Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Acta Naturae ; 4(4): 65-72, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23346381

ABSTRACT

For prokaryotes in vitro, 16S rRNA and 20 ribosomal proteins are capable of hierarchical self- assembly yielding a 30S ribosomal subunit. The self-assembly is initiated by interactions between 16S rRNA and three key ribosomal proteins: S4, S8, and S7. These proteins also have a regulatory function in the translation of their polycistronic operons recognizing a specific region of mRNA. Therefore, studying the RNA-protein interactions within binary complexes is obligatory for understanding ribosome biogenesis. The non-conventional RNA-protein contact within the binary complex of recombinant ribosomal protein S7 and its 16S rRNA binding site (236 nucleotides) was identified. UV-induced RNA-protein cross-links revealed that S7 cross-links to nucleotide U1321 of 16S rRNA. The careful consideration of the published RNA- protein cross-links for protein S7 within the 30S subunit and their correlation with the X-ray data for the 30S subunit have been performed. The RNA - protein cross-link within the binary complex identified in this study is not the same as the previously found cross-links for a subunit both in a solution, and in acrystal. The structure of the binary RNA-protein complex formed at the initial steps of self-assembly of the small subunit appears to be rearranged during the formation of the final structure of the subunit.

2.
Biochemistry (Mosc) ; 73(6): 652-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18620530

ABSTRACT

In E. coli cells ribosomal small subunit biogenesis is regulated by RNA-protein interactions involving protein S7. S7 initiates the subunit assembly interacting with 16S rRNA. During shift-down of rRNA synthesis level, free S7 inhibits self-translation by interacting with 96 nucleotides long specific region of streptomycin (str) mRNA between cistrons S12 and S7 (intercistron). Many bacteria do not have the extended intercistron challenging development of specific approaches for searching putative mRNA regulatory regions, which are able to interact with proteins. The paper describes application of SERF approach (Selection of Random RNA Fragments) to reveal regulatory regions of str mRNA. Set of random DNA fragments has been generated from str operon by random hydrolysis and then transcribed into RNA; the fragments being able to bind protein S7 (serfamers) have been selected by iterative rounds. S7 binds to single serfamer, 109 nucleotide long (RNA109), derived from the intercistron. After multiple copying and selection, the intercistronic mutant (RNA109) has been isolated; it has enhanced affinity to S7. RNA109 binds to the protein better than authentic intercistronic str mRNA; apparent dissociation constants are 26 +/- 5 and 60 +/- 8 nM, respectively. Location of S7 binding site on the mRNA, as well as putative mode of regulation of coupled translation of S12 and S7 cistrons have been hypothesized.


Subject(s)
Escherichia coli/genetics , Protein Biosynthesis , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , Regulatory Sequences, Nucleic Acid , Ribosomal Proteins/metabolism , Streptomycin/metabolism , Base Sequence , Binding Sites/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Gene Library , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Random Allocation
3.
J Nutr ; 131(11): 2978S-82S, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11694632

ABSTRACT

Trans-translation is a quality-control process, activated upon premature termination of protein elongation, which recycles stalled ribosomes and degrades incomplete polypeptides. These functions are facilitated by transfer-messenger RNA (tmRNA, also called 10Sa RNA or SsrA RNA), a small stable RNA molecule encoded by the SsrA gene found in bacteria, chloroplasts and mitochondria. Most tmRNAs consist of a tRNA- and an mRNA-like domain connected by up to four pseudoknots. Comparative sequence analysis provided the first insight into tmRNA secondary and three-dimensional structure. Studies of the E. coli tmRNA in vitro and in vivo demonstrated that tmRNA functions as a ribonucleoprotein (RNP) complex with elongation factor Tu (EF-Tu), protein SmpB and ribosomal protein S1. The tRNA-like and mRNA-like activities of tmRNA mark prematurely terminated proteins for degradation by attaching to their C-termini peptide tags, which are recognized by numerous proteases. Studies aimed at understanding the details of the molecular mechanisms of trans-translation are ongoing.


Subject(s)
Peptide Elongation Factors/physiology , Protein Biosynthesis , RNA, Bacterial/physiology , Quality Control , Sequence Analysis
4.
J Mol Biol ; 314(1): 9-21, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11724528

ABSTRACT

Transfer-messenger RNA (tmRNA) mimics functions of aminoacyl-tRNA and mRNA, subsequently, when rescuing stalled ribosomes on a 3' truncated mRNA without stop codon in bacteria. In addition, this mechanism marks prematurely terminated proteins by a C-terminal peptide tag as a signal for degradation by specific cellular proteases. For Escherichia coli, previous studies on initial steps of this "trans-translation" mechanism revealed that tmRNA alanylation by Ala-tRNA synthetase and binding of Ala-tmRNA by EF-Tu-GTP for subsequent delivery to stalled ribosomes are inefficient when compared to analogous reactions with canonical tRNA(Ala). In other studies, protein SmpB and ribosomal protein S1 appeared to bind directly to tmRNA and to be indispensable for trans-translation. Here, we have searched for additional and synergistic effects of the latter two on tmRNA alanylation and its subsequent binding to EF-Tu-GTP. Kinetic analysis of functioning combined with band-shift experiments and structural probing demonstrate, that tmRNA may indeed form a multimeric complex with SmpB, S1 and EF-Tu-GTP, which leads to a considerably enhanced efficiency of the initial steps of trans-translation. Whereas S1 binds to the mRNA region of tmRNA, we have found that SmpB and EF-Tu both interact with its acceptor arm region. Interaction with SmpB and EF-Tu was also observed at the acceptor arm of Ala-tRNA(Ala), but there the alanylation efficiency was inhibited rather than stimulated by SmpB. Therefore, SmpB may function as an essential modulator of the tRNA-like acceptor arm of tmRNA during its successive steps in trans-translation.


Subject(s)
Alanine/genetics , Escherichia coli/genetics , Guanosine Triphosphate/metabolism , Peptide Elongation Factor Tu/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA-Binding Proteins/metabolism , Acylation , Alanine/metabolism , Alanine-tRNA Ligase/metabolism , Base Sequence , Binding Sites , Electrophoretic Mobility Shift Assay , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Kinetics , Molecular Sequence Data , Nuclease Protection Assays , Nucleic Acid Conformation , Protein Binding , RNA, Bacterial/genetics , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism , Ribonucleases/metabolism , Ribosomal Proteins/metabolism
5.
Eur J Biochem ; 268(13): 3807-15, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11432749

ABSTRACT

In Streptomyces coelicolor, the regular and abundant elongation factor (EF)-Tu1 is encoded by tuf1, while the actual function of the highly divergent tuf3 gene product is not yet known. Expression of the latter could so far only be detected on the transcriptional level under stress conditions. In this paper we demonstrate the presence of low levels of EF-Tu3 in strains of the J1501 lineage. Enhanced expression was observed for J1501 glkA and relA deletion mutants, which lack glucose kinase and ribosome-bound ppGpp synthetase, respectively. To assess the putative translational capacities of EF-Tu3, a novel Streptomyces in vitro translation assay was designed, based on the full elimination by Ni2+ affinity adsorption of chromosomally encoded (His)6-tagged EF-Tu1 from a S. coelicolor cell-free extract. Translational activity of this system is totally dependent on the addition of purified EF-Tu species or on the presence of an additional elongation factor Tu in the extract, e.g. encoded by a plasmid-borne tuf gene. Using this EF-Tu-dependent translation system, we have established that S. coelicolor EF-Tu3 has translational capacities despite its striking deviation from the common prokaryotic EF-Tu sequence at positions involved in either aminoacyl-tRNA binding or interaction with the guanine-nucleotide exchange factor EF-Ts.


Subject(s)
Genetic Variation , Peptide Elongation Factor Tu/genetics , Protein Biosynthesis , Streptomyces/genetics , Transcription, Genetic , Amino Acid Sequence , Bacterial Proteins/genetics , Base Sequence , Gene Expression Regulation, Bacterial , Genetic Vectors , Kinetics , Models, Molecular , Molecular Sequence Data , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/metabolism , Protein Conformation , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid
6.
J Mol Biol ; 304(5): 995-1005, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11124042

ABSTRACT

The antibiotic GE2270A prevents stable complex formation between elongation factor Tu (EF-Tu) and aminoacyl-tRNA (aatRNA). In Escherichia coli we characterized two mutant EF-Tu species with either G257S or G275A that lead to high GE2270A resistance in poly(Phe) synthesis, which at least partially explains the high resistance of EF-Tu1 from GE2270A producer Planobispora rosea to its own antibiotic. Both E. coli mutants were unexpectedly found to bind GE2270A nearly as well as wild-type (wt) EF-Tu in their GTP-bound conformations. Both G257S and G275A are in or near the binding site for the 3' end of aatRNA. The G257S mutation causes a 2.5-fold increase in affinity for aatRNA, whereas G275A causes a 40-fold decrease. In the presence of GE2270A, wt EF-Tu shows a drop in aatRNA affinity of at least four orders of magnitude. EF-Tu[G275S] and EF-Tu[G275A] curtail this drop to about two or one order, respectively. It thus appears that the resistance mutations do not prevent GE2270A from binding to EF-Tu.GTP and that the mutant EF-Tus may accommodate GE2270A and aatRNA simultaneously. Interestingly, in their GDP-bound conformations the mutant EF-Tus have much less affinity for GE2270A than wt EF-Tu. The latter is explained by a recent crystal structure of the EF-Tu.GDP.GE2270A complex, which predicts direct steric problems between GE2270A and the mutated G257S or G275A. These mutations may cause a dislocation of GE2270A in complex with GTP-bound EF-Tu, which then no longer prevents aatRNA binding as in the wt situation. Altogether, the data lead to the following novel resistance scenario. Upon arrival of the mutant EF-Tu.GTP.GE2270.aatRNA complex at the ribosomal A-site, the GTPase centre is triggered. The affinities of aatRNA and GE2270A for the GDP-bound EF-Tu are negligible; the former stays at the A-site for subsequent interaction with the peptidyltransferase centre and the latter two dissociate from the ribosome.


Subject(s)
Aminoglycosides , Escherichia coli , Guanosine Triphosphate/metabolism , Mutation/genetics , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Peptides, Cyclic/pharmacology , RNA, Transfer, Amino Acyl/metabolism , Thiazoles/metabolism , Actinomycetales/chemistry , Adenine/metabolism , Amino Acid Substitution/genetics , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Resistance, Microbial , Escherichia coli/chemistry , Escherichia coli/drug effects , Escherichia coli/genetics , Guanosine Diphosphate/metabolism , Models, Molecular , Peptide Elongation Factor Tu/chemistry , Peptides/metabolism , Peptides, Cyclic/chemistry , Poly U/genetics , Poly U/metabolism , Protein Binding/drug effects , Protein Biosynthesis/drug effects , Protein Conformation , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Transfer, Amino Acyl/genetics , Thermodynamics , Thermus/chemistry , Thiazoles/chemistry , Thiazoles/pharmacology
7.
J Bacteriol ; 182(20): 5653-62, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11004161

ABSTRACT

The role of ssgA in cell division and development of streptomycetes was analyzed. An ssgA null mutant of Streptomyces coelicolor produced aerial hyphae but failed to sporulate, and ssgA can therefore be regarded as a novel whi gene. In addition to the morphological changes, antibiotic production was also disturbed, with strongly reduced actinorhodin production. These defects could be complemented by plasmid-borne ssgA. In the wild-type strain, transcription of ssgA was induced by nutritional shift-down and was shown to be linked to that of the upstream-located gene ssgR, which belongs to the family of iclR-type transcriptional regulator genes. Analysis of mycelium harvested from liquid-grown cultures by transmission electron microscopy showed that septum formation had strongly increased in ssgA-overexpressing strains in comparison to wild-type S. coelicolor and that spore-like compartments were produced at high frequency. Furthermore, the hyphae were significantly wider and contained irregular and often extremely thick septa. These data underline the important role for ssgA in Streptomyces cell division.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Streptomyces/physiology , Amino Acid Sequence , Bacterial Proteins/chemistry , Base Sequence , Cell Division , Gene Deletion , Genetic Complementation Test , Molecular Sequence Data , Mutagenesis , Plasmids , Polymerase Chain Reaction , Restriction Mapping , Sequence Alignment , Sequence Homology, Amino Acid , Spores, Bacterial/physiology , Spores, Bacterial/ultrastructure , Streptomyces/cytology , Streptomyces/genetics
8.
Biochemistry ; 39(10): 2652-8, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10704215

ABSTRACT

Aminoacylation and transportation of tmRNA to stalled ribosomes constitute prerequisite steps for trans-translation, a process facilitating the release of stalled ribosomes from 3' ends of truncated mRNAs and the degradation of incompletely synthesized proteins. Kinetic analysis of the aminoacylation of tmRNA indicates that tmRNA has both a lower affinity and a lower turnover number than cognate tRNA(Ala) for alanyl-tRNA synthetase, resulting in a 75-fold lower k(cat)/K(M) value. The association rate constant of Ala-tmRNA for elongation factor Tu in complex with GTP is about 150-fold lower than that of Ala-tRNA(Ala), whereas its dissocation rate constant is about 5-fold lower. These observations can be interpreted to suggest that additional factors facilitate tmRNA binding to ribosomes.


Subject(s)
Alanine-tRNA Ligase/metabolism , Escherichia coli/metabolism , Peptide Elongation Factor Tu/metabolism , RNA, Messenger/metabolism , RNA, Transfer, Ala/metabolism , Acylation , Alanine-tRNA Ligase/genetics , Biopolymers/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Peptide Elongation Factor Tu/genetics , RNA, Transfer, Ala/genetics , Thermodynamics , Transcription, Genetic
9.
J Biochem Biophys Methods ; 42(1-2): 1-14, 2000 Jan 03.
Article in English | MEDLINE | ID: mdl-10647810

ABSTRACT

Elongation factor Tu is essential for binding and a correct delivery of aminoacyl-tRNA during protein biosynthesis. For a good characterization of its interaction with tRNA in terms of structure-function relationship, determinations of kinetic equilibrium parameters are of great value. We describe two novel methods for that purpose. One method is based on EF-Tu protection of the tRNA 3' acceptor end against RNase A cleavage and yields the Kd value together with the corresponding dissociation and association rate constants from one single set of experiments. The other is a rapid method for screening relative affinities of mutant EF-Tus for tRNA. It is based on competition between EF-Tu species with and without a (His)6 extension for the same aminoacyl-tRNA and yields a relative Kd value. The method can be of general importance for the measuring of ligand affinities of all sorts of His-tagged proteins. Both methods are illustrated by their application in the analysis of mutant EF-Tus with changed interactions with tRNA and antibiotics. Raising the assay temperature from 4 to 37 degrees C causes a 30-fold increase of Kd for EF-Tu x GTP x Phe-tRNA complexes. The mutation K237E leads to rapid inactivation at the latter temperature. A parallel is found between the order of increasing Kd values for EF-Tus with mutation G316D, A375T and Q124K, respectively, and their order of increasing resistance to kirromycin.


Subject(s)
Biochemistry/methods , Histidine , Mutation , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , RNA, Transfer, Amino Acyl/metabolism , Binding, Competitive , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Models, Chemical , Peptide Elongation Factor Tu/analysis , Peptides/genetics , Peptides/metabolism , RNA, Transfer, Phe/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reproducibility of Results , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/metabolism
10.
Antonie Van Leeuwenhoek ; 78(3-4): 269-76, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11386349

ABSTRACT

This paper describes the effects of increased expression of the cell division genes ftsZ, ftsQ, and ssgA on the development of both solid- and liquid-grown mycelium of Streptomyces coelicolor and Streptomyces lividans. Over-expression of ftsZ in S. coelicolor M145 inhibited aerial mycelium formation and blocked sporulation. Such deficient sporulation was also observed for the ftsZ mutant. Over-expression of ftsZ also inhibited morphological differentiation in S. lividans 1326, although aerial mycelium formation was less reduced. Furthermore, antibiotic production was increased in both strains, and in particular the otherwise dormant actinorhodin biosynthesis cluster of S. lividans was activated in liquid- and solid-grown cultures. No significant alterations were observed when the gene dosage of ftsQ was increased. Analysis by transmission electron microscopy of an S. coelicolor strain overexpressing ssgA showed that septum formation had strongly increased in comparison to wild-type S. coelicolor, showing that SsgA clearly influences Streptomyces cell division. The morphology of the hyphae was affected such that irregular septa were produced with a significantly wider diameter, thereby forming spore-like compartments. This suggests that ssgA can induce a process similar to submerged sporulation in Streptomyces strains that otherwise fail to do so. A working model is proposed for the regulation of septum formation and of submerged sporulation.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Cytoskeletal Proteins , Gene Expression Regulation, Bacterial , Streptomyces/cytology , Streptomyces/genetics , Bacterial Proteins/genetics , Cell Division/genetics , Gene Dosage , Plasmids , Restriction Mapping , Species Specificity , Spores, Bacterial , Streptomyces/physiology
11.
Folia Microbiol (Praha) ; 44(2): 131-41, 1999.
Article in English | MEDLINE | ID: mdl-10588048

ABSTRACT

EF-Tu from E. coli, one of the superfamily of GTPase switch proteins, plays a central role in the fast and accurate delivery of aminoacyl-tRNAs to the translating ribosome. An overview is given about the regulatory effects of methylation, phosphorylation and phage-induced cleavage of EF-Tu on its function. During exponential growth, EF-Tu becomes monomethylated at Lys56 which is converted to Me2Lys upon entering the stationary phase. Lys56 is in the GTPase switch-1 region (residues 49-62), a strongly conserved site involved in interactions with the nucleotide and the 5' end of tRNA. Methylation was found to attenuate GTP hydrolysis and may thus enhance translational accuracy. In vivo 5-10% of EF-Tu is phosphorylated at Thr382 by a ribosome-associated kinase. In EF-Tu-GTP, Thr382 in domain 3 has a strategic position in the interface with domain 1; it is hydrogen-bonded to Glu117 that takes part in the switch-2 mechanism, and is close to the T-stem binding site of the tRNA, in a region known for many kirromycin-resistance mutations. Phosphorylation is enhanced by EF-Ts, but inhibited by kirromycin. In reverse, phosphorylated EF-Tu has an increased affinity for EF-Ts, does not bind kirromycin and can no longer bind aminoacyi tRNA. The in vivo role of this reversible modification is still a matter of speculation. T4 infection of E. coli may trigger a phase-exclusion mechanism by activation of Lit, a host-encoded proteinase. As a result, EF-Tu is cleaved site-specifically between Gly59-Ile60 in the switch-1 region. Translation was found to drop beyond a minimum level. Interestingly, the identical sequence in the related EF-G appeared to remain fully intact. Although the Lit cleavage-mechanism may eventually lead to programmed cell death, the very efficient prevention of phage multiplication may be caused by a novel mechanism of in cis inhibition of late T4 mRNA translation.


Subject(s)
Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Peptide Elongation Factor Tu/metabolism , Protein Biosynthesis , Methylation , Phosphorylation
12.
J Mol Biol ; 294(3): 627-37, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10610785

ABSTRACT

For clarification of the action of a new antibiotic, the analysis of resistant mutants is often indispensable. For enacyloxin IIa we discovered four resistant elongation factor Tu (EF-Tu) species in Escherichia coli with the mutations Q124K, G316D, Q329H, and A375T, respectively. They revealed that enacyloxin IIa sensitivity is dominant in a mixed population of resistant and wild-type EF-Tus. This points to an inhibition mechanism in which EF-Tu is the dominant target of enacyloxin IIa and in which a ribosome with a sensitive EF-Tu blocks mRNA translation for upstream ribosomes with resistant EF-Tus, a mechanism similar to that of the unrelated antibiotic kirromycin. Remarkably, the same mutations are also linked to kirromycin resistance, though the order of their levels of resistance is different from that for enacyloxin IIa. Among the mutant EF-Tus, three different resistance mechanisms can be distinguished: (i) by obstructing enacyloxin IIa binding to EF-Tu. GTP; (ii) by enabling the release of enacyloxin IIa after GTP hydrolysis; and (iii) by reducing the affinity of EF-Tu.GDP. enacyloxin IIa for aminoacyl-tRNA at the ribosomal A-site, which then allows the release of EF-Tu.GDP.enacyloxin IIa. Ala375 seems to contribute directly to enacyloxin IIa binding at the domain 1-3 interface of EF-Tu.GTP, a location that would easily explain the pleiotropic effects of enacyloxin IIa on the functioning of EF-Tu.


Subject(s)
Peptide Elongation Factor Tu/genetics , Ribosomes/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , GTP Phosphohydrolases/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Kinetics , Models, Molecular , Peptide Elongation Factor Tu/metabolism , Phenylalanine/biosynthesis , Polyenes/metabolism , Polymers/metabolism , Protein Biosynthesis , Pyridones/metabolism , RNA, Transfer, Amino Acyl/metabolism , Structure-Activity Relationship
13.
Mol Gen Genet ; 260(6): 603-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9928940

ABSTRACT

A method of lambda-mediated gene replacement was used to disrupt tufA or tufB on the chromosome of the E. coli K-12 strain MG1655. Both tuf genes, which are almost identical but map in different chromosomal contexts, encode the essential peptide chain elongation factor EF-Tu, one of the most abundant cytoplasmic proteins. Southern analysis confirmed replacement of the chromosomal tufA or tufB gene by a chloramphenicol resistance marker, demonstrating that both tuf genes are individually dispensable for growth. Under conditions of rapid growth, deletion of tufB had no significant effect on growth rate, but deletion of tufA resulted in a 35% increase in generation time. In minimal medium we observed no negative effects of tufA deletion on growth rate. Strains with a single tuf gene are useful for the expression of mutant forms of EF-Tu as the sole species in cells; this was demonstrated by introducing the hybrid tufAhis gene, encoding EF-TuA extended with a C-terminal (His)6 tag, into the chromosome of a strain lacking tufB.


Subject(s)
Bacterial Proteins/genetics , Escherichia coli/growth & development , Escherichia coli/genetics , Gene Deletion , Peptide Elongation Factor Tu/genetics , Bacteriophage lambda/genetics , Chromosomes, Bacterial , Plasmids , Transduction, Genetic
14.
J Mol Biol ; 283(2): 371-81, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9769211

ABSTRACT

The tuf gene coding for elongation factor Tu (EF-Tu) of Bacillus stearothermophilus was cloned and sequenced. This gene maps in the same context as the tufA gene of Escherichia coli str operon. Northern-blot analysis and primer extension experiments revealed that the transcription of the tuf gene is driven from two promoter regions. One of these is responsible for producing a 4.9-kb transcript containing all the genes of B. stearothermophilus str operon and the other, identified adjacent to the stop codon of the fus gene and designated tufp, for producing a 1.3-kb transcript of the tuf gene only. In contrast to the situation in E. coli, the ratio between the transcription products was found to be about 10:1 in favour of the tuf gene transcript. This high transcription activity from the tufp promoter might be accounted for by the presence of an extremely A+T-rich block consisting of 29 nucleotides which immediately precedes the consensus -35 region of the promoter. A very similar tuf gene transcription strategy and the same tufp promoter organization with the identical A/T block were found in Bacillus subtilis. The tuf gene specifies a protein of 395 amino acid residues with a molecular mass of 43,290 Da, including the N-terminal methionine. A computer-generated three-dimensional homology model shows that all the structural elements essential for binding guanine nucleotides and aminoacyl-tRNA are conserved. The presence of serine at position 376 and a low affinity for kirromycin determined by zone-interference gel electrophoresis (Kd approximately 8 microM) and by polyacrylamide gel electrophoresis under non-denaturing conditions are in agreement with the reported resistance of this EF-Tu to the antibiotic. The replacement of the highly conserved Leu211 by Met was identified as a possible cause of pulvomycin resistance.


Subject(s)
Aminoglycosides , Geobacillus stearothermophilus/genetics , Peptide Elongation Factor Tu/genetics , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Base Sequence , DNA, Bacterial , Drug Resistance, Microbial , Geobacillus stearothermophilus/drug effects , Geobacillus stearothermophilus/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Elongation Factor Tu/biosynthesis , Peptide Elongation Factor Tu/chemistry , Protein Conformation , Pyridones/pharmacology , Sequence Alignment
15.
Proc Natl Acad Sci U S A ; 95(6): 2891-5, 1998 Mar 17.
Article in English | MEDLINE | ID: mdl-9501186

ABSTRACT

Phage exclusion is a form of programmed cell death in prokaryotes in which death is triggered by infection with phage, a seemingly altruistic response that limits multiplication of the phage and its spread through the population. One of the best-characterized examples of phage exclusion is the exclusion of T-even phages such as T4 by the e14-encoded Lit protein in many Escherichia coli K-12 strains. In this exclusion system, transcription and translation of a short region of the major head coat protein gene late in phage infection activates proteolysis of translation elongation factor Tu (EF-Tu), blocking translation and multiplication of the phage. The cleavage occurs between Gly-59 and Ile-60 in the nucleotide-binding domain. In the present work, we show that a 29-residue synthetic peptide spanning the activating region of the major head coat protein can activate the cleavage of GDP-bound EF-Tu in a purified system containing only purified EF-Tu and purified Lit protein. Lit behaves as a bona fide enzyme in this system, cleaving EF-Tu to completion when present at substoichiometric amounts. Two mutant peptides with amino acid changes that reduce the activation of cleavage of EF-Tu in vivo were also greatly reduced in their ability to activate EF-Tu cleavage in vitro but were still able to activate cleavage at a high concentration. Elongation factor G, which has the same sequence at the cleavage site and a nucleotide-binding domain similar to EF-Tu, was not cleaved by this system, and neither was heat-inactivated EF-Tu, suggesting that the structural context of the cleavage site may be important for specificity. This system apparently represents an activation mechanism for proteolysis that targets one of nature's most evolutionarily conserved proteins for site-specific cleavage.


Subject(s)
Bacterial Proteins/metabolism , Capsid Proteins , Capsid/metabolism , Endopeptidases/metabolism , Escherichia coli Proteins , Membrane Proteins/metabolism , Peptide Elongation Factor Tu/metabolism , Amino Acid Sequence , Bacteriophage T4/growth & development , Enzyme Activation , Escherichia coli/virology , Molecular Sequence Data , Peptide Elongation Factor G , Peptide Elongation Factors/metabolism , Peptide Fragments/metabolism , Substrate Specificity
16.
J Bacteriol ; 179(11): 3619-24, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9171408

ABSTRACT

The str operon of Streptomyces ramocissimus contains the genes for ribosomal proteins S12 (rpsL) and S7 (rpsG) and for the polypeptide chain elongation factors G (EF-G) (fus) and Tu (EF-Tu) (tuf). This kirromycin producer contains three tuf or tuf-like genes; tuf1 encodes the regular EF-Tu and is located immediately downstream of fus. In vivo and in vitro transcription analysis revealed a transcription start site directly upstream of S. ramocissimus tuf1, in addition to the operon promoter rpsLp. Transcription from these promoters appeared to be growth phase dependent, diminishing drastically upon entry into stationary phase and at the onset of production of the EF-Tu-targeted antibiotic kirromycin. In surface-grown cultures, a second round of tuf1 transcription, coinciding with aerial mycelium formation and kirromycin production, was observed. The tuf1-specific promoter (tuf1p) was located in the intercistronic region between fus and tuf1 by high-resolution S1 mapping, in vitro transcription, and in vivo promoter probing. During logarithmic growth, the tuf1p and rpsLp transcripts are present at comparable levels. In contrast to Escherichia coli, which has two almost identical tuf genes, the gram-positive S. ramocissimus contains only tuf1 for its regular EF-Tu. High levels of EF-Tu may therefore be achieved by the compensatory activity of tuf1p.


Subject(s)
Gene Expression Regulation, Bacterial , Genes, Bacterial , Peptide Elongation Factor Tu/genetics , Promoter Regions, Genetic/genetics , Streptomyces/genetics , Bacterial Proteins/genetics , Base Sequence , Escherichia coli Proteins , Molecular Sequence Data , Ribosomal Protein S9 , Streptomyces/growth & development , Transcription, Genetic
17.
Biochem Biophys Res Commun ; 230(2): 320-6, 1997 Jan 13.
Article in English | MEDLINE | ID: mdl-9016775

ABSTRACT

Sensitivity of EF-Tu1 of the GE2270A producer Planobispora rosea towards GE2270A, pulvomycin and kirromycin was determined by band-shift assays for EF-Tu1-antibiotic complex formation and by in vitro translation experiments. EF-Tu1 of P. rosea appeared to be not only totally resistant to GE2270A, but also ten times more resistant to kirromycin than EF-Tu1 of Streptomyces coelicolor. In contrast, P. rosea EF-Tu1 was found to be not resistant to pulvomycin, an antibiotic that just like GE2270A blocks EF-Tu x GTP x aminoacyl-tRNA complex formation. Previous in vivo and in vitro experiments with mixed populations of antibiotic resistant and sensitive EF-Tu species had shown that sensitivity to kirromycin and pulvomycin is dominant over resistance. In the case of GE2270A we observed, however, that sensitivity is recessive to resistance, which again points to a different action mechanism than in the case of pulvomycin. Besides the tuf1 gene encoding the regular elongation factor EF-Tu1 a gene similar to S. coelicolor tuf3 for a specialized EF-Tu was located in the P. rosea genome. The tuf1 gene was isolated and sequenced. The amino acid sequence of EF-Tul of P. rosea not only exhibits an unusual Tyr160 substitution (comparable to those described for kirromycin-resistant EF-Tus), but also shows significant changes of conserved amino acids in domain 2 that may be responsible for GE2270A resistance (the latter do not resemble those leading to pulvomycin resistance). P. rosea EF-Tu1 thus is a first example of a bacterial EF-Tu with resistance against two divergently acting antibiotics.


Subject(s)
Actinomycetales/metabolism , Aminoglycosides , Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Peptide Elongation Factor Tu/biosynthesis , Peptides , Actinomycetales/drug effects , Actinomycetales/genetics , Amino Acid Sequence , Cell-Free System , Cloning, Molecular , Drug Resistance, Microbial , Genes, Bacterial , Kinetics , Molecular Sequence Data , Peptide Biosynthesis , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/isolation & purification , Peptides, Cyclic/pharmacology , Protein Biosynthesis , Protein Structure, Secondary , Pyridones/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid , Streptomyces/metabolism , Thiazoles/pharmacology
18.
EMBO J ; 15(23): 6766-74, 1996 Dec 02.
Article in English | MEDLINE | ID: mdl-8978702

ABSTRACT

Elongation factor Tu (EF-Tu) from Escherichia coli carrying the mutation G222D is unable to hydrolyze GTP on the ribosome and to sustain polypeptide synthesis at near physiological Mg2+ concentration, although the interactions with guanine nucleotides and aminoacyl-tRNA are not changed significantly. GTPase and polypeptide synthesis activities are restored by increasing the Mg2+ concentration. Here we report a pre-steady-state kinetic study of the binding of the ternary complexes of wild-type and mutant EF-Tu with Phe-tRNA(Phe) and GTP to the A site of poly(U)-programed ribosomes. The kinetic parameters of initial binding to the ribosome and subsequent codon-anticodon interaction are similar for mutant and wild-type EF-Tu, independent of the Mg2+ concentration, suggesting that the initial interaction with the ribosome is not affected by the mutation. Codon recognition following initial binding is also not affected by the mutation. The main effect of the G222D mutation is the inhibition, at low Mg2+ concentration, of codon-induced structural transitions of the tRNA and, in particular, their transmission to EF-Tu that precedes GTP hydrolysis and the subsequent steps of A-site binding. Increasing the Mg2+ concentration to 10 mM restores the complete reaction sequence of A-site binding at close to wild-type rates. The inhibition of the structural transitions is probably due to the interference of the negative charge introduced by the mutation with negative charges either of the 3' terminus of the tRNA, bound in the vicinity of the mutated amino acid in domain 2 of EF-Tu, or of the ribosome. Increasing the Mg2+ concentration appears to overcome the inhibition by screening the negative charges.


Subject(s)
Escherichia coli/metabolism , GTP Phosphohydrolase-Linked Elongation Factors/metabolism , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/metabolism , Point Mutation , Protein Conformation , Protein Structure, Secondary , Ribosomes/metabolism , Thermus/metabolism , Codon , Enzyme Activation , Guanosine Triphosphate/metabolism , Guanylyl Imidodiphosphate/metabolism , Kinetics , Magnesium/metabolism , Models, Structural , Models, Theoretical , Mutagenesis, Site-Directed , Peptide Elongation Factor Tu/biosynthesis , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Phe/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
19.
Biochem Biophys Res Commun ; 225(3): 961-7, 1996 Aug 23.
Article in English | MEDLINE | ID: mdl-8780718

ABSTRACT

We have developed a cell-free system of E. coli that enables us to study the in vitro translation of natural mRNA mediated by wild-type or mutant EF-Tu. Various mutant EF-Tu species have been analyzed, one of which, EF-Tu [G222D], appeared to be virtually unable to mediate the translation of natural mRNA. Since this mutant factor is able to participate in translation in vivo by suppressing nonsense and frameshift mutations in cooperation with EF-Tu [A375T], a revision of the generally accepted scheme of the elongation cycle has been proposed (Bosch, L., Vijgenboom, E., & Zeef, L.A.H., 1996, Biochemistry 36).


Subject(s)
Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Peptide Chain Elongation, Translational , Point Mutation , Protein Biosynthesis , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer, Phe/genetics , RNA, Transfer, Phe/metabolism
20.
Gene ; 165(1): 39-43, 1995 Nov 07.
Article in English | MEDLINE | ID: mdl-7489913

ABSTRACT

This study has investigated the cause of a growth-defect phenotype of a mutation in the elongation factor EF-Tu from Escherichia coli. An M13-based genetic retrieval system reported by Zeef and Bosch [Mol. Gen. Genet. 238 (1993) 252-260] was used to segregate and identify an extremely growth-defective kirromycin-resistant (KrR) tufA mutation, encoding Gln124-->Lys (Q124K), from a KrR parent strain. This original strain also contained mutations, 124com1 and 124com2, that appear to have evolved to suppress the Q124K tufA mutation. In this communication we present these M13-based genetic experiments together with additional genetic and protein characterization experiments to clarify the basis of this complementation. The data indicate that the serious growth defect of Q124K originates from a defective GTP/GDP interaction. The GTP/GDP binding and GTP hydrolysis characteristics of ET-Tu Q124K were different from wild-type EF-Tu and especially of another KrR EF-Tu mutant A375T. In line with this, 124com1 specifically complemented EF-Tu Q124K, whereas the growth defects of strains containing EF-Tu mutated at aa 375 were aggravated. We also show that strains containing the segregated tufA Q124K mutation formed filaments.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Peptide Elongation Factor Tu/genetics , Suppression, Genetic , Drug Resistance, Microbial/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , GTP Phosphohydrolase-Linked Elongation Factors/metabolism , Guanosine Diphosphate/metabolism , Peptide Elongation Factor Tu/metabolism , Pyridones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...