Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nat Struct Mol Biol ; 31(5): 810-816, 2024 May.
Article in English | MEDLINE | ID: mdl-38538914

ABSTRACT

The frequency of errors upon decoding of messenger RNA by the bacterial ribosome is low, with one misreading event per 1 × 104 codons. In the universal genetic code, the AUN codon box specifies two amino acids, isoleucine and methionine. In bacteria and archaea, decoding specificity of the AUA and AUG codons relies on the wobble avoidance strategy that requires modification of C34 in the anticodon loop of isoleucine transfer RNAIleCAU (tRNAIleCAU). Bacterial tRNAIleCAU with 2-lysylcytidine (lysidine) at the wobble position deciphers AUA while avoiding AUG. Here we report cryo-electron microscopy structures of the Escherichia coli 70S ribosome complexed with elongation factor thermo unstable (EF-Tu) and isoleucine-tRNAIleLAU in the process of decoding AUA and AUG. Lysidine in tRNAIleLAU excludes AUG by promoting the formation of an unusual Hoogsteen purine-pyrimidine nucleobase geometry at the third position of the codon, weakening the interactions with the mRNA and destabilizing the EF-Tu ternary complex. Our findings elucidate the molecular mechanism by which tRNAIleLAU specifically decodes AUA over AUG.


Subject(s)
Cryoelectron Microscopy , Escherichia coli , Models, Molecular , Peptide Elongation Factor Tu , RNA, Transfer, Ile , Ribosomes , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Ribosomes/metabolism , Ribosomes/ultrastructure , Ribosomes/chemistry , RNA, Transfer, Ile/metabolism , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/genetics , Codon/metabolism , Codon/genetics , Anticodon/chemistry , Anticodon/metabolism , Nucleic Acid Conformation , Isoleucine/metabolism , Isoleucine/chemistry , RNA, Messenger/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Lysine/analogs & derivatives , Pyrimidine Nucleosides
2.
Nat Struct Mol Biol ; 31(5): 817-825, 2024 May.
Article in English | MEDLINE | ID: mdl-38538915

ABSTRACT

The anticodon modifications of transfer RNAs (tRNAs) finetune the codon recognition on the ribosome for accurate translation. Bacteria and archaea utilize the modified cytidines, lysidine (L) and agmatidine (agm2C), respectively, in the anticodon of tRNAIle to decipher AUA codon. L and agm2C contain long side chains with polar termini, but their functions remain elusive. Here we report the cryogenic electron microscopy structures of tRNAsIle recognizing the AUA codon on the ribosome. Both modifications interact with the third adenine of the codon via a unique C-A geometry. The side chains extend toward 3' direction of the mRNA, and the polar termini form hydrogen bonds with 2'-OH of the residue 3'-adjacent to the AUA codon. Biochemical analyses demonstrated that AUA decoding is facilitated by the additional interaction between the polar termini of the modified cytidines and 2'-OH of the fourth mRNA residue. We also visualized cyclic N6-threonylcarbamoyladenosine (ct6A), another tRNA modification, and revealed a molecular basis how ct6A contributes to efficient decoding.


Subject(s)
Anticodon , Cryoelectron Microscopy , RNA, Transfer, Ile , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/metabolism , RNA, Transfer, Ile/genetics , Anticodon/chemistry , Anticodon/metabolism , Ribosomes/metabolism , Ribosomes/chemistry , Nucleic Acid Conformation , Models, Molecular , Codon/genetics , Lysine/metabolism , Lysine/chemistry , Lysine/analogs & derivatives , Cytidine/analogs & derivatives , Cytidine/chemistry , Cytidine/metabolism , RNA, Transfer/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , Protein Biosynthesis , Pyrimidine Nucleosides
3.
FEBS J ; 289(16): 4888-4900, 2022 08.
Article in English | MEDLINE | ID: mdl-35122395

ABSTRACT

In almost all eubacteria, the AUA codon is translated by tRNAIle2 bearing lysidine at the wobble position. Lysidine is introduced by tRNAIle lysidine synthetase (TilS) via post-transcriptional modification of the cytidine of tRNAIle2 (CAU). Lactobacillus casei and Lactobacillus plantarum have tilS homologues and tRNAIle2 (CAU) genes. In addition, L. casei also has another tRNAIle2 gene with an UAU anticodon. L. plantarum has a tRNAIle (UAU)-like RNA. Here, we demonstrate that L. casei tRNAIle2 (UAU) is charged with isoleucine by L. casei isoleucyl-tRNA synthetase (IleRS) but not by L. plantarum IleRS, even though the amino acid identity of these two enzymes is over 60%. It has been reported that, in Mycoplasma mobile, which has its tRNAIle2 (UAU) but no tilS homologue, an Arg residue at position 865 of the IleRS is required for recognition of the UAU anticodon. This position is occupied by an Arg also in the IleRSs from both of the Lactobacillus species. Thus, other residues in L. casei, IleRS should also contribute to the recognition of tRNAIle2 (UAU). We found that a chimeric L. casei IleRS in which the N-terminal domain was replaced by the corresponding region of L. plantatarum IleRS has very low aminoacylation activity towards both tRNAIle2 (UAU) and tRNAIle1 (GAU). The A18G mutant had barely detectable aminoacylation activity towards either of the tRNAsIle . However, a double point mutant of A18G and G19N aminoacylated tRNAIle1 (GAU), but not tRNAIle2 (UAU). Our results suggest that, for L. casei IleRS, Ala18 and Gly19 also play a critical role in recognition of tRNAIle2 (UAU).


Subject(s)
Anticodon , Lactobacillales , Anticodon/genetics , Isoleucine/genetics , Isoleucine-tRNA Ligase/chemistry , Lactobacillales/genetics , Lactobacillales/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/genetics , RNA, Transfer, Ile/metabolism
4.
J Am Soc Nephrol ; 33(2): 305-325, 2022 02.
Article in English | MEDLINE | ID: mdl-34607911

ABSTRACT

BACKGROUND: Gitelman syndrome is the most frequent hereditary salt-losing tubulopathy characterized by hypokalemic alkalosis and hypomagnesemia. Gitelman syndrome is caused by biallelic pathogenic variants in SLC12A3, encoding the Na+-Cl- cotransporter (NCC) expressed in the distal convoluted tubule. Pathogenic variants of CLCNKB, HNF1B, FXYD2, or KCNJ10 may result in the same renal phenotype of Gitelman syndrome, as they can lead to reduced NCC activity. For approximately 10 percent of patients with a Gitelman syndrome phenotype, the genotype is unknown. METHODS: We identified mitochondrial DNA (mtDNA) variants in three families with Gitelman-like electrolyte abnormalities, then investigated 156 families for variants in MT-TI and MT-TF, which encode the transfer RNAs for phenylalanine and isoleucine. Mitochondrial respiratory chain function was assessed in patient fibroblasts. Mitochondrial dysfunction was induced in NCC-expressing HEK293 cells to assess the effect on thiazide-sensitive 22Na+ transport. RESULTS: Genetic investigations revealed four mtDNA variants in 13 families: m.591C>T (n=7), m.616T>C (n=1), m.643A>G (n=1) (all in MT-TF), and m.4291T>C (n=4, in MT-TI). Variants were near homoplasmic in affected individuals. All variants were classified as pathogenic, except for m.643A>G, which was classified as a variant of uncertain significance. Importantly, affected members of six families with an MT-TF variant additionally suffered from progressive chronic kidney disease. Dysfunction of oxidative phosphorylation complex IV and reduced maximal mitochondrial respiratory capacity were found in patient fibroblasts. In vitro pharmacological inhibition of complex IV, mimicking the effect of the mtDNA variants, inhibited NCC phosphorylation and NCC-mediated sodium uptake. CONCLUSION: Pathogenic mtDNA variants in MT-TF and MT-TI can cause a Gitelman-like syndrome. Genetic investigation of mtDNA should be considered in patients with unexplained Gitelman syndrome-like tubulopathies.


Subject(s)
DNA, Mitochondrial/genetics , Gitelman Syndrome/genetics , Mutation , Adolescent , Adult , Aged , Base Sequence , Child , Child, Preschool , Female , Genotype , Gitelman Syndrome/metabolism , Gitelman Syndrome/pathology , HEK293 Cells , Humans , Infant , Kidney/metabolism , Kidney/ultrastructure , Male , Middle Aged , Mitochondria/metabolism , Models, Biological , Nucleic Acid Conformation , Pedigree , Phenotype , Polymorphism, Single Nucleotide , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/genetics , RNA, Transfer, Phe/chemistry , RNA, Transfer, Phe/genetics , Solute Carrier Family 12, Member 3/genetics , Young Adult
5.
J Phys Chem Lett ; 11(10): 4001-4007, 2020 May 21.
Article in English | MEDLINE | ID: mdl-32354218

ABSTRACT

Living systems maintain a high fidelity in information processing through kinetic proofreading, a mechanism for preferentially removing incorrect substrates at the cost of energy dissipation and slower speed. Proofreading mechanisms must balance their demand for higher speed, fewer errors, and lower dissipation, but it is unclear how rates of individual reaction steps are evolutionarily tuned to balance these needs, especially when multiple proofreading mechanisms are present. Here, using a discrete-state stochastic model, we analyze the optimization strategies in Escherichia coli isoleucyl-tRNA synthetase. Surprisingly, this enzyme adopts an economic proofreading strategy and improves speed and dissipation as long as the error is tolerable. Through global parameter sampling, we reveal a fundamental dissipation-error relation that bounds the enzyme's optimal performance and explains the importance of the post-transfer editing mechanism. The proximity of native system parameters to this bound demonstrates the importance of energy dissipation as an evolutionary force affecting fitness.


Subject(s)
RNA, Transfer, Ile/chemical synthesis , Aminoacylation , Models, Molecular , RNA, Transfer, Ile/chemistry
6.
Nat Struct Mol Biol ; 26(12): 1114-1122, 2019 12.
Article in English | MEDLINE | ID: mdl-31792448

ABSTRACT

T-box riboswitches are modular bacterial noncoding RNAs that sense and regulate amino acid availability through direct interactions with tRNAs. Between the 5' anticodon-binding stem I domain and the 3' amino acid sensing domains of most T-boxes lies the stem II domain of unknown structure and function. Here, we report a 2.8-Å cocrystal structure of the Nocardia farcinica ileS T-box in complex with its cognate tRNAIle. The structure reveals a perpendicularly arranged ultrashort stem I containing a K-turn and an elongated stem II bearing an S-turn. Both stems rest against a compact pseudoknot, dock via an extended ribose zipper and jointly create a binding groove specific to the anticodon of its cognate tRNA. Contrary to proposed distal contacts to the tRNA elbow region, stem II locally reinforces the codon-anticodon interactions between stem I and tRNA, achieving low-nanomolar affinity. This study illustrates how mRNA junctions can create specific binding sites for interacting RNAs of prescribed sequence and structure.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Isoleucine-tRNA Ligase/genetics , Nocardia/genetics , Nucleotide Motifs , RNA, Bacterial/chemistry , RNA, Transfer, Ile/chemistry , Riboswitch/genetics , Binding Sites , Crystallography, X-Ray , Models, Molecular , RNA, Bacterial/metabolism , RNA, Transfer, Ile/metabolism , Structure-Activity Relationship
7.
Mol Med Rep ; 18(6): 5159-5165, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30272361

ABSTRACT

Mutations in the mitochondrial genome have been identified to be associated with hearing loss. The aim of the present study was to investigate the role of mitochondrial DNA (mtDNA) variants in a Chinese family with hearing loss. Polymerase chain reaction (PCR)­Sanger sequencing was used to screen the mtDNA variants and nuclear genes [gap junction protein ß2 (GJB2) and transfer (t)RNA 5­methylaminomethyle­2­thiouridylate methyltransferase (TRMU)]; in addition, the mtDNA copy number was determined by quantitative PCR. The present study characterized the molecular features of a Chinese family with maternally­inherited hearing loss and identified mtDNA A1555G and tRNAIle A4317G mutations. The A4317G mutation was localized at the TΨC arm of tRNAIle (position 59) and created a novel base­pairing (G59­C54), which may alter the secondary structure of the tRNA. In addition, patients carrying the A4317G mutation exhibited a lower mtDNA copy number compared with the controls, suggesting that this mutation may cause mitochondrial dysfunction that is responsible for the deafness. However, no functional variants in the GJB2 and TRMU genes were detected. mtDNA A1555G and A4317G mutations may contribute to the clinical manifestation of hearing loss in this family.


Subject(s)
Genes, Mitochondrial , Genetic Association Studies , Genetic Predisposition to Disease , Hearing Loss/genetics , Mutation , RNA, Transfer, Ile/genetics , Adult , Asian People/genetics , China , Connexin 26 , Connexins/genetics , DNA Copy Number Variations , DNA, Mitochondrial , Genome, Mitochondrial , Genotype , Hearing Loss/diagnosis , Humans , Middle Aged , Mitochondrial Proteins/genetics , Pedigree , Phenotype , Phylogeny , RNA, Transfer, Ile/chemistry , Severity of Illness Index , tRNA Methyltransferases/genetics
8.
Biochem Biophys Res Commun ; 497(4): 1049-1054, 2018 03 18.
Article in English | MEDLINE | ID: mdl-29481798

ABSTRACT

Mitochondria are essential for early cardiac development and impaired mitochondrial function was described associated with heart diseases such as hypertrophic or dilated mitochondrial cardiomyopathy. In this study, we report a family including two individuals with severe dilated mitochondrial cardiomyopathy. The whole mitochondrial genome screening showed the presence of several variations and a novel homoplasmic mutation m.4318-4322delC in the MT-TI gene shared by the two patients and their mother and leading to a disruption of the tRNAIle secondary structure. In addition, a mitochondrial depletion was present in blood leucocyte of the two affected brother whereas a de novo heteroplasmic multiple deletion in the major arc of mtDNA was present in blood leucocyte and mucosa of only one of them. These deletions in the major arc of the mtDNA resulted to the loss of several protein-encoding genes and also some tRNA genes. The mtDNA deletion and depletion could result to an impairment of the oxidative phosphorylation and energy metabolism in the respiratory chain in the studied patients. Our report is the first description of a family with severe lethal dilated mitochondrial cardiomyopathy and presenting several mtDNA abnormalities including punctual mutation, deletion and depletion.


Subject(s)
Cardiomyopathy, Dilated/genetics , DNA, Mitochondrial/genetics , Mitochondria, Heart/genetics , Mutation , RNA, Transfer, Ile/genetics , Energy Metabolism , Family , Genome, Mitochondrial/genetics , Humans , Infant , Infant, Newborn , Mitochondria/genetics , Oxidative Phosphorylation , RNA, Transfer, Ile/chemistry , Sequence Deletion
9.
Mol Cell ; 63(1): 110-24, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27345152

ABSTRACT

The Lupus autoantigen La is an RNA-binding protein that stabilizes RNA polymerase III (Pol III) transcripts and supports RNA folding and has in addition been implicated in the mammalian microRNA (miRNA) pathway. Here, we have analyzed effects of La depletion on Argonaute (Ago)-bound small RNAs in human cells. We find that in the absence of La, distinct tRNA fragments are loaded into Ago proteins. Thus, La functions as gatekeeper ensuring correct tRNA maturation and protecting the miRNA pathway from potentially functional tRNA fragments. However, one specific isoleucin pre-tRNA produces both a functional tRNA and a miRNA even when La is present. We demonstrate that the fully complementary 5' leader and 3' trailer of the pre-tRNA-Ile form a double-stranded RNA molecule that has low affinity to La. Instead, Exportin-5 (Xpo5) recognizes it as miRNA precursor and transports it into the cytoplasm for Dicer processing and Ago loading.


Subject(s)
Autoantigens/metabolism , MicroRNAs/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Transfer, Ile/metabolism , Ribonucleoproteins/metabolism , A549 Cells , Argonaute Proteins/metabolism , Autoantigens/genetics , Binding Sites , DEAD-box RNA Helicases/metabolism , HEK293 Cells , HeLa Cells , Hep G2 Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Humans , Karyopherins/metabolism , MCF-7 Cells , MicroRNAs/genetics , Nucleic Acid Conformation , Protein Binding , RNA Interference , RNA Polymerase III/metabolism , RNA Precursors/chemistry , RNA Precursors/genetics , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Ribonuclease III/metabolism , Ribonucleoproteins/genetics , Structure-Activity Relationship , Transfection , SS-B Antigen
10.
Plant Physiol ; 169(1): 627-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26152711

ABSTRACT

Plastid gene expression is crucial for organelle function, but the factors that control it are still largely unclear. Members of the so-called mitochondrial transcription termination factor (mTERF) family are found in metazoans and plants and regulate organellar gene expression at different levels. Arabidopsis (Arabidopsis thaliana) mTERF6 is localized in chloroplasts and mitochondria, and its knockout perturbs plastid development and results in seedling lethality. In the leaky mterf6-1 mutant, a defect in photosynthesis is associated with reduced levels of photosystem subunits, although corresponding messenger RNA levels are unaffected, whereas translational capacity and maturation of chloroplast ribosomal RNAs (rRNAs) are perturbed in mterf6-1 mutants. Bacterial one-hybrid screening, electrophoretic mobility shift assays, and coimmunoprecipitation experiments reveal a specific interaction between mTERF6 and an RNA sequence in the chloroplast isoleucine transfer RNA gene (trnI.2) located in the rRNA operon. In vitro, recombinant mTERF6 bound to its plastid DNA target site can terminate transcription. At present, it is unclear whether disturbed rRNA maturation is a primary or secondary defect. However, it is clear that mTERF6 is required for the maturation of trnI.2. This points to an additional function of mTERFs.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Chloroplasts/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , RNA, Transfer, Ile/metabolism , Transcription Termination, Genetic , 5' Untranslated Regions/genetics , Aminoacylation , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , DNA, Bacterial/genetics , Gene Expression Regulation, Plant , Genetic Loci , Mitochondrial Proteins/genetics , Molecular Sequence Data , Mutagenesis, Insertional/genetics , Mutation , Phenotype , Photosynthesis , Protein Binding , Protein Transport , RNA Processing, Post-Transcriptional , RNA, Ribosomal/genetics , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/genetics , Ribosomes/metabolism , Seedlings/metabolism , Seeds/ultrastructure
11.
FEBS Lett ; 589(16): 2124-30, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26143376

ABSTRACT

Protozoan mitochondrial tRNAs (mt-tRNAs) are repaired by a process known as 5'-editing. Mt-tRNA sequencing revealed organism-specific patterns of editing G-U base pairs, wherein some species remove G-U base pairs during 5'-editing, while others retain G-U pairs in the edited tRNA. We tested whether 3'-5' polymerases that catalyze the repair step of 5'-editing exhibit organism-specific preferences that explain the treatment of G-U base pairs. Biochemical and kinetic approaches revealed that a 3'-5' polymerase from Acanthamoeba castellanii tolerates G-U wobble pairs in editing substrates much more readily than several other enzymes, consistent with its biological pattern of editing.


Subject(s)
Acanthamoeba castellanii/enzymology , Mitochondria/enzymology , Protozoan Proteins/metabolism , RNA Editing , RNA Nucleotidyltransferases/metabolism , RNA, Protozoan/metabolism , RNA, Transfer/metabolism , Acanthamoeba castellanii/metabolism , Dictyostelium/enzymology , Dictyostelium/metabolism , Electrophoresis, Polyacrylamide Gel , Isoenzymes/genetics , Isoenzymes/metabolism , Isotope Labeling , Kinetics , Mitochondria/metabolism , Models, Molecular , Phosphorus Radioisotopes , Protozoan Proteins/genetics , RNA Nucleotidyltransferases/genetics , RNA, Protozoan/chemistry , RNA, Transfer/chemistry , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/metabolism , Recombinant Proteins/metabolism , Species Specificity , Substrate Specificity
12.
Biochemistry ; 53(39): 6189-98, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25207837

ABSTRACT

The accurate expression of genetic information relies on the fidelity of amino acid-tRNA coupling by aminoacyl-tRNA synthetases (aaRS). When the specificity against structurally similar noncognate amino acids in the synthetic reaction does not support a threshold fidelity level for translation, the aaRS employ intrinsic hydrolytic editing to correct errors in aminoacylation. Escherichia coli isoleucyl-tRNA synthetase (EcIleRS) is a class I aaRS that is notable for its use of tRNA-dependent pretransfer editing to hydrolyze noncognate valyl-adenylate prior to aminoacyl-tRNA formation. On the basis of the finding that IleRS possessing an inactivated post-transfer editing domain is still capable of robust tRNA-dependent editing, we have recently proposed that the pretransfer editing activity resides within the synthetic site. Here we apply an improved methodology that allows quantitation of the AMP fraction that arises particularly from tRNA-dependent aa-AMP hydrolysis. By this approach, we demonstrate that tRNA-dependent pretransfer editing accounts for nearly one-third of the total proofreading by EcIleRS and that a highly conserved tyrosine within the synthetic site modulates both editing and aminoacylation. Therefore, synthesis of aminoacyl-tRNA and hydrolysis of aminoacyl-adenylates employ overlapping amino acid determinants. We suggest that this overlap hindered the evolution of synthetic site-based pretransfer editing as the predominant proofreading pathway, because that activity is difficult to accommodate in the context of efficient aminoacyl-tRNA synthesis. Instead, the acquisition of a spatially separate domain dedicated to post-transfer editing alone allowed for the development of a powerful deacylation machinery that effectively competes with dissociation of misacylated tRNAs.


Subject(s)
Escherichia coli Proteins/metabolism , Isoleucine-tRNA Ligase/metabolism , RNA Editing , RNA, Transfer/metabolism , Amino Acid Sequence , Binding Sites/genetics , Biosynthetic Pathways/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Hydrolysis , Isoleucine-tRNA Ligase/chemistry , Isoleucine-tRNA Ligase/genetics , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/genetics , RNA, Transfer, Ile/metabolism , Sequence Homology, Amino Acid , Transfer RNA Aminoacylation , Tyrosine/genetics , Tyrosine/metabolism
13.
Nucleic Acids Res ; 42(3): 1904-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24194599

ABSTRACT

Translation of the isoleucine codon AUA in most prokaryotes requires a modified C (lysidine or agmatidine) at the wobble position of tRNA2(Ile) to base pair specifically with the A of the AUA codon but not with the G of AUG. Recently, a Bacillus subtilis strain was isolated in which the essential gene encoding tRNA(Ile)-lysidine synthetase was deleted for the first time. In such a strain, C34 at the wobble position of tRNA2(Ile) is expected to remain unmodified and cells depend on a mutant suppressor tRNA derived from tRNA1(Ile), in which G34 has been changed to U34. An important question, therefore, is how U34 base pairs with A without also base pairing with G. Here, we show (i) that unlike U34 at the wobble position of all B. subtilis tRNAs of known sequence, U34 in the mutant tRNA is not modified, and (ii) that the mutant tRNA binds strongly to the AUA codon on B. subtilis ribosomes but only weakly to AUG. These in vitro data explain why the suppressor strain displays only a low level of misreading AUG codons in vivo and, as shown here, grows at a rate comparable to that of the wild-type strain.


Subject(s)
Bacillus subtilis/genetics , Codon , Isoleucine/metabolism , Protein Biosynthesis , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/metabolism , Amino Acyl-tRNA Synthetases/genetics , Bacillus subtilis/growth & development , Gene Deletion , Phenotype , RNA, Transfer, Ile/isolation & purification , Ribosomes/metabolism , Transfer RNA Aminoacylation
14.
RNA ; 20(2): 177-88, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344322

ABSTRACT

Most archaea and bacteria use a modified C in the anticodon wobble position of isoleucine tRNA to base pair with A but not with G of the mRNA. This allows the tRNA to read the isoleucine codon AUA without also reading the methionine codon AUG. To understand why a modified C, and not U or modified U, is used to base pair with A, we mutated the C34 in the anticodon of Haloarcula marismortui isoleucine tRNA (tRNA2(Ile)) to U, expressed the mutant tRNA in Haloferax volcanii, and purified and analyzed the tRNA. Ribosome binding experiments show that although the wild-type tRNA2(Ile) binds exclusively to the isoleucine codon AUA, the mutant tRNA binds not only to AUA but also to AUU, another isoleucine codon, and to AUG, a methionine codon. The G34 to U mutant in the anticodon of another H. marismortui isoleucine tRNA species showed similar codon binding properties. Binding of the mutant tRNA to AUG could lead to misreading of the AUG codon and insertion of isoleucine in place of methionine. This result would explain why most archaea and bacteria do not normally use U or a modified U in the anticodon wobble position of isoleucine tRNA for reading the codon AUA. Biochemical and mass spectrometric analyses of the mutant tRNAs have led to the discovery of a new modified nucleoside, 5-cyanomethyl U in the anticodon wobble position of the mutant tRNAs. 5-Cyanomethyl U is present in total tRNAs from euryarchaea but not in crenarchaea, eubacteria, or eukaryotes.


Subject(s)
Anticodon/genetics , Haloarcula marismortui/genetics , RNA, Archaeal/genetics , RNA, Transfer, Ile/genetics , Uridine/analogs & derivatives , Base Pairing , Base Sequence , Codon/genetics , Escherichia coli/genetics , Haloferax/genetics , Molecular Structure , Point Mutation , RNA, Archaeal/chemistry , RNA, Archaeal/metabolism , RNA, Bacterial/genetics , RNA, Fungal/genetics , RNA, Transfer, Ile/chemistry , RNA, Transfer, Ile/metabolism , Ribosomes/chemistry , Saccharomyces cerevisiae/genetics , Sulfolobus/genetics , Transfer RNA Aminoacylation , Uridine/chemistry , Uridine/genetics
15.
Biochem Biophys Res Commun ; 441(4): 999-1004, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24239547

ABSTRACT

Polyamines, putrescine, spermidine (SPD), and spermine are closely linked to cell growth, and highly regulate the levels of transcription, translation and protein turnover. We propose that SPD stimulates the formation of Ile-tRNA(Ile) by inducing a selective structural change of the G-G mismatch containing acceptor stem in tRNA(Ile). Here, we provide insight into how SPD recognizes and stabilizes the G-G mismatch containing acceptor stem in tRNA(Ile) with molecular modeling (MM), density functional theory (DFT) calculations, and molecular dynamics (MD) simulations. The results of the MM and DFT calculations indicate that the negatively charged region of the G-G mismatch containing acceptor stem in tRNA(Ile) is preferentially recognized by positively charged SPD. In addition, MD simulations indicate that all of the positively charged amino groups of SPD under physiological conditions (N1(NH3(+)), N5(NH2(+)), and N10(NH3(+)) could form hydrogen bonds with tRNA(Ile) and trigger the SPD-induced stabilization and structural change of the G-G mismatch containing acceptor stem in tRNA(Ile). Thus, this approach should be useful for determining the preferential binding site and appropriate binding mode of polyamines on tRNA(Ile).


Subject(s)
Computer Simulation , Models, Molecular , RNA, Transfer, Ile/chemistry , Spermidine/chemistry , Base Pair Mismatch , Binding Sites , Hydrogen Bonding , Molecular Dynamics Simulation , RNA, Transfer, Ile/genetics
16.
PLoS One ; 8(6): e66112, 2013.
Article in English | MEDLINE | ID: mdl-23840412

ABSTRACT

Proofreading/editing in protein synthesis is essential for accurate translation of information from the genetic code. In this article we present a theoretical investigation of efficiency of a kinetic proofreading mechanism that employs hydrolysis of the wrong substrate as the discriminatory step in enzyme catalytic reactions. We consider aminoacylation of tRNA(Ile) which is a crucial step in protein synthesis and for which experimental results are now available. We present an augmented kinetic scheme and then employ methods of stochastic simulation algorithm to obtain time dependent concentrations of different substances involved in the reaction and their rates of formation. We obtain the rates of product formation and ATP hydrolysis for both correct and wrong substrates (isoleucine and valine in our case, respectively), in single molecular enzyme as well as ensemble enzyme kinetics. The present theoretical scheme correctly reproduces (i) the amplitude of the discrimination factor in the overall rates between isoleucine and valine which is obtained as (1.8×10(2)).(4.33×10(2)) = 7.8×10(4), (ii) the rates of ATP hydrolysis for both Ile and Val at different substrate concentrations in the aminoacylation of tRNA(Ile). The present study shows a non-michaelis type dependence of rate of reaction on tRNA(Ile) concentration in case of valine. The overall editing in steady state is found to be independent of amino acid concentration. Interestingly, the computed ATP hydrolysis rate for valine at high substrate concentration is same as the rate of formation of Ile-tRNA(Ile) whereas at intermediate substrate concentration the ATP hydrolysis rate is relatively low. We find that the presence of additional editing domain in class I editing enzyme makes the kinetic proofreading more efficient through enhanced hydrolysis of wrong product at the editing CP1 domain.


Subject(s)
Bacterial Proteins/chemistry , Isoleucine-tRNA Ligase/chemistry , Models, Chemical , RNA, Transfer, Ile/chemistry , Water/chemistry , Adenosine Monophosphate/chemistry , Adenosine Triphosphate/chemistry , Algorithms , Computer Simulation , Hydrolysis , Isoleucine/chemistry , Kinetics , Stochastic Processes , Transfer RNA Aminoacylation , Valine/chemistry
17.
Nat Struct Mol Biol ; 20(5): 641-3, 2013 May.
Article in English | MEDLINE | ID: mdl-23542153

ABSTRACT

Decoding of the AUA isoleucine codon in bacteria and archaea requires modification of a C in the anticodon wobble position of the isoleucine tRNA. Here, we report the crystal structure of the archaeal tRNA2(Ile), which contains the modification agmatidine in its anticodon, in complex with the AUA codon on the 70S ribosome. The structure illustrates how agmatidine confers codon specificity for AUA over AUG.


Subject(s)
Archaea/genetics , Codon , Isoleucine/genetics , Protein Biosynthesis , RNA, Transfer, Ile/chemistry , Ribosomes/chemistry , Archaea/chemistry , Archaea/metabolism , Isoleucine/metabolism , Models, Molecular , Nucleic Acid Conformation , RNA, Transfer, Ile/metabolism , Ribosomes/metabolism
18.
Nucleic Acids Res ; 41(4): 2621-31, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23295668

ABSTRACT

Deciphering the genetic code is a fundamental process in all living organisms. In many bacteria, AUA codons are deciphered by tRNA(Ile2) bearing lysidine (L) at the wobble position. L is a modified cytidine introduced post-transcriptionally by tRNA(Ile)-lysidine synthetase (TilS). Some bacteria, including Mycoplasma mobile, do not carry the tilS gene, indicating that they have established a different system to decode AUA codons. In this study, tRNA(Ile2) has been isolated from M. mobile and was found to contain a UAU anticodon without any modification. Mycoplasma mobile isoleucyl-tRNA synthetase (IleRS) recognized the UAU anticodon, whereas Escherichia coli IleRS did not efficiently aminoacylate tRNA(Ile2)(UAU). In M. mobile IleRS, a single Arg residue at position 865 was critical for specificity for the UAU anticodon and, when the corresponding site (W905) in E. coli IleRS was substituted with Arg, the W905R mutant efficiently aminoacylated tRNA with UAU anticodon. Mycoplasma mobile tRNA(Ile2) cannot distinguish between AUA and AUG codon on E. coli ribosome. However, on M. mobile ribosome, M. mobile tRNA(Ile2)(UAU) specifically recognized AUA codon, and not AUG codon, suggesting M. mobile ribosome has a property that prevents misreading of AUG codon. These findings provide an insight into the evolutionary reorganization of the AUA decoding system.


Subject(s)
Anticodon/chemistry , Codon/chemistry , Isoleucine-tRNA Ligase/chemistry , Mycoplasma/genetics , RNA, Transfer, Ile/chemistry , Amino Acid Sequence , Arginine/chemistry , Isoleucine-tRNA Ligase/metabolism , Kinetics , Molecular Sequence Data , Mycoplasma/enzymology , RNA, Transfer, Ile/isolation & purification , RNA, Transfer, Ile/metabolism , Ribosomes/metabolism , Sequence Alignment
20.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 11): 1414-6, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22102245

ABSTRACT

The cytidine at the first anticodon position of archaeal tRNA(Ile2), which decodes the isoleucine AUA codon, is modified to 2-agmatinylcytidine (agm(2)C) to guarantee the fidelity of protein biosynthesis. This post-transcriptional modification is catalyzed by tRNA(Ile)-agm(2)C synthetase (TiaS) using ATP and agmatine as substrates. Archaeoglobus fulgidus TiaS was overexpressed in Escherichia coli cells and purified. tRNA(Ile2) was prepared by in vitro transcription with T7 RNA polymerase. TiaS was cocrystallized with both tRNA(Ile2) and ATP by the vapour-diffusion method. The crystals of the TiaS-tRNA(Ile2)-ATP complex diffracted to 2.9 Å resolution using synchrotron radiation at the Photon Factory. The crystals belonged to the primitive hexagonal space group P3(2)21, with unit-cell parameters a = b = 131.1, c = 86.6 Å. The asymmetric unit is expected to contain one TiaS-tRNA(Ile2)-ATP complex, with a Matthews coefficient of 2.8 Å(3) Da(-1) and a solvent content of 61%.


Subject(s)
Adenosine Triphosphate/chemistry , Archaeoglobales/enzymology , Isoleucine-tRNA Ligase/chemistry , RNA, Transfer, Ile/chemistry , Adenosine Triphosphate/metabolism , Crystallization , Crystallography, X-Ray , Isoleucine-tRNA Ligase/metabolism , Protein Binding , RNA, Transfer, Ile/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...