Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
FEBS J ; 286(3): 536-554, 2019 02.
Article in English | MEDLINE | ID: mdl-30570212

ABSTRACT

The rules of the genetic code are established by aminoacyl-tRNA synthetases (aaRSs) enzymes, which covalently link tRNA with the cognate amino acid. Many aaRSs are involved in diverse cellular processes beyond translation, acting alone, or in complex with other proteins. However, studies of aaRS noncanonical assembly and functions in plants are scarce, as are structural studies of plant aaRSs. Here, we have solved the crystal structure of Arabidopsis thaliana cytosolic seryl-tRNA synthetase (SerRS), which is the first crystallographic structure of a plant aaRS. Arabidopsis SerRS displays structural features typical of canonical SerRSs, except for a unique intrasubunit disulfide bridge. In a yeast two-hybrid screen, we identified BEN1, a protein involved in the metabolism of plant brassinosteroid hormones, as a protein interactor of Arabidopsis SerRS. The SerRS:BEN1 complex is one of the first protein complexes of plant aaRSs discovered so far, and is a rare example of an aaRS interacting with an enzyme involved in primary or secondary metabolism. To pinpoint regions responsible for this interaction, we created truncated variants of SerRS and BEN1, and identified that the interaction interface involves the SerRS globular catalytic domain and the N-terminal extension of BEN1 protein. BEN1 does not have a strong impact on SerRS aminoacylation activity, indicating that the primary function of the complex is not the modification of SerRS canonical activity. Perhaps SerRS performs as yet unknown noncanonical functions mediated by BEN1. These findings indicate that - via SerRS and BEN1 - a link exists between the protein translation and steroid metabolic pathways of the plant cell. DATABASE: Structural data are available in the PDB under the accession number PDB ID 6GIR.


Subject(s)
Alcohol Oxidoreductases/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis/chemistry , Serine-tRNA Ligase/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Sequence , Arabidopsis/enzymology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Binding Sites , Brassinosteroids/biosynthesis , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Kinetics , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine-tRNA Ligase/genetics , Serine-tRNA Ligase/metabolism , Substrate Specificity , Two-Hybrid System Techniques
2.
Biochimie ; 147: 36-45, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29273296

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) decode genetic information by coupling tRNAs with cognate amino acids. In the archaeon Methanothermobacter thermautotrophicus arginyl- and seryl-tRNA synthetase (ArgRS and SerRS, respectively) form a complex which enhances serylation and facilitates tRNASer recycling through its association with the ribosome. Yet, the way by which complex formation participates in Arg-tRNAArg synthesis is still unresolved. Here we utilized pull down and surface plasmon resonance experiments with truncated ArgRS variants to demonstrate that ArgRS uses its N-terminal domain to establish analogous interactions with both SerRS and cognate tRNAArg, providing a rationale for the lack of detectable SerRS•[ArgRS•tRNAArg] complex. In contrast, stable ternary ArgRS•[SerRS•tRNASer] complex was easily detected supporting the model wherein ArgRS operates in serylation by modulating SerRS affinity toward tRNASer. We also found that the interaction with SerRS suppresses arginylation of unmodified tRNAArg by ArgRS, which, by itself, does not discriminate against tRNAArg substrates lacking posttranscriptional modifications. Hence, there is a fundamentally different participation of the protein partners in Arg-tRNA and Ser-tRNA synthesis. Propensity of the ArgRS•SerRS complex to exclude unmodified tRNAs from translation leads to an attractive hypothesis that SerRS•ArgRS complex might act in vivo as a safeguarding switch that improves translation accuracy.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Methanobacteriaceae/enzymology , Amino Acyl-tRNA Synthetases/chemistry , Arginine/metabolism , Molecular Docking Simulation , Protein Conformation , Substrate Specificity
3.
Mol Biosyst ; 10(12): 3207-16, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25272963

ABSTRACT

Recently described and characterized Bradyrhizobium japonicum glycine:[carrier protein] ligase 1 (Bj Gly:CP ligase 1), a homologue of methanogenic type seryl-tRNA synthetase (SerRS) is an intriguing enzyme whose physiological role is not yet known. While aminoacyl-tRNA synthetases supply ribosome with amino acids for protein biosynthesis, this homologue transfers the activated amino acid to a specific carrier protein. Despite remarkable structural similarity between the Bj Gly:CP ligase 1 and the catalytic core domain of methanogenic type SerRS, the ligase displays altered and relaxed substrate specificity. In contrast to methanogenic SerRS which exclusively activates serine, the Bj Gly:CP ligase 1 predominantly activates glycine. Besides, it shows low activity in the presence of alanine, but it is incapable of activating serine. The detailed computational study aiming to address this unexpected substrate specificity toward the small aliphatic amino acids revealed the A281G Bj Gly:CP ligase 1 mutant as the most promising candidate with reconstituted catalytic activity toward the larger substrates. The A281G mutation is predicted to increase the active site volume, allowing alanine and serine to establish important hydrogen bonds within the active site, and to adopt an optimal orientation for the reaction. The results were tested by the site-directed mutagenesis experiments coupled with in vitro kinetic assays. It was found that the A281G substitution greatly affects the enzyme specificity and allows efficient activation of both polar and small aliphatic amino acids (serine, glycine and alanine), confirming predictions and conclusions based on molecular dynamics simulations.


Subject(s)
Amino Acid Substitution , Bradyrhizobium/enzymology , Serine-tRNA Ligase/chemistry , Alanine/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Bradyrhizobium/genetics , Catalytic Domain , Ligands , Magnesium/chemistry , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Serine/chemistry , Serine-tRNA Ligase/genetics , Substrate Specificity , Zinc/chemistry
4.
Nucleic Acids Res ; 42(8): 5191-201, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569352

ABSTRACT

Aminoacyl-tRNA synthetases (aaRS) are essential enzymes catalyzing the formation of aminoacyl-tRNAs, the immediate precursors for encoded peptides in ribosomal protein synthesis. Previous studies have suggested a link between tRNA aminoacylation and high-molecular-weight cellular complexes such as the cytoskeleton or ribosomes. However, the structural basis of these interactions and potential mechanistic implications are not well understood. To biochemically characterize these interactions we have used a system of two interacting archaeal aaRSs: an atypical methanogenic-type seryl-tRNA synthetase and an archaeal ArgRS. More specifically, we have shown by thermophoresis and surface plasmon resonance that these two aaRSs bind to the large ribosomal subunit with micromolar affinities. We have identified the L7/L12 stalk and the proteins located near the stalk base as the main sites for aaRS binding. Finally, we have performed a bioinformatics analysis of synonymous codons in the Methanothermobacter thermautotrophicus genome that supports a mechanism in which the deacylated tRNAs may be recharged by aaRSs bound to the ribosome and reused at the next occurrence of a codon encoding the same amino acid. These results suggest a mechanism of tRNA recycling in which aaRSs associate with the L7/L12 stalk region to recapture the tRNAs released from the preceding ribosome in polysomes.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Archaea/enzymology , Protein Biosynthesis , RNA, Transfer/metabolism , Ribosomes/enzymology , Arginine-tRNA Ligase/metabolism , Genome, Archaeal , Methanobacteriaceae/genetics , Ribosomal Proteins/metabolism , Serine-tRNA Ligase/metabolism
5.
Structure ; 21(4): 614-26, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23541895

ABSTRACT

Amino acid:[carrier protein] ligases (aa:CP ligases) are recently discovered enzymes that are highly similar to class II aminoacyl-tRNA synthetases (aaRSs). However, while aaRSs aminoacylate tRNA and supply building blocks for ribosomal translation, aa:CP ligases transfer activated amino acids to the phosphopantetheine group of small carrier proteins. We have solved the crystal structure of an aa:CP ligase complexed with the carrier protein (CP). The CP prosthetic group enters the active site from a different direction than tRNA in class II aaRS complexes through an idiosyncratic tunnel. CP binds to aa:CP ligase in a fundamentally different manner compared to tRNA binding by structurally closely related aaRSs. Based on crystallographic analysis, an enzyme of altered CP specificity was designed, and the mechanism of amino acid transfer to the prosthetic group was proposed. The presented study reveals how a conserved class II aaRS catalytic core can adapt to another function through minor structural alterations.


Subject(s)
Amino Acyl-tRNA Synthetases/chemistry , Bradyrhizobium/chemistry , Carrier Proteins/chemistry , Ligases/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Protein Conformation , Amino Acyl-tRNA Synthetases/metabolism , Aminoacylation , Carrier Proteins/metabolism , Catalytic Domain/genetics , Crystallization , Escherichia coli , Kinetics , Ligases/metabolism , Molecular Structure , Multiprotein Complexes/metabolism , Protein Binding
6.
Arch Biochem Biophys ; 529(2): 122-30, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23228595

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) catalyze the attachment of amino acids to their cognate tRNAs to establish the genetic code. To obtain the high degree of accuracy that is observed in translation, these enzymes must exhibit strict substrate specificity for their cognate amino acids and tRNAs. We studied the requirements for tRNA(Ser) recognition by maize cytosolic seryl-tRNA synthetase (SerRS). The enzyme efficiently recognized bacterial and eukaryotic tRNAs(Ser) indicating that it can accommodate various types of tRNA(Ser) structures. Discriminator base G73 is crucial for recognition by cytosolic SerRS. Although cytosolic SerRS efficiently recognized bacterial tRNAs(Ser), it is localized exclusively in the cytosol. The fidelity of maize cytosolic and dually targeted organellar SerRS with respect to amino acid recognition was compared. Organellar SerRS exhibited higher discrimination against tested non-cognate substrates as compared with cytosolic counterpart. Both enzymes showed pre-transfer editing activity implying their high overall accuracy. The contribution of various reaction pathways in the pre-transfer editing reactions by maize enzymes were different and dependent on the non-cognate substrate. The fidelity mechanisms of maize organellar SerRS, high discriminatory power and proofreading, indicate that aaRSs in general may play an important role in translational quality control in plant mitochondria and chloroplasts.


Subject(s)
Amino Acids/chemistry , Amino Acids/metabolism , Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/metabolism , Zea mays/enzymology , Enzyme Activation , Substrate Specificity
7.
Eur J Pharmacol ; 691(1-3): 251-60, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22687816

ABSTRACT

A series of porphyrins, tetrapyrrole natural organic compounds, are evaluated here as endogenous anti-inflammatory agents. They directly inhibit the activity of Fyn, a non-receptor Src-family tyrosine kinase, triggering anti-inflammatory events associated with down-regulation of T-cell receptor signal transduction, leading to inhibition of tumor necrosis factor alpha (TNF-α) production. This is one of the major pro-inflammatory cytokines, associated with diseases such as diabetes, tumorigenesis, rheumatoid arthritis, and inflammatory bowel disease. Porphyrins, as a chemical class, inhibited Fyn kinase activity in a non-competitive, linear-mixed fashion. In cell-based in vitro experiments on polymorphonuclear cells, porphyrins inhibited TNF-α cytokine production, T-cell proliferation, and the generation of free radicals in the oxidative burst, in a concentration-related manner. In vivo, lipopolysaccharide-induced TNF-α production in mice was inhibited by several of the porphyrins. These findings may be very important for the overall understanding of the role(s) of porphyrins in inflammation and their possible application as new anti-inflammatory agents.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Porphyrins/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Computational Biology , Humans , Kinetics , Lipopolysaccharides/pharmacology , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Mice , Molecular Docking Simulation , Porphyrins/metabolism , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/chemistry , Proto-Oncogene Proteins c-fyn/metabolism , Respiratory Burst/drug effects , Sf9 Cells , Spodoptera , Thymidine/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
8.
J Mol Biol ; 418(3-4): 145-60, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22306461

ABSTRACT

Translation of mRNA into proteins by the ribosome is universally conserved in all cellular life. The composition and complexity of the translation machinery differ markedly between the three domains of life. Organisms from the domain Archaea show an intermediate level of complexity, sharing several additional components of the translation machinery with eukaryotes that are absent in bacteria. One of these translation factors is initiation factor 6 (IF6), which associates with the large ribosomal subunit. We have reconstructed the 50S ribosomal subunit from the archaeon Methanothermobacter thermautotrophicus in complex with archaeal IF6 at 6.6 Å resolution using cryo-electron microscopy (EM). The structure provides detailed architectural insights into the 50S ribosomal subunit from a methanogenic archaeon through identification of the rRNA expansion segments and ribosomal proteins that are shared between this archaeal ribosome and eukaryotic ribosomes but are mostly absent in bacteria and in some archaeal lineages. Furthermore, the structure reveals that, in spite of highly divergent evolutionary trajectories of the ribosomal particle and the acquisition of novel functions of IF6 in eukaryotes, the molecular binding of IF6 on the ribosome is conserved between eukaryotes and archaea. The structure also provides a snapshot of the reductive evolution of the archaeal ribosome and offers new insights into the evolution of the translation system in archaea.


Subject(s)
Archaeal Proteins/chemistry , Cryoelectron Microscopy/methods , Methanobacteriaceae/metabolism , Prokaryotic Initiation Factors/chemistry , Ribosome Subunits, Large, Archaeal/ultrastructure , Ribosomes/genetics , Archaeal Proteins/ultrastructure , Binding Sites , Methanobacteriaceae/genetics , Prokaryotic Initiation Factors/ultrastructure , Protein Biosynthesis , Ribosome Subunits, Large, Archaeal/chemistry , Ribosomes/metabolism
9.
Biochimie ; 93(10): 1761-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704670

ABSTRACT

Seryl-tRNA synthetases (SerRS) covalently attach serine to cognate tRNA(Ser). Atypical SerRSs, considerably different from canonical enzymes, have been found in methanogenic archaea. A crystal structure of methanogenic-type SerRS revealed a motif within the active site (serine ordering loop; SOL), which undergoes a notable induced-fit rearrangement during serine binding. The loop rearranges from a disordered conformation in the unliganded enzyme, to an ordered structure comprising an α-helix followed by a loop. We performed kinetic and thermodynamic analyses of SerRS variants to establish the role of the SOL in serylation. Thermodynamic data confirmed a linkage between binding of serine and α-helix formation, previously described by the crystallographic analysis. The ability of the SOL to adopt the observed secondary structure was recognized as essential for serine activation. Mutation of Gln400, which according to the structural data establishes the main connection between the serine and the SOL, produced only modest kinetic effects. Kinetic data offer new insights into the coupling of the conformational change with active site assembly. Productive positioning of the SOL may be driven by the interaction between Trp396 and the serine α-amino group. Rapid kinetics reveals that His250, a non-SOL residue, is essential for transfer of serine to tRNA. Modeling data established that accommodation of the tRNA within the active site may require movement of the SOL. This would enable His250 to assist in productive positioning of the 3'-end of the tRNA for the aminoacyl transfer. Thus, the rearrangements of the SOL conformationally adjust the active site for both reaction steps.


Subject(s)
Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/metabolism , Binding Sites , Calorimetry , Catalytic Domain , Chromatography, Ion Exchange , Protein Structure, Secondary , Serine-tRNA Ligase/genetics , Substrate Specificity , Thermodynamics
10.
J Biol Chem ; 286(5): 3396-404, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21098026

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) play an integral role in protein synthesis, functioning to attach the correct amino acid with its cognate tRNA molecule. AaRSs are known to associate into higher-order multi-aminoacyl-tRNA synthetase complexes (MSC) involved in archaeal and eukaryotic translation, although the precise biological role remains largely unknown. To gain further insights into archaeal MSCs, possible protein-protein interactions with the atypical Methanothermobacter thermautotrophicus seryl-tRNA synthetase (MtSerRS) were investigated. Yeast two-hybrid analysis revealed arginyl-tRNA synthetase (MtArgRS) as an interacting partner of MtSerRS. Surface plasmon resonance confirmed stable complex formation, with a dissociation constant (K(D)) of 250 nM. Formation of the MtSerRS·MtArgRS complex was further supported by the ability of GST-MtArgRS to co-purify MtSerRS and by coelution of the two enzymes during gel filtration chromatography. The MtSerRS·MtArgRS complex also contained tRNA(Arg), consistent with the existence of a stable ribonucleoprotein complex active in aminoacylation. Steady-state kinetic analyses revealed that addition of MtArgRS to MtSerRS led to an almost 4-fold increase in the catalytic efficiency of serine attachment to tRNA, but had no effect on the activity of MtArgRS. Further, the most pronounced improvements in the aminoacylation activity of MtSerRS induced by MtArgRS were observed under conditions of elevated temperature and osmolarity. These data indicate that formation of a complex between MtSerRS and MtArgRS provides a means by which methanogenic archaea can optimize an early step in translation under a wide range of extreme environmental conditions.


Subject(s)
Aminoacylation , Arginine-tRNA Ligase/metabolism , Methanobacteriaceae/enzymology , Osmolar Concentration , Serine-tRNA Ligase/metabolism , Temperature , Archaea , Bacterial Proteins/metabolism , Multiprotein Complexes , Protein Biosynthesis , Protein Interaction Mapping/methods
12.
Proc Natl Acad Sci U S A ; 107(33): 14585-90, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20663952

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are ancient and evolutionary conserved enzymes catalyzing the formation of aminoacyl-tRNAs, that are used as substrates for ribosomal protein biosynthesis. In addition to full length aaRS genes, genomes of many organisms are sprinkled with truncated genes encoding single-domain aaRS-like proteins, which often have relinquished their canonical role in genetic code translation. We have identified the genes for putative seryl-tRNA synthetase homologs widespread in bacterial genomes and characterized three of them biochemically and structurally. The proteins encoded are homologous to the catalytic domain of highly diverged, atypical seryl-tRNA synthetases (aSerRSs) found only in methanogenic archaea and are deprived of the tRNA-binding domain. Remarkably, in comparison to SerRSs, aSerRS homologs display different and relaxed amino acid specificity. aSerRS homologs lack canonical tRNA aminoacylating activity and instead transfer activated amino acid to phosphopantetheine prosthetic group of putative carrier proteins, whose genes were identified in the genomic surroundings of aSerRS homologs. Detailed kinetic analysis confirmed that aSerRS homologs aminoacylate these carrier proteins efficiently and specifically. Accordingly, aSerRS homologs were renamed amino acid:[carrier protein] ligases (AMP forming). The enzymatic activity of aSerRS homologs is reminiscent of adenylation domains in nonribosomal peptide synthesis, and thus they represent an intriguing link between programmable ribosomal protein biosynthesis and template-independent nonribosomal peptide synthesis.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Carrier Proteins/metabolism , Peptide Biosynthesis, Nucleic Acid-Independent , Ribosomes/metabolism , Acylation , Agrobacterium tumefaciens/genetics , Agrobacterium tumefaciens/metabolism , Alanine/metabolism , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Bradyrhizobium/genetics , Bradyrhizobium/metabolism , Carrier Proteins/genetics , Catalytic Domain , Crystallography, X-Ray , Kinetics , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/genetics , Serine-tRNA Ligase/metabolism , Transfer RNA Aminoacylation
13.
J Biol Chem ; 284(44): 30643-51, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19734148

ABSTRACT

Seryl-tRNA synthetase (SerRS) from methanogenic archaeon Methanosarcina barkeri, contains an idiosyncratic N-terminal domain, composed of an antiparallel beta-sheet capped by a helical bundle, connected to the catalytic core by a short linker peptide. It is very different from the coiled-coil tRNA binding domain in bacterial-type SerRS. Because the crystal structure of the methanogenic-type SerRSxtRNA complex has not been obtained, a docking model was produced, which indicated that highly conserved helices H2 and H3 of the N-terminal domain may be important for recognition of the extra arm of tRNA(Ser). Based on structural information and the docking model, we have mutated various positions within the N-terminal region and probed their involvement in tRNA binding and serylation. Total loss of activity and inability of the R76A variant to form the complex with cognate tRNA identifies Arg(76) located in helix H2 as a crucial tRNA-interacting residue. Alteration of Lys(79) positioned in helix H2 and Arg(94) in the loop between helix H2 and beta-strand A4 have a pronounced effect on SerRSxtRNA(Ser) complex formation and dissociation constants (K(D)) determined by surface plasmon resonance. The replacement of residues Arg(38) (located in the loop between helix H1 and beta-strand A2), Lys(141) and Asn(142) (from H3), and Arg(143) (between H3 and H4) moderately affect both the serylation activity and the K(D) values. Furthermore, we have obtained a striking correlation between these results and in vivo effects of these mutations by quantifying the efficiency of suppression of bacterial amber mutations, after coexpression of the genes for M. barkeri suppressor tRNA(Ser) and a set of mMbSerRS variants in Escherichia coli.


Subject(s)
Archaea/enzymology , Archaeal Proteins/chemistry , Methanobacteriales/enzymology , Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/genetics , Amino Acid Sequence , Amino Acid Substitution , Amino Acids , Archaeal Proteins/genetics , Binding Sites , Cloning, Molecular , Computer Simulation , Escherichia coli/genetics , Models, Molecular , Protein Conformation
14.
FEMS Microbiol Lett ; 294(1): 111-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19309487

ABSTRACT

Two dissimilar seryl-tRNA synthetases (SerRSs) exist in Methanosarcina barkeri: one of bacterial type (bMbSerRS) and the other resembling SerRSs present only in methanogenic archaea (mMbSerRS). While the expression of the archaeal bMbSerRS gene in Escherichia coli complements the function of thermolabile SerRS at a nonpermissive temperature, mMbSerRS does not. Our recent X-ray structural analysis of mMbSerRS revealed an idiosyncratic N-terminal domain and a catalytic zinc ion in the active site, identifying methanogenic-type SerRSs as atypical members of the SerRS family. To shed further light on substrate discrimination by methanogenic-type SerRS, we developed an in vivo system in E. coli to study tRNA serylation by mMbSerRS variants. We show that coexpression of the M. barkeri SerRS gene, encoding either bacterial- or methanogenic-type SerRS, with the gene for cognate archaeal suppressor tRNA leads to suppression of bacterial amber mutations, implying that the E. coli translation machinery can use serylated tRNA from methanogenic archaea as a substrate in protein synthesis. Furthermore, because serylation of M. barkeri serine-specific tRNA by endogenous E. coli SerRS is negligible, suppression is entirely dependent on recognition between archaeal partners (mMbSerRS/suppressor tRNA(Ser)). Thus, the efficiency of suppression by mMbSerRS variants quantified in the described beta-galactosidase-based reporter system, accurately reflects enzymes' serylation propensity obtained by in vitro kinetic measurements.


Subject(s)
Codon, Nonsense , Methanosarcina barkeri/enzymology , Protein Biosynthesis , RNA, Transfer, Ser/metabolism , RNA, Transfer/metabolism , Serine-tRNA Ligase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genes, Reporter , Models, Molecular , Protein Structure, Tertiary , beta-Galactosidase/metabolism
15.
J Biol Chem ; 284(16): 10706-13, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19228694

ABSTRACT

All seryl-tRNA synthetases (SerRSs) are functional homodimers with a C-terminal active site domain typical for class II aminoacyl-tRNA synthetases and an N-terminal domain involved in tRNA binding. The recently solved three-dimensional structure of Methanosarcina barkeri SerRS revealed the idiosyncratic features of methanogenic-type SerRSs; that is, an active site zinc ion, a unique tRNA binding domain, and an insertion of approximately 30 residues in the catalytic domain, which adopt a helix-turn-helix (HTH) fold. Here, we present biochemical evidence for multiple roles of the HTH motif; it is important for dimerization of the enzyme, contributes to the overall stability, and is critical for the proper positioning of the tRNA binding domain relative to the catalytic domain. The changes in intrinsic fluorescence during denaturation of the wild-type M. barkeri SerRS and of the mutated variant lacking the HTH motif combined with cross-linking and gel analysis of protein subunits during various stages of the unfolding process revealed significantly reduced stability of the mutant dimers. In vitro kinetic analysis of enzymes, mutated in one of the N-terminal helices and the HTH motif, shows impaired tRNA binding and aminoacylation and emphasizes the importance of this domain for the overall architecture of the enzyme. The role of the idiosyncratic HTH motif in dimer stabilization and association between the catalytic and tRNA binding domain has been additionally confirmed by a yeast two-hybrid approach. Furthermore, we provide experimental evidence that tRNA binds across the dimer.


Subject(s)
Helix-Turn-Helix Motifs , Methanosarcina barkeri/enzymology , Protein Structure, Tertiary , Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/genetics , Dimerization , Enzyme Stability , Methanosarcina barkeri/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Quaternary , RNA, Transfer, Amino Acyl/metabolism , Serine-tRNA Ligase/metabolism , Two-Hybrid System Techniques
16.
Plant Cell Rep ; 27(7): 1157-68, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18392626

ABSTRACT

Aminoacyl-tRNA synthetases (AARSs) play a critical role in translation and are thus required in three plant protein-synthesizing compartments: cytosol, mitochondria and plastids. A systematic study had previously shown extensive sharing of organellar AARSs from Arabidopsis thaliana, mostly between mitochondria and chloroplasts. However, distribution of AARSs from monocot species, such as maize, has never been experimentally investigated. Here we demonstrate dual targeting of maize seryl-tRNA synthetase, SerZMo, into both mitochondria and chloroplasts using combination of complementary methods, including in vitro import assay, transient expression analysis of green fluorescent protein (GFP) fusions and immunodetection. We also show that SerZMo dual localization is established by the virtue of an ambiguous targeting peptide. Full-length SerZMo protein fused to GFP is targeted to chloroplast stromules, indicating that SerZMo protein performs its function in plastid stroma. The deletion mutant lacking N-terminal region of the ambiguous SerZMo targeting peptide was neither targeted into mitochondria nor chloroplasts, indicating the importance of this region in both mitochondrial and chloroplastic import.


Subject(s)
Chloroplasts/metabolism , Mitochondria/metabolism , Serine-tRNA Ligase/metabolism , Zea mays/metabolism , Amino Acid Sequence , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Molecular Sequence Data , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine-tRNA Ligase/genetics , Zea mays/enzymology , Zea mays/genetics
17.
FEBS J ; 275(11): 2831-44, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422966

ABSTRACT

Seryl-tRNA synthetase (SerRS) is a class II aminoacyl-tRNA synthetase that catalyzes serine activation and its transfer to cognate tRNA(Ser). Previous biochemical and structural studies have revealed that bacterial- and methanogenic-type SerRSs employ different strategies of substrate recognition. In addition to other idiosyncratic features, such as the active site zinc ion and the unique fold of the N-terminal tRNA-binding domain, methanogenic-type SerRS is, in comparison with bacterial homologues, characterized by a notable shortening of the motif 2 loop. Mutational analysis of Methanosarcina barkeri SerRS (mMbSerRS) was undertaken to identify the active site residues that ensure the specificity of amino acid and tRNA 3'-end recognition. Residues predicted to contribute to the amino acid specificity were selected for mutation according to the crystal structure of mMbSerRS complexed with its cognate aminoacyl-adenylate, whereas those involved in binding of the tRNA 3'-end were identified and mutagenized on the basis of modeling the mMbSerRS:tRNA complex. Although mMbSerRSs variants with an altered serine-binding pocket (W396A, N435A, S437A) were more sensitive to inhibition by threonine and cysteine, none of the mutants was able to activate noncognate amino acids to greater extent than the wild-type enzyme. In vitro kinetics results also suggest that conformational changes in the motif 2 loop are required for efficient serylation.


Subject(s)
Serine-tRNA Ligase/chemistry , Amino Acids/chemistry , Binding Sites , Catalysis , Circular Dichroism , DNA Mutational Analysis , Kinetics , Magnesium/chemistry , Methane/chemistry , Methanosarcina barkeri/enzymology , Models, Molecular , Molecular Conformation , RNA, Transfer/chemistry , Serine/chemistry , Substrate Specificity
18.
Arch Biochem Biophys ; 470(2): 129-38, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18067851

ABSTRACT

Eukaryotic cytosolic seryl-tRNA synthetases (SerRS) have idiosyncratic C-terminal extensions not present in prokaryotic counterparts. The extensions of two eukaryotic SerRSs were subjected to mutagenesis and partial truncation. Only minor parts of the yeast or maize SerRS extensions, adjacent to the catalytic core (7 of 20 and 8 of 26 amino acids, respectively), were found to be indispensable for protein stability. Truncated proteins with substantially shortened extensions displayed unaltered catalytic properties and could complement a Saccharomyces cerevisiae strain with a disrupted SerRS gene, if these proximal regions were left intact. Although the yeast C-terminal SerRS extension is required for Pex21p binding, the maize counterpart with an appended yeast SerRS extension remained incapable of Pex21p binding, implying that additional regions of yeast SerRS may also contribute to the interaction with the peroxin. The proximal region of the eukaryotic SerRS C-terminal extension is indispensable for protein stability, while the remaining part of the extension remains available for other functions, such as species-specific protein:protein interactions.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/metabolism , Binding Sites , Catalysis , Enzyme Activation , Enzyme Stability , Protein Binding , Protein Denaturation , Structure-Activity Relationship
19.
FEBS Lett ; 581(26): 5110-4, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17931630

ABSTRACT

Aminoacyl-tRNA synthetases, a group of enzymes catalyzing aminoacyl-tRNA formation, may possess inherent editing activity to clear mistakes arising through the selection of non-cognate amino acid. It is generally assumed that both editing substrates, non-cognate aminoacyl-adenylate and misacylated tRNA, are hydrolyzed at the same editing domain, distant from the active site. Here, we present the first example of an aminoacyl-tRNA synthetase (seryl-tRNA synthetase) that naturally lacks an editing domain, but possesses a hydrolytic activity toward non-cognate aminoacyl-adenylates. Our data reveal that tRNA-independent pre-transfer editing may proceed within the enzyme active site without shuttling the non-cognate aminoacyl-adenylate intermediate to the remote editing site.


Subject(s)
Adenosine Monophosphate/chemistry , Escherichia coli Proteins/chemistry , RNA Editing , Saccharomyces cerevisiae Proteins/chemistry , Serine-tRNA Ligase/chemistry , Binding Sites , Cysteine/chemistry , Escherichia coli Proteins/genetics , Hydrolysis , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/genetics , Serine/analogs & derivatives , Serine/chemistry , Serine-tRNA Ligase/genetics , Substrate Specificity , Threonine/chemistry
20.
FEBS J ; 274(11): 2788-99, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17451428

ABSTRACT

The seryl-tRNA synthetase from Saccharomyces cerevisiae interacts with the peroxisome biogenesis-related factor Pex21p. Several deletion mutants of seryl-tRNA synthetase were constructed and inspected for their ability to interact with Pex21p in a yeast two-hybrid assay, allowing mapping of the synthetase domain required for complex assembly. Deletion of the 13 C-terminal amino acids abolished Pex21p binding to seryl-tRNA synthetase. The catalytic parameters of purified truncated seryl-tRNA synthetase, determined in the serylation reaction, were found to be almost identical to those of the native enzyme. In vivo loss of interaction with Pex21p was confirmed in vitro by coaffinity purification. These data indicate that the C-terminally appended domain of yeast seryl-tRNA synthetase does not participate in substrate binding, but instead is required for association with Pex21p. We further determined that Pex21p does not directly bind tRNA, and nor does it possess a tRNA-binding motif, but it instead participates in the formation of a specific ternary complex with seryl-tRNA synthetase and tRNA(Ser), strengthening the interaction of seryl-tRNA synthetase with its cognate tRNA(Ser).


Subject(s)
Carrier Proteins/metabolism , RNA, Transfer, Ser/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Serine-tRNA Ligase/metabolism , Amino Acid Sequence , Electrophoretic Mobility Shift Assay , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL