were between 5.3 and 20.8 microM, making these compounds among the strongest known inhibitors of this ribozyme, and the first inhibitors of it not based on natural products. These compounds effect their inhibition by binding to the substrate of the enzyme reaction, making them examples of an unusual class of enzyme inhibitors. They provide novel, small-molecule, inhibitor frameworks for this endoribonuclease ribozyme.


Subject(s)
Endoribonucleases/metabolism , Enzyme Inhibitors/metabolism , Escherichia coli Proteins , Protein Processing, Post-Translational/drug effects , RNA Precursors/metabolism , RNA, Bacterial/metabolism , RNA, Catalytic/metabolism , RNA, Transfer/metabolism , Binding Sites , Bisbenzimidazole/metabolism , DNA Footprinting , Endoribonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Escherichia coli/enzymology , Ligands , RNA Precursors/antagonists & inhibitors , RNA, Bacterial/antagonists & inhibitors , RNA, Catalytic/antagonists & inhibitors , RNA, Transfer/antagonists & inhibitors , RNA, Transfer, Ala/antagonists & inhibitors , RNA, Transfer, Ala/metabolism , RNA, Transfer, His/antagonists & inhibitors , RNA, Transfer, His/metabolism , RNA, Transfer, Phe/antagonists & inhibitors , RNA, Transfer, Phe/metabolism , RNA, Transfer, Val/antagonists & inhibitors , RNA, Transfer, Val/metabolism , Ribonuclease P , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Substrate Specificity
15.
Biochim Biophys Acta ; 1092(2): 218-25, 1991 Apr 17.
Article in English | MEDLINE | ID: mdl-2018789

ABSTRACT

Estradiol (E2) induces an increase in the peptide elongation rate of isolated uterine ribosomes assayed in a cell-free protein synthesis system. An inhibitory factor, extracted from ribosomes of E2-deprived rats, was found to inhibit the peptide elongation reaction by acting on certain tRNAs to render them incapable of binding to aminoacyl-tRNA synthetases, thus reducing the availability of specific aminoacylated tRNAs required for the sequential translation of the codons in mRNA. The uterine ribosome-associated tRNA inactivator (RATI) has been partially purified and monoclonal antibodies (MABs) to RATI have been prepared. Specificity of the MABs for RATI was indicated by the inactivation of RATI in vitro by the anti-RATI MABs. RATI selectively inactivates deacylated, but not acylated, tRNAs and the inactivation does not appear to involve nuclease cleavage of the tRNA. Within 1 h after E2 treatment 50% of both RATI activity and immunoreactivity were lost from the uterine ribosome extracts, suggesting that E2 regulation of tRNA reutilization may occur through dissociation of RATI from the ribosomal site of tRNA deacylation or alteration in the structure of RATI resulting in inactivation both biologically and immunologically. We propose that RATI may function as an E2-regulatable 'switch' mechanism which inactivates, delays or defers the aminoacylation of certain tRNAs in the absence of E2 and which participates in the regulation of protein synthesis at the translational level by creating rate-limiting levels of certain tRNAs in the E2-deprived uterus.


Subject(s)
Estradiol/pharmacology , RNA, Transfer/antagonists & inhibitors , Ribosomes/chemistry , Uterus/chemistry , Acylation , Amino Acyl-tRNA Synthetases/metabolism , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Female , RNA, Transfer/metabolism , Rats , Ribosomes/metabolism , Uterus/metabolism
16.
J Natl Cancer Inst ; 81(22): 1743-7, 1989 Nov 15.
Article in English | MEDLINE | ID: mdl-2681797

ABSTRACT

The active methylene compounds acetylacetone, 1,1,1-trifluoroacetylacetone, benzoylacetone, dibenzoylmethane, and 1,3-indandione inhibited the mutagenicity of 2-nitrofluorene in Salmonella typhimurium. They also inhibited the N,O-acetyltransferase-catalyzed transfer RNA binding of N-hydroxy-2-acetylaminofluorene, but they did not inhibit N,O-acetyltransferase. However, only 1,3-indandione and 1,1,1-trifluoroacetylacetone significantly inhibited the binding of N-acetoxy-2-acetylaminofluorene to transfer RNA. Reaction of the trifluoro compound with the acetoxy compound yielded 1-(N-2-fluorenylacetamido)acetone. These results demonstrate that active methylene compounds can inhibit mutagenicity and nucleic acid binding of chemical carcinogens.


Subject(s)
Benzoates/pharmacology , Butanones/pharmacology , Chalcones , Fluorenes , Indans/pharmacology , Indenes/pharmacology , Ketones/pharmacology , Mutagens/antagonists & inhibitors , Mutation , Nucleic Acids/metabolism , Pentanones/pharmacology , 2-Acetylaminofluorene/metabolism , Acetyltransferases/antagonists & inhibitors , Animals , Drug Evaluation, Preclinical , Drug Interactions , Hydroxyacetylaminofluorene/metabolism , Male , Mutagens/metabolism , RNA, Transfer/antagonists & inhibitors , RNA, Transfer/metabolism , Rats , Salmonella typhimurium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...