Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
RNA ; 30(3): 298-307, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38164606

ABSTRACT

Several methods are available to visualize and assess the kinetics and efficiency of elemental steps of protein biosynthesis. However, each of these methods has its own limitations. Here, we present a novel, simple and convenient tool for monitoring stepwise in vitro translation initiated by BODIPY-Met-tRNA. Synthesis and release of very short, 1-7 amino acids, BODIPY-labeled peptides, can be monitored using urea-polyacrylamide gel electrophoresis. Very short BODIPY-labeled oligopeptides might be resolved this way, in contrast to widely used Tris-tricine gel electrophoresis, which is suitable to separate peptides larger than 1 kDa. The method described in this manuscript allows one to monitor the steps of translation initiation, peptide transfer, translocation, and termination as well as their inhibition at an unprecedented single amino acid resolution.


Subject(s)
Boron Compounds , Peptides , RNA, Transfer, Amino Acyl , RNA, Transfer, Amino Acyl/chemistry , Peptides/metabolism , RNA, Transfer/metabolism , Electrophoresis, Polyacrylamide Gel , Protein Biosynthesis
2.
PLoS Biol ; 18(1): e3000593, 2020 01.
Article in English | MEDLINE | ID: mdl-31995552

ABSTRACT

During host colonization, bacteria use the alarmones (p)ppGpp to reshape their proteome by acting pleiotropically on DNA, RNA, and protein synthesis. Here, we elucidate how the initiating ribosome senses the cellular pool of guanosine nucleotides and regulates the progression towards protein synthesis. Our results show that the affinity of guanosine triphosphate (GTP) and the inhibitory concentration of ppGpp for the 30S-bound initiation factor IF2 vary depending on the programmed mRNA. The TufA mRNA enhanced GTP affinity for 30S complexes, resulting in improved ppGpp tolerance and allowing efficient protein synthesis. Conversely, the InfA mRNA allowed ppGpp to compete with GTP for IF2, thus stalling 30S complexes. Structural modeling and biochemical analysis of the TufA mRNA unveiled a structured enhancer of translation initiation (SETI) composed of two consecutive hairpins proximal to the translation initiation region (TIR) that largely account for ppGpp tolerance under physiological concentrations of guanosine nucleotides. Furthermore, our results show that the mechanism enhancing ppGpp tolerance is not restricted to the TufA mRNA, as similar ppGpp tolerance was found for the SETI-containing Rnr mRNA. Finally, we show that IF2 can use pppGpp to promote the formation of 30S initiation complexes (ICs), albeit requiring higher factor concentration and resulting in slower transitions to translation elongation. Altogether, our data unveil a novel regulatory mechanism at the onset of protein synthesis that tolerates physiological concentrations of ppGpp and that bacteria can exploit to modulate their proteome as a function of the nutritional shift happening during stringent response and infection.


Subject(s)
Guanosine Tetraphosphate/pharmacology , Peptide Chain Initiation, Translational , RNA, Messenger/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Binding, Competitive , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Guanosine Tetraphosphate/metabolism , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Host-Pathogen Interactions/physiology , Kinetics , Nucleic Acid Conformation , Peptide Chain Initiation, Translational/drug effects , Peptide Chain Initiation, Translational/physiology , Peptide Elongation Factor Tu/metabolism , Protein Biosynthesis/drug effects , RNA, Messenger/chemistry , RNA, Messenger/drug effects , RNA, Messenger/genetics
3.
RNA ; 26(6): 715-723, 2020 06.
Article in English | MEDLINE | ID: mdl-32144191

ABSTRACT

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


Subject(s)
Anti-Bacterial Agents/chemistry , Erythromycin/analogs & derivatives , Protein Synthesis Inhibitors/chemistry , Ribosomes/chemistry , Anti-Bacterial Agents/pharmacology , Cryoelectron Microscopy , Erythromycin/chemistry , Erythromycin/pharmacology , Escherichia coli/genetics , Models, Molecular , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Ribosomal, 23S/chemistry
4.
Int J Mol Sci ; 22(17)2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34502523

ABSTRACT

The growth of the polypeptide chain occurs due to the fast and coordinated work of the ribosome and protein elongation factors, EF-Tu and EF-G. However, the exact contribution of each of these components in the overall balance of translation kinetics remains not fully understood. We created an in vitro translation system Escherichia coli replacing either elongation factor with heterologous thermophilic protein from Thermus thermophilus. The rates of the A-site binding and decoding reactions decreased an order of magnitude in the presence of thermophilic EF-Tu, indicating that the kinetics of aminoacyl-tRNA delivery depends on the properties of the elongation factor. On the contrary, thermophilic EF-G demonstrated the same translocation kinetics as a mesophilic protein. Effects of translocation inhibitors (spectinomycin, hygromycin B, viomycin and streptomycin) were also similar for both proteins. Thus, the process of translocation largely relies on the interaction of tRNAs and the ribosome and can be efficiently catalysed by thermophilic EF-G even at suboptimal temperatures.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli/metabolism , Peptide Chain Elongation, Translational , Peptide Elongation Factor Tu/metabolism , Ribosomes/metabolism , Thermus thermophilus , Peptide Elongation Factor G/metabolism , RNA, Bacterial/metabolism , RNA, Transfer/metabolism
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.
Proc Natl Acad Sci U S A ; 112(19): 6033-7, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25902496

ABSTRACT

The reduction of specific uridines to dihydrouridine is one of the most common modifications in tRNA. Increased levels of the dihydrouridine modification are associated with cancer. Dihydrouridine synthases (Dus) from different subfamilies selectively reduce distinct uridines, located at spatially unique positions of folded tRNA, into dihydrouridine. Because the catalytic center of all Dus enzymes is conserved, it is unclear how the same protein fold can be reprogrammed to ensure that nucleotides exposed at spatially distinct faces of tRNA can be accommodated in the same active site. We show that the Escherichia coli DusC is specific toward U16 of tRNA. Unexpectedly, crystal structures of DusC complexes with tRNA(Phe) and tRNA(Trp) show that Dus subfamilies that selectively modify U16 or U20 in tRNA adopt identical folds but bind their respective tRNA substrates in an almost reverse orientation that differs by a 160° rotation. The tRNA docking orientation appears to be guided by subfamily-specific clusters of amino acids ("binding signatures") together with differences in the shape of the positively charged tRNA-binding surfaces. tRNA orientations are further constrained by positional differences between the C-terminal "recognition" domains. The exquisite substrate specificity of Dus enzymes is therefore controlled by a relatively simple mechanism involving major reorientation of the whole tRNA molecule. Such reprogramming of the enzymatic specificity appears to be a unique evolutionary solution for altering tRNA recognition by the same protein fold.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Oxidoreductases/chemistry , RNA, Transfer/chemistry , Amino Acids/chemistry , Catalytic Domain , Crystallography, X-Ray , Evolution, Molecular , Protein Binding , Protein Folding , RNA/chemistry , RNA-Binding Proteins/chemistry , Substrate Specificity , Uridine/chemistry , X-Ray Diffraction
7.
Front Microbiol ; 12: 618857, 2021.
Article in English | MEDLINE | ID: mdl-33643246

ABSTRACT

Amicoumacin A (Ami) halts bacterial growth by inhibiting the ribosome during translation. The Ami binding site locates in the vicinity of the E-site codon of mRNA. However, Ami does not clash with mRNA, rather stabilizes it, which is relatively unusual and implies a unique way of translation inhibition. In this work, we performed a kinetic and thermodynamic investigation of Ami influence on the main steps of polypeptide synthesis. We show that Ami reduces the rate of the functional canonical 70S initiation complex (IC) formation by 30-fold. Additionally, our results indicate that Ami promotes the formation of erroneous 30S ICs; however, IF3 prevents them from progressing towards translation initiation. During early elongation steps, Ami does not compromise EF-Tu-dependent A-site binding or peptide bond formation. On the other hand, Ami reduces the rate of peptidyl-tRNA movement from the A to the P site and significantly decreases the amount of the ribosomes capable of polypeptide synthesis. Our data indicate that Ami progressively decreases the activity of translating ribosomes that may appear to be the main inhibitory mechanism of Ami. Indeed, the use of EF-G mutants that confer resistance to Ami (G542V, G581A, or ins544V) leads to a complete restoration of the ribosome functionality. It is possible that the changes in translocation induced by EF-G mutants compensate for the activity loss caused by Ami.

8.
J Mol Biol ; 430(6): 842-852, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29410130

ABSTRACT

Antibiotic chloramphenicol (CHL) binds with a moderate affinity at the peptidyl transferase center of the bacterial ribosome and inhibits peptide bond formation. As an approach for modifying and potentially improving properties of this inhibitor, we explored ribosome binding and inhibitory activity of a number of amino acid analogs of CHL. The L-histidyl analog binds to the ribosome with the affinity exceeding that of CHL by 10 fold. Several of the newly synthesized analogs were able to inhibit protein synthesis and exhibited the mode of action that was distinct from the action of CHL. However, the inhibitory properties of the semi-synthetic CHL analogs did not correlate with their affinity and in general, the amino acid analogs of CHL were less active inhibitors of translation in comparison with the original antibiotic. The X-ray crystal structures of the Thermus thermophilus 70S ribosome in complex with three semi-synthetic analogs showed that CHL derivatives bind at the peptidyl transferase center, where the aminoacyl moiety of the tested compounds established idiosyncratic interactions with rRNA. Although still fairly inefficient inhibitors of translation, the synthesized compounds represent promising chemical scaffolds that target the peptidyl transferase center of the ribosome and potentially are suitable for further exploration.


Subject(s)
Amino Acids/pharmacology , Anti-Bacterial Agents/pharmacology , Chloramphenicol/pharmacology , Protein Binding/drug effects , Ribosome Subunits, Large, Bacterial/drug effects , Binding Sites , Chloramphenicol/metabolism , Crystallography, X-Ray , Escherichia coli/metabolism , Models, Molecular , Peptidyl Transferases/metabolism , Protein Biosynthesis/drug effects , Protein Conformation , Ribosome Subunits, Large, Bacterial/metabolism , Thermus thermophilus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL