Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Adv Protein Chem Struct Biol ; 138: 67-99, 2024.
Article in English | MEDLINE | ID: mdl-38220433

ABSTRACT

Mammalian translation elongation factors eEF1A1 and eEF1A2 are 92% homologous isoforms whose mutually exclusive tissue-specific expression is regulated during development. The isoforms have similar translation functionality, but show differences in spatial organization and participation in various processes, such as oncogenesis and virus reproduction. The differences may be due to their ability to interact with isoform-specific partner proteins. We used the identified sets of eEF1A1 or eEF1A2 partner proteins to identify cell complexes and/or processes specific to one particular isoform. As a result, we found isoform-specific interactions reflecting the involvement of different eEF1A isoforms in different cellular processes, including actin-related, chromatin-remodeling, ribonuclease H2, adenylyl cyclase, and Cul3-RING ubiquitin ligase complexes as well as initiation of mitochondrial transcription. An essential by-product of our analysis is the elucidation of a number of cellular processes beyond protein biosynthesis, where both isoforms appear to participate such as large ribosomal subunit biogenesis, mRNA splicing, DNA mismatch repair, 26S proteasome activity, P-body and exosomes formation, protein targeting to the membrane. This information suggests that a relatively high content of eEF1A in the cell may be necessary not only to maintain efficient translation, but also to ensure its participation in various cellular processes, where some roles of eEF1A have not yet been described. We believe that the data presented here will be useful for deciphering new auxiliary functions of eEF1A and its isoforms, and provide a new look at the known non-canonical functions of this main component of the human translation-elongation machinery.


Subject(s)
Protein Biosynthesis , Proteomics , Animals , Humans , Mammals , Protein Isoforms/genetics
2.
Nucleic Acids Res ; 46(16): 8651-8661, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30107527

ABSTRACT

The GTPase elongation factor EF-Tu delivers aminoacyl-tRNAs to the mRNA-programmed ribosome during translation. Cognate codon-anticodon interaction stimulates GTP hydrolysis within EF-Tu. It has been proposed that EF-Tu undergoes a large conformational change subsequent to GTP hydrolysis, which results in the accommodation of aminoacyl-tRNA into the ribosomal A-site. However, this proposal has never been tested directly. Here, we apply single-molecule total internal reflection fluorescence microscopy to study the conformational dynamics of EF-Tu when bound to the ribosome. Our studies show that GTP hydrolysis initiates a partial, comparatively small conformational change of EF-Tu on the ribosome, not directly along the path from the solution 'GTP' to the 'GDP' structure. The final motion is completed either concomitant with or following dissociation of EF-Tu from the ribosome. The structural transition of EF-Tu on the ribosome is slower when aa-tRNA binds to a cognate versus a near-cognate codon. The resulting longer residence time of EF-Tu on the ribosome may be important for promoting accommodation of the cognate aminoacyl-tRNA into the A-site.


Subject(s)
GTP Phosphohydrolases/chemistry , Peptide Elongation Factor Tu/chemistry , RNA, Transfer, Amino Acyl/genetics , Ribosomes/genetics , Anticodon/genetics , Codon/genetics , Escherichia coli/genetics , GTP Phosphohydrolases/genetics , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/chemistry , Hydrolysis , Kinetics , Peptide Elongation Factor Tu/genetics , Protein Biosynthesis/genetics , Protein Conformation , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Transfer, Amino Acyl/chemistry , Ribosomes/chemistry
3.
Nucleic Acids Res ; 46(16): 8641-8650, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30107565

ABSTRACT

According to the traditional view, GTPases act as molecular switches, which cycle between distinct 'on' and 'off' conformations bound to GTP and GDP, respectively. Translation elongation factor EF-Tu is a GTPase essential for prokaryotic protein synthesis. In its GTP-bound form, EF-Tu delivers aminoacylated tRNAs to the ribosome as a ternary complex. GTP hydrolysis is thought to cause the release of EF-Tu from aminoacyl-tRNA and the ribosome due to a dramatic conformational change following Pi release. Here, the crystal structure of Escherichia coli EF-Tu in complex with a non-hydrolysable GTP analogue (GDPNP) has been determined. Remarkably, the overall conformation of EF-Tu·GDPNP displays the classical, open GDP-bound conformation. This is in accordance with an emerging view that the identity of the bound guanine nucleotide is not 'locking' the GTPase in a fixed conformation. Using a single-molecule approach, the conformational dynamics of various ligand-bound forms of EF-Tu were probed in solution by fluorescence resonance energy transfer. The results suggest that EF-Tu, free in solution, may sample a wider set of conformations than the structurally well-defined GTP- and GDP-forms known from previous X-ray crystallographic studies. Only upon binding, as a ternary complex, to the mRNA-programmed ribosome, is the well-known, closed GTP-bound conformation, observed.


Subject(s)
Escherichia coli/chemistry , Guanosine Triphosphate/chemistry , Peptide Elongation Factor Tu/chemistry , Protein Conformation , Crystallography, X-Ray , Escherichia coli/genetics , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/genetics , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/analogs & derivatives , Peptide Elongation Factor Tu/genetics , Protein Biosynthesis/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribosomes/chemistry , Ribosomes/genetics
4.
Bioconjug Chem ; 27(9): 2176-87, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27501274

ABSTRACT

Approximately 30% of the ATP generated in the living cell is utilized by P-type ATPase primary active transporters to generate and maintain electrochemical gradients across biological membranes. P-type ATPases undergo large conformational changes during their functional cycle to couple ATP hydrolysis in the cytoplasmic domains to ion transport across the membrane. The Ca(2+)-ATPase from Listeria monocytogenes, LMCA1, was found to be a suitable model of P-type ATPases and was engineered to facilitate single-molecule FRET studies of transport-related structural changes. Mutational analyses of the endogenous cysteine residues in LMCA1 were performed to reduce background labeling without compromising activity. Pairs of cysteines were introduced into the optimized low-reactivity background, and labeled with maleimide derivatives of Cy3 and Cy5 resulting in site-specifically double-labeled protein with moderate activity. Ensemble and confocal single-molecule FRET studies revealed changes in FRET distribution related to structural changes during the transport cycle, consistent with those observed by X-ray crystallography for the sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA). Notably, the cytosolic headpiece of LMCA1 was found to be distinctly more compact in the E1 state than in the E2 state. Thus, the established experimental system should allow future real-time FRET studies of the structural dynamics of LMCA1 as a representative P-type ATPase.


Subject(s)
Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Fluorescence Resonance Energy Transfer , Listeria monocytogenes/enzymology , Protein Engineering , Calcium-Transporting ATPases/chemistry , Maleimides/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation
5.
Nucleic Acids Res ; 43(22): 10893-906, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26578560

ABSTRACT

Upon infection of Escherichia coli by bacteriophage Qß, the virus-encoded ß-subunit recruits host translation elongation factors EF-Tu and EF-Ts and ribosomal protein S1 to form the Qß replicase holoenzyme complex, which is responsible for amplifying the Qß (+)-RNA genome. Here, we use X-ray crystallography, NMR spectroscopy, as well as sequence conservation, surface electrostatic potential and mutational analyses to decipher the roles of the ß-subunit and the first two oligonucleotide-oligosaccharide-binding domains of S1 (OB1-2) in the recognition of Qß (+)-RNA by the Qß replicase complex. We show how three basic residues of the ß subunit form a patch located adjacent to the OB2 domain, and use NMR spectroscopy to demonstrate for the first time that OB2 is able to interact with RNA. Neutralization of the basic residues by mutagenesis results in a loss of both the phage infectivity in vivo and the ability of Qß replicase to amplify the genomic RNA in vitro. In contrast, replication of smaller replicable RNAs is not affected. Taken together, our data suggest that the ß-subunit and protein S1 cooperatively bind the (+)-stranded Qß genome during replication initiation and provide a foundation for understanding template discrimination during replication initiation.


Subject(s)
Allolevivirus/physiology , Genome, Viral , Q beta Replicase/chemistry , Virus Replication , Allolevivirus/genetics , Dimerization , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Mutation , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Q beta Replicase/genetics , Q beta Replicase/metabolism , RNA, Viral/biosynthesis , RNA, Viral/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism
6.
Nucleic Acids Res ; 43(19): 9519-28, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26338772

ABSTRACT

The G-protein EF-Tu, which undergoes a major conformational change when EF-Tu·GTP is converted to EF-Tu·GDP, forms part of an aminoacyl(aa)-tRNA·EF-Tu·GTP ternary complex (TC) that accelerates the binding of aa-tRNA to the ribosome during peptide elongation. Such binding, placing a portion of EF-Tu in contact with the GTPase Associated Center (GAC), is followed by GTP hydrolysis and Pi release, and results in formation of a pretranslocation (PRE) complex. Although tRNA movement through the ribosome during PRE complex formation has been extensively studied, comparatively little is known about the dynamics of EF-Tu interaction with either the ribosome or aa-tRNA. Here we examine these dynamics, utilizing ensemble and single molecule assays employing fluorescent labeled derivatives of EF-Tu, tRNA, and the ribosome to measure changes in either FRET efficiency or fluorescence intensity during PRE complex formation. Our results indicate that ribosome-bound EF-Tu separates from the GAC prior to its full separation from aa-tRNA, and suggest that EF-Tu·GDP dissociates from the ribosome by two different pathways. These pathways correspond to either reversible EF-Tu·GDP dissociation from the ribosome prior to the major conformational change in EF-Tu that follows GTP hydrolysis, or irreversible dissociation after or concomitant with this conformational change.


Subject(s)
Guanosine Diphosphate/metabolism , Peptide Chain Elongation, Translational , Peptide Elongation Factor Tu/metabolism , Ribosomes/metabolism , Fluorescence Resonance Energy Transfer , Guanosine Triphosphate/metabolism , RNA, Transfer, Amino Acyl/metabolism , Ribosomal Proteins/metabolism
7.
J Struct Biol ; 191(1): 10-21, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26073967

ABSTRACT

Translation elongation factor EF-Tu belongs to the superfamily of guanine-nucleotide binding proteins, which play key cellular roles as regulatory switches. All G-proteins require activation via exchange of GDP for GTP to carry out their respective tasks. Often, guanine-nucleotide exchange factors are essential to this process. During translation, EF-Tu:GTP transports aminoacylated tRNA to the ribosome. GTP is hydrolyzed during this process, and subsequent reactivation of EF-Tu is catalyzed by EF-Ts. The reaction path of guanine-nucleotide exchange is structurally poorly defined for EF-Tu and EF-Ts. We have determined the crystal structures of the following reaction intermediates: two structures of EF-Tu:GDP:EF-Ts (2.2 and 1.8Å resolution), EF-Tu:PO4:EF-Ts (1.9Å resolution), EF-Tu:GDPNP:EF-Ts (2.2Å resolution) and EF-Tu:GDPNP:pulvomycin:Mg(2+):EF-Ts (3.5Å resolution). These structures provide snapshots throughout the entire exchange reaction and suggest a mechanism for the release of EF-Tu in its GTP conformation. An inferred sequence of events during the exchange reaction is presented.


Subject(s)
Guanine Nucleotides/chemistry , Guanine Nucleotides/metabolism , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/metabolism , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Models, Molecular
8.
ACS Chem Biol ; 9(10): 2421-31, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25126896

ABSTRACT

The universally conserved translation elongation factor EF-Tu delivers aminoacyl(aa)-tRNA in the form of an aa-tRNA·EF-Tu·GTP ternary complex (TC) to the ribosome where it binds to the cognate mRNA codon within the ribosomal A-site, leading to formation of a pretranslocation (PRE) complex. Here we describe preparation of QSY9 and Cy5 derivatives of the variant E348C-EF-Tu that are functional in translation elongation. Together with fluorophore derivatives of aa-tRNA and of ribosomal protein L11, located within the GTPase associated center (GAC), these labeled EF-Tus allow development of two new FRET assays that permit the dynamics of distance changes between EF-Tu and both L11 (Tu-L11 assay) and aa-tRNA (Tu-tRNA assay) to be determined during the decoding process. We use these assays to examine: (i) the relative rates of EF-Tu movement away from the GAC and from aa-tRNA during decoding, (ii) the effects of the misreading-inducing antibiotics streptomycin and paromomycin on tRNA selection at the A-site, and (iii) how strengthening the binding of aa-tRNA to EF-Tu affects the rate of EF-Tu movement away from L11 on the ribosome. These FRET assays have the potential to be adapted for high throughput screening of ribosomal antibiotics.


Subject(s)
Peptide Elongation Factor Tu/metabolism , Protein Biosynthesis/physiology , RNA, Transfer, Amino Acyl/chemistry , Ribosomal Proteins/metabolism , Ribosomes/physiology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Guanosine Triphosphate/metabolism , Kinetics , Models, Molecular , Mutation/genetics , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/genetics , Protein Conformation , RNA, Transfer, Amino Acyl/metabolism
9.
Cancer Cell Int ; 14(1): 17, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24571548

ABSTRACT

BACKGROUND: The oncogene PTI-1 was originally isolated from a prostate cancer cell line by its capability to transform rat fibroblasts. The PTI-1 mRNA has a very eccentric structure as the 5'UTR is similar to prokaryotic 23S rRNA, while the major open reading frame and the 3'UTR corresponds to a part of the mRNA encoding human translation elongation factor eEF1A1. Thus, the largest open reading frame encodes a truncated version of eEF1A1 lacking the first 67 amino acids, while having three unique N-terminal amino acids. Previously, the UTRs were shown to be a prerequisite for the transforming capacity of the PTI-1 transcript. In this study, we have investigated the possible role of the UTRs in regulating protein expression and localization. METHODS: The protein expression profiles of a number of PTI-1 mRNA variants were studied in vitro and in vivo. Furthermore, the oncogenic potentials of the same PTI-1 mRNAs were determined by monitoring the capacities of stably transfected cells expressing these mRNAs to induce tumors in nude mice and form foci in cell culture. Finally, the cellular localizations of PTI-1 proteins expressed from these mRNAs were determined by fluorescence microscopy. RESULTS: The PTI-1 mRNA was found to give rise to multiple protein products that potentially originate from translation initiation at downstream, inframe AUGs within the major open reading frame. At least one of the truncated protein variants was also found to be oncogenic. However, the UTRs did not appear to influence the amount and identities of these truncated protein products. In contrast, our localization studies showed that the UTRs of the transcript promote a nuclear localization of the encoded protein(s). CONCLUSIONS: Translation of the PTI-1 mRNA results in multiple protein products of which (a) truncated variant(s) may play a predominant role during cellular transformation. The PTI-1 UTRs did not seem to play a role in translation regulation, but appeared to contribute to a nuclear localization of the PTI-1 protein(s). This indicates that the PTI-1 protein(s) exert(s) its/their oncogenic function inside the nucleus.

10.
Biochemistry ; 51(13): 2642-51, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22409271

ABSTRACT

During translation, the nucleic acid language employed by genes is translated into the amino acid language used by proteins. The translator is the ribosome, while the dictionary employed is known as the genetic code. The genetic information is presented to the ribosome in the form of a mRNA, and tRNAs connect the two languages. Translation takes place in three steps: initiation, elongation, and termination. After a protein has been synthesized, the components of the translation apparatus are recycled. During each phase of translation, the ribosome collaborates with specific translation factors, which secure a proper balance between speed and fidelity. Notably, initiation, termination, and ribosomal recycling occur only once per protein produced during normal translation, while the elongation step is repeated a large number of times, corresponding to the number of amino acids constituting the protein of interest. In bacteria, elongation factor Tu plays a central role during the selection of the correct amino acids throughout the elongation phase of translation. Elongation factor Tu is the main subject of this review.


Subject(s)
Peptide Elongation Factor Tu/metabolism , Ribosomes/metabolism , Models, Molecular , Peptide Elongation Factor Tu/chemistry , Protein Biosynthesis
11.
Mol Cell ; 41(4): 419-31, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21329880

ABSTRACT

Transcript-selective translational regulation of epithelial-mesenchymal transition (EMT) by transforming growth factor-ß (TGF-ß) is directed by the hnRNP E1-containing TGF-ß-activated-translational (BAT) mRNP complex. Herein, eukaryotic elongation factor-1 A1 (eEF1A1) is identified as an integral component of the BAT complex. Translational silencing of Dab2 and ILEI, two EMT transcripts, is mediated by the binding of hnRNP E1 and eEF1A1 to their 3'UTR BAT element, whereby hnRNP E1 stalls translational elongation by inhibiting the release of eEF1A1 from the ribosomal A site. TGF-ß-mediated hnRNP E1 phosphorylation, through Akt2, disrupts the BAT complex, thereby restoring translation of target EMT transcripts. Attenuation of hnRNP E1 expression in two noninvasive breast epithelial cells (NMuMG and MCF-7) not only induced EMT but also enabled cells to form metastatic lesions in vivo. Thus, translational regulation by TGF-ß at the elongation stage represents a critical checkpoint coordinating the expression of EMT transcripts required during development and in tumorigenesis and metastatic progression.


Subject(s)
Neoplasms/genetics , Peptide Chain Elongation, Translational/physiology , Ribonucleoproteins/metabolism , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Eukaryotic Initiation Factor-1/genetics , Eukaryotic Initiation Factor-1/metabolism , Mice , Mice, Inbred BALB C , Neoplasms/metabolism , Peptide Biosynthesis, Nucleic Acid-Independent/physiology , Ribonucleoproteins/genetics , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
PLoS One ; 6(12): e29525, 2011.
Article in English | MEDLINE | ID: mdl-22216304

ABSTRACT

Legionella pneumophila, which is the causative organism of Legionnaires disease, translocates numerous effector proteins into the host cell cytosol by a type IV secretion system during infection. Among the most potent effector proteins of Legionella are glucosyltransferases (lgt's), which selectively modify eukaryotic elongation factor (eEF) 1A at Ser-53 in the GTP binding domain. Glucosylation results in inhibition of protein synthesis. Here we show that in vitro glucosylation of yeast and mouse eEF1A by Lgt3 in the presence of the factors Phe-tRNA(Phe) and GTP was enhanced 150 and 590-fold, respectively. The glucosylation of eEF1A catalyzed by Lgt1 and 2 was increased about 70-fold. By comparison of uncharged tRNA with two distinct aminoacyl-tRNAs (His-tRNA(His) and Phe-tRNA(Phe)) we could show that aminoacylation is crucial for Lgt-catalyzed glucosylation. Aminoacyl-tRNA had no effect on the enzymatic properties of lgt's and did not enhance the glucosylation rate of eEF1A truncation mutants, consisting of the GTPase domain only or of a 5 kDa peptide covering Ser-53 of eEF1A. Furthermore, binding of aminoacyl-tRNA to eEF1A was not altered by glucosylation. Taken together, our data suggest that the ternary complex, consisting of eEF1A, aminoacyl-tRNA and GTP, is the bona fide substrate for lgt's.


Subject(s)
Glucosyltransferases/metabolism , Legionella pneumophila/enzymology , Peptide Elongation Factor 1/metabolism , RNA, Transfer, Amino Acyl/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Biocatalysis , Cell Line , Chromatography, Liquid , Cloning, Molecular , Glucosyltransferases/genetics , Glycosylation , Macrophages/parasitology , Mice , Models, Molecular , Peptide Elongation Factor 1/chemistry , Substrate Specificity , Tandem Mass Spectrometry
13.
Proc Natl Acad Sci U S A ; 107(24): 10884-9, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20534494

ABSTRACT

The RNA-dependent RNA polymerase core complex formed upon infection of Escherichia coli by the bacteriophage Qbeta is composed of the viral catalytic beta-subunit as well as the host translation elongation factors EF-Tu and EF-Ts, which are required for initiation of RNA replication. We have determined the crystal structure of the complex between the beta-subunit and the two host proteins to 2.5-A resolution. Whereas the basic catalytic machinery in the viral subunit appears similar to other RNA-dependent RNA polymerases, a unique C-terminal region of the beta-subunit engages in extensive interactions with EF-Tu and may contribute to the separation of the transient duplex formed between the template and the nascent product to allow exponential amplification of the phage genome. The evolution of resistance by the host appears to be impaired because of the interactions of the beta-subunit with parts of EF-Tu essential in recognition of aminoacyl-tRNA.


Subject(s)
Q beta Replicase/chemistry , Allolevivirus/enzymology , Allolevivirus/genetics , Amino Acid Sequence , Base Sequence , Catalytic Domain , Crystallography, X-Ray , DNA Primers/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factors/chemistry , Peptide Elongation Factors/genetics , Protein Multimerization , Protein Structure, Tertiary , Q beta Replicase/genetics , Q beta Replicase/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sequence Homology, Amino Acid , Static Electricity , Viral Proteins/chemistry , Viral Proteins/genetics
14.
Protein Eng Des Sel ; 23(3): 129-36, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20083494

ABSTRACT

Here we describe the design, preparation and characterization of 10 EF-Tu mutants of potential utility for the study of Escherichia coli elongation factor Tu (EF-Tu) interaction with tRNA by a fluorescence resonance energy transfer assay. Each mutant contains a single cysteine residue at positions in EF-Tu that are proximal to tRNA sites within the aminoacyl-tRNA.EF-Tu.GTP ternary complex that have previously been labeled with fluorophores. These positions fall in the 323-326 and 344-348 regions of EF-Tu, and at the C terminus. The EF-Tus were isolated as N-terminal fusions to glutathione S-transferase (GST), which was cleaved to yield intact EF-Tus. The mutant EF-Tus were tested for binding to GDP, binding to tRNA in gel retardation and protection assays, and activity in poly-U translation in vitro. The results indicate that at least three EF-Tu mutants, K324C, G325C and E348C, are suitable for further studies. Remarkably, GST fusions that were not cleaved were also active in the various assays, despite the N-terminal fusion.


Subject(s)
Escherichia coli/enzymology , Fluorescence Resonance Energy Transfer , Mutant Proteins/metabolism , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/metabolism , Protein Engineering , RNA, Transfer/metabolism , Binding Sites , Electrophoretic Mobility Shift Assay , Escherichia coli/genetics , Factor Xa/metabolism , Guanosine Diphosphate/metabolism , Hydrolysis , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/isolation & purification , Mutation , Nucleic Acid Conformation , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/isolation & purification , Peptides/metabolism , Protein Conformation , RNA, Transfer/chemistry , Sequence Analysis, DNA , Staining and Labeling
15.
J Cell Biochem ; 105(3): 847-58, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18756455

ABSTRACT

Eukaryotic translation elongation factor 1A (eEF1A) is a guanine-nucleotide binding protein, which transports aminoacylated tRNA to the ribosomal A site during protein synthesis. In a yeast two-hybrid screening of a human skeletal muscle cDNA library, a novel eEF1A binding protein, immunoglobulin-like and fibronectin type III domain containing 1 (IGFN1), was discovered, and its interaction with eEF1A was confirmed in vitro. IGFN1 is specifically expressed in skeletal muscle and presents immunoglobulin I and fibronectin III sets of domains characteristic of sarcomeric proteins. IGFN1 shows sequence and structural homology to myosin binding protein-C fast and slow-type skeletal muscle isoforms. IGFN1 is substantially upregulated during muscle denervation. We propose a model in which this increased expression of IGFN1 serves to down-regulate protein synthesis via interaction with eEF1A during denervation.


Subject(s)
Carrier Proteins/metabolism , Peptide Elongation Factor 1/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cells, Cultured , Humans , Mice , Microscopy, Confocal , Molecular Sequence Data , Peptide Elongation Factor 1/genetics , Protein Biosynthesis/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Alignment
16.
Biol Chem ; 389(9): 1239-49, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18713011

ABSTRACT

The ribosome is the macromolecular machine responsible for translating the genetic code into polypeptide chains. Despite impressive structural and kinetic studies of the translation process, a number of challenges remain with respect to understanding the dynamic properties of the translation apparatus. Single-molecule techniques hold the potential of characterizing the structural and mechanical properties of macromolecules during their functional cycles in real time. These techniques often necessitate the specific coupling of biologically active molecules to a surface. Here, we describe a procedure for such coupling of functionally active ribosomes that permits single-molecule studies of protein synthesis. Oxidation with NaIO4 at the 3' end of 23S rRNA and subsequent reaction with a biotin hydrazide produces biotinylated 70S ribosomes, which can be immobilized on a streptavidin-coated surface. The surface-attached ribosomes are fully active in poly(U) translation in vitro, synthesizing poly(Phe) at a rate of 3-6 peptide bonds/s per active ribosome at 37 degrees C. Specificity of binding of biotinylated ribosomes to a streptavidin-coated quartz surface was confirmed by observation of individual fluorescently labeled, biotinylated 70S ribosomes, using total internal reflection fluorescence microscopy. Functional interactions of the immobilized ribosomes with various components of the protein synthesis apparatus are shown by use of surface plasmon resonance.


Subject(s)
Biotin/analogs & derivatives , Protein Biosynthesis , Protein Modification, Translational , Ribosomes/metabolism , Streptavidin/metabolism , Biotin/metabolism , Biotinylation , Escherichia coli , Poly U/metabolism , Surface Plasmon Resonance
17.
J Biol Chem ; 282(49): 35629-37, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17925388

ABSTRACT

The interactions of elongation factor 1A (eEF1A) from Saccharomyces cerevisiae with elongation factor 1Balpha (eEF1Balpha), guanine nucleotides, and aminoacyl-tRNA were studied kinetically by fluorescence stopped-flow. eEF1A has similar affinities for GDP and GTP, 0.4 and 1.1 microm, respectively. Dissociation of nucleotides from eEF1A in the absence of the guanine nucleotide exchange factor is slow (about 0.1 s(-1)) and is accelerated by eEF1Balpha by 320-fold and 250-fold for GDP and GTP, respectively. The rate constant of eEF1Balpha binding to eEF1A (10(7)-10(8) M (-1) s(-1)) is independent of guanine nucleotides. At the concentrations of nucleotides and factors prevailing in the cell, the overall exchange rate is expected to be in the range of 6 s(-1), which is compatible with the rate of protein synthesis in the cell. eEF1A.GTP binds Phe-tRNA(Phe) with a K(d) of 3 nm, whereas eEF1A.GDP shows no significant binding, indicating that eEF1A has similar tRNA binding properties as its prokaryotic homolog, EF-Tu.


Subject(s)
Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Peptide Elongation Factor 1/metabolism , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Phe/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Guanosine Diphosphate/chemistry , Guanosine Triphosphate/genetics , Kinetics , Peptide Elongation Factor 1/chemistry , Peptide Elongation Factor Tu , Protein Binding , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Phe/genetics , Saccharomyces cerevisiae Proteins/chemistry
18.
J Biol Chem ; 281(30): 21139-21146, 2006 Jul 28.
Article in English | MEDLINE | ID: mdl-16717093

ABSTRACT

Elongation factor Ts (EF-Ts) is the guanine nucleotide exchange factor for elongation factor Tu (EF-Tu). An important feature of the nucleotide exchange is the structural rearrangement of EF-Tu in the EF-Tu.EF-Ts complex caused by insertion of Phe-81 of EF-Ts between His-84 and His-118 of EF-Tu. In this study, the contribution of His-118 to nucleotide release was studied by pre-steady state kinetic analysis of nucleotide exchange in EF-Tu mutants in which His-118 was replaced by Ala or Glu. Intrinsic as well as EF-Ts-catalyzed release of GDP/GTP was affected by the mutations, resulting in an approximately 10-fold faster spontaneous nucleotide release and a 10-50-fold slower EF-Ts-catalyzed nucleotide release. The effects are attributed to the interference of the mutations with the EF-Ts-induced movements of the P-loop of EF-Tu and changes at the domain 1/3 interface, leading to the release of the beta-phosphate group of GTP/GDP. The K(d) for GTP is increased by more than 40 times when His-118 is replaced with Glu, which may explain the inhibition by His-118 mutations of aminoacyl-tRNA binding to EF-Tu. The mutations had no effect on EF-Tu-dependent delivery of aminoacyl-tRNA to the ribosome.


Subject(s)
Escherichia coli/metabolism , Guanine Nucleotides/chemistry , Peptide Elongation Factor Tu/chemistry , Catalysis , Guanine Nucleotide Exchange Factors/chemistry , Histidine/chemistry , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Nucleotides/chemistry , Peptide Elongation Factor Tu/physiology , Phenylalanine/chemistry , Protein Binding , Protein Structure, Tertiary , Ribosomes/chemistry
19.
Biochim Biophys Acta ; 1727(2): 116-24, 2005 Feb 14.
Article in English | MEDLINE | ID: mdl-15716006

ABSTRACT

The prostate tumor-inducing gene 1 (PTI-1) transcript is detected in various human carcinoma cells. PTI-1 is reported to consist of a 5' untranslated region (5' UTR) homologous to mycoplasma 23S rRNA and a coding region corresponding to a truncated and mutated form of the translation elongation factor 1A, eEF1A. We have found that the PTI-1 transcript may encode a truncated, but not mutated, form of the human isoform eEF1A1. Additionally, the 5' UTR sequence of PTI-1 from genomic DNA of different cell lines and blood samples varies from the original sequence. This 5' -UTR region of PTI-1 presents a fusion of E. coli and Mycoplasma hyorhinis 23S rRNA. We have overexpressed the potential PTI-1 protein in E. coli and various human cell lines. The resulting protein could be detected by western blotting using anti-eEF1A antibodies. However, we were unable to detect the PTI-1 protein in LNCaP cell extracts. The potential roles of the PTI-1 protein in carcinogenesis and the origin of the PTI-1 gene in the human genome are discussed.


Subject(s)
Oncogene Proteins/genetics , Peptide Chain Elongation, Translational/genetics , Transcription, Genetic/genetics , Base Sequence , Cell Line, Tumor , DNA Primers , Escherichia coli/genetics , Humans , Mycoplasma/genetics , Peptide Elongation Factor 1 , Polymerase Chain Reaction
20.
J Biol Chem ; 279(3): 1878-84, 2004 Jan 16.
Article in English | MEDLINE | ID: mdl-14583631

ABSTRACT

Elongation factor Ts (EF-Ts) is the guanine-nucleotide exchange factor of elongation factor Tu (EF-Tu), which promotes the binding of aminoacyl-tRNA to the mRNA-programmed ribosome in prokaryotes. The EF-Tu.EF-Ts complex, one of the EF-Tu complexes during protein synthesis, is also a component of RNA-dependent RNA polymerases like the polymerase from coliphage Qbeta. The present study shows that the Escherichia coli mutant GRd.tsf lacking the coiled-coil motif of EF-Ts is completely resistant to phage Qbeta and that Qbeta-polymerase complex formation is not observed. GRd.tsf is the first E. coli mutant ever described that is unable to form a Qbeta-polymerase complex while still maintaining an almost normal growth behavior. The phage resistance correlates with an observed instability of the mutant EF-Tu.EF-Ts complex in the presence of guanine nucleotides. Thus, the mutant EF-Tu.EF-Ts is the first EF-Tu.EF-Ts complex ever described that is completely inactive in the Qbeta-polymerase complex despite its almost full activity in protein synthesis. We propose that the role of EF-Ts in the Qbeta-polymerase complex is to control and trap EF-Tu in a stable conformation with affinity for RNA templates while unable to bind aminoacyl-tRNA.


Subject(s)
Coliphages/physiology , Escherichia coli Proteins/physiology , Escherichia coli/virology , Peptide Elongation Factors/physiology , DNA-Directed RNA Polymerases/metabolism , Peptide Elongation Factors/chemistry , Protein Biosynthesis , Protein Conformation , RNA, Viral/biosynthesis , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...