Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Nucleic Acids Res ; 51(8): 4000-4011, 2023 05 08.
Article in English | MEDLINE | ID: mdl-36912079

ABSTRACT

Two types of glycyl-tRNA synthetase (GlyRS) are known, the α2 and the α2ß2 GlyRSs. Both types of synthetase employ a class II catalytic domain to aminoacylate tRNAGly. In plastids and some bacteria, the α and ß subunits are fused and are designated as (αß)2 GlyRSs. While the tRNA recognition and aminoacylation mechanisms are well understood for α2 GlyRSs, little is known about the mechanisms for α2ß2/(αß)2 GlyRSs. Here we describe structures of the (αß)2 GlyRS from Oryza sativa chloroplast by itself and in complex with cognate tRNAGly. The set of structures reveals that the U-shaped ß half of the synthetase selects the tRNA in a two-step manner. In the first step, the synthetase engages the elbow and the anticodon base C35 of the tRNA. In the second step, the tRNA has rotated ∼9° toward the catalytic centre. The synthetase probes the tRNA for the presence of anticodon base C36 and discriminator base C73. This intricate mechanism enables the tRNA to access the active site of the synthetase from a direction opposite to that of most other class II synthetases.


Subject(s)
Glycine-tRNA Ligase , Glycine-tRNA Ligase/genetics , Anticodon , RNA, Transfer, Gly/chemistry , RNA, Transfer , Plastids
2.
Nucleic Acids Res ; 49(22): 13045-13061, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34871455

ABSTRACT

Dnmt2, a member of the DNA methyltransferase superfamily, catalyzes the formation of 5-methylcytosine at position 38 in the anticodon loop of tRNAs. Dnmt2 regulates many cellular biological processes, especially the production of tRNA-derived fragments and intergenerational transmission of paternal metabolic disorders to offspring. Moreover, Dnmt2 is closely related to human cancers. The tRNA substrates of mammalian Dnmt2s are mainly detected using bisulfite sequencing; however, we lack supporting biochemical data concerning their substrate specificity or recognition mechanism. Here, we deciphered the tRNA substrates of human DNMT2 (hDNMT2) as tRNAAsp(GUC), tRNAGly(GCC) and tRNAVal(AAC). Intriguingly, for tRNAAsp(GUC) and tRNAGly(GCC), G34 is the discriminator element; whereas for tRNAVal(AAC), the inosine modification at position 34 (I34), which is formed by the ADAT2/3 complex, is the prerequisite for hDNMT2 recognition. We showed that the C32U33(G/I)34N35 (C/U)36A37C38 motif in the anticodon loop, U11:A24 in the D stem, and the correct size of the variable loop are required for Dnmt2 recognition of substrate tRNAs. Furthermore, mammalian Dnmt2s possess a conserved tRNA recognition mechanism.


Subject(s)
5-Methylcytosine/metabolism , Anticodon/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , RNA, Transfer/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Anticodon/genetics , Base Sequence , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/genetics , HEK293 Cells , HeLa Cells , Humans , Inosine/metabolism , Mice , Models, Molecular , NIH 3T3 Cells , Nucleic Acid Conformation , Protein Binding , RNA, Transfer/chemistry , RNA, Transfer/genetics , RNA, Transfer, Asp/chemistry , RNA, Transfer, Asp/genetics , RNA, Transfer, Asp/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/genetics , RNA, Transfer, Gly/metabolism , RNA, Transfer, Val/chemistry , RNA, Transfer, Val/genetics , RNA, Transfer, Val/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Substrate Specificity
3.
Nucleic Acids Res ; 49(17): 10106-10119, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34390350

ABSTRACT

AaRSs (aminoacyl-tRNA synthetases) group into two ten-member classes throughout evolution, with unique active site architectures defining each class. Most are monomers or homodimers but, for no apparent reason, many bacterial GlyRSs are heterotetramers consisting of two catalytic α-subunits and two tRNA-binding ß-subunits. The heterotetrameric GlyRS from Escherichia coli (EcGlyRS) was historically tested whether its α- and ß-polypeptides, which are encoded by a single mRNA with a gap of three in-frame codons, are replaceable by a single chain. Here, an unprecedented X-shaped structure of EcGlyRS shows wide separation of the abutting chain termini seen in the coding sequences, suggesting strong pressure to avoid a single polypeptide format. The structure of the five-domain ß-subunit is unique across all aaRSs in current databases, and structural analyses suggest these domains play different functions on α-subunit binding, ATP coordination and tRNA recognition. Moreover, the X-shaped architecture of EcGlyRS largely fits with a model for how two classes of tRNA synthetases arose, according to whether enzymes from opposite classes can simultaneously co-dock onto separate faces of the same tRNA acceptor stem. While heterotetrameric GlyRS remains the last structurally uncharacterized member of aaRSs, our study contributes to a better understanding of this ancient and essential enzyme family.


Subject(s)
Catalytic Domain/genetics , Escherichia coli/genetics , Glycine-tRNA Ligase/genetics , RNA, Transfer, Gly/chemistry , Adenosine Triphosphate/metabolism , Crystallography, X-Ray , Glycine/chemistry , Models, Molecular , RNA, Transfer, Gly/genetics
4.
Leuk Res ; 87: 106234, 2019 12.
Article in English | MEDLINE | ID: mdl-31669784

ABSTRACT

Chronic lymphocytic leukemia (CLL) is one of the most common types of leukemia in adults. Several studies have identified various prognostic biomarkers in CLL. In this study, we investigated the potential value of an internal fragment of the tRNAs bearing the Glycine anticodon CCC (i-tRF-GlyCCC), which is a small non-coding RNA, as a prognostic and screening biomarker in CLL. For this purpose, blood samples were collected from 90 CLL patients and 43 non-leukemic blood donors. Peripheral blood mononuclear cells (PBMCs) were isolated, total RNA was extracted and in-vitro polyadenylated, and first-strand cDNA was synthesized using an oligo-dT-adaptor primer. A real-time quantitative PCR assay was developed and applied for the quantification of i-tRF-GlyCCC in our samples. The biostatistical analysis revealed that i-tRF-GlyCCC levels are significantly lower in PBMCs of CLL patients, compared to PBMCs of non-leukemic controls, and that i-tRF-GlyCCC could be considered as a screening biomarker. Kaplan-Meier overall survival (OS) analysis revealed reduced OS for CLL patients with positive i-tRF-GlyCCC expression (P = 0.001). Multivariate Cox regression confirmed its independent unfavorable prognostic power with regard to OS. In conclusion, i-tRF-GlyCCC may constitute a promising molecular biomarker in CLL, for screening and prognostic purposes.


Subject(s)
Biomarkers, Tumor/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , RNA, Transfer, Gly/genetics , Real-Time Polymerase Chain Reaction/methods , Aged , Aged, 80 and over , Base Sequence/physiology , Biomarkers, Tumor/chemistry , Case-Control Studies , Cells, Cultured , Cohort Studies , Female , Follow-Up Studies , Humans , Inventions , K562 Cells , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Mass Screening/methods , Middle Aged , Prognosis , RNA, Transfer, Gly/chemistry , Real-Time Polymerase Chain Reaction/trends , Sequence Analysis, RNA
5.
Nucleic Acids Res ; 46(17): 9081-9093, 2018 09 28.
Article in English | MEDLINE | ID: mdl-29893896

ABSTRACT

We have previously shown that 5' halves from tRNAGlyGCC and tRNAGluCUC are the most enriched small RNAs in the extracellular space of human cell lines, and especially in the non-vesicular fraction. Extracellular RNAs are believed to require protection by either encapsulation in vesicles or ribonucleoprotein complex formation. However, deproteinization of non-vesicular tRNA halves does not affect their retention in size-exclusion chromatography. Thus, we considered alternative explanations for their extracellular stability. In-silico analysis of the sequence of these tRNA-derived fragments showed that tRNAGly 5' halves can form homodimers or heterodimers with tRNAGlu 5' halves. This capacity is virtually unique to glycine tRNAs. By analyzing synthetic oligonucleotides by size exclusion chromatography, we provide evidence that dimerization is possible in vitro. tRNA halves with single point substitutions preventing dimerization are degraded faster both in controlled nuclease digestion assays and after transfection in cells, showing that dimerization can stabilize tRNA halves against the action of cellular nucleases. Finally, we give evidence supporting dimerization of endogenous tRNAGlyGCC 5' halves inside cells. Considering recent reports have shown that 5' tRNA halves from Ala and Cys can form tetramers, our results highlight RNA intermolecular structures as a new layer of complexity in the biology of tRNA-derived fragments.


Subject(s)
Dimerization , RNA Stability , RNA, Transfer, Glu/metabolism , RNA, Transfer, Gly/metabolism , Ribonucleases/metabolism , 5' Flanking Region , Base Sequence , Glutamic Acid/metabolism , Glycine/metabolism , Humans , MCF-7 Cells , Nucleic Acid Conformation , RNA, Transfer, Glu/chemistry , RNA, Transfer, Gly/chemistry
6.
Nucleic Acids Res ; 45(13): 8079-8090, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28531275

ABSTRACT

A T-box regulator or riboswitch actively monitors the levels of charged/uncharged tRNA and participates in amino acid homeostasis by regulating genes involved in their utilization or biosynthesis. It has an aptamer domain for cognate tRNA recognition and an expression platform to sense the charge state and modulate gene expression. These two conserved domains are connected by a variable linker that harbors additional secondary structural elements, such as Stem III. The structural basis for specific tRNA binding is known, but the structural basis for charge sensing and the role of other elements remains elusive. To gain new structural insights on the T-box mechanism, a molecular envelope was calculated from small angle X-ray scattering data for the Bacillus subtilis glyQS T-box riboswitch in complex with an uncharged tRNAGly. A structural model of an anti-terminated glyQS T-box in complex with its cognate tRNAGly was derived based on the molecular envelope. It shows the location and relative orientation of various secondary structural elements. The model was validated by comparing the envelopes of the wild-type complex and two variants. The structural model suggests that in addition to a possible regulatory role, Stem III could aid in preferential stabilization of the T-box anti-terminated state allowing read-through of regulated genes.


Subject(s)
RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/metabolism , Riboswitch/genetics , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Base Sequence , Models, Molecular , Nucleic Acid Conformation , RNA, Bacterial/genetics , RNA, Transfer, Gly/genetics , Scattering, Small Angle , X-Ray Diffraction
7.
J Mol Biol ; 428(18): 3603-14, 2016 09 11.
Article in English | MEDLINE | ID: mdl-27261259

ABSTRACT

Aminoacyl-tRNA synthetases are essential components of the protein translational machinery in all living species, among which the human glycyl-tRNA synthetase (hGlyRS) is of great research interest because of its unique species-specific aminoacylation properties and noncanonical roles in the Charcot-Marie-Tooth neurological disease. However, the molecular mechanisms of how the enzyme carries out its classical and alternative functions are not well understood. Here, we report a complex structure of the wild-type hGlyRS bound with tRNA(Gly) at 2.95Å. In the complex, the flexible Whep-TRS domain is visible in one of the subunits of the enzyme dimer, and the tRNA molecule is also completely resolved. At the active site, a glycyl-AMP molecule is synthesized and is waiting for the transfer of the glycyl moiety to occur. This cocrystal structure provides us with new details about the recognition mechanism in the intermediate stage during glycylation, which was not well elucidated in the previous crystal structures where the inhibitor AMPPNP was used for crystallization. More importantly, the structural and biochemical work conducted in the current and previous studies allows us to build a model of the full-length hGlyRS in complex with tRNA(Gly), which greatly helps us to understand the roles that insertions and the Whep-TRS domain play in the tRNA-binding process. Finally, through structure comparison with other class II aminoacyl-tRNA synthetases bound with their tRNA substrates, we found some commonalities of the aminoacylation mechanism between these enzymes.


Subject(s)
Glycine-tRNA Ligase/chemistry , Glycine-tRNA Ligase/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/metabolism , Crystallography, X-Ray , Humans , Models, Biological , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation
8.
PLoS Biol ; 14(5): e1002465, 2016 05.
Article in English | MEDLINE | ID: mdl-27224426

ABSTRACT

D-aminoacyl-tRNA deacylase (DTD) removes D-amino acids mischarged on tRNAs and is thus implicated in enforcing homochirality in proteins. Previously, we proposed that selective capture of D-aminoacyl-tRNA by DTD's invariant, cross-subunit Gly-cisPro motif forms the mechanistic basis for its enantioselectivity. We now show, using nuclear magnetic resonance (NMR) spectroscopy-based binding studies followed by biochemical assays with both bacterial and eukaryotic systems, that DTD effectively misedits Gly-tRNAGly. High-resolution crystal structure reveals that the architecture of DTD's chiral proofreading site is completely porous to achiral glycine. Hence, L-chiral rejection is the only design principle on which DTD functions, unlike other chiral-specific enzymes such as D-amino acid oxidases, which are specific for D-enantiomers. Competition assays with elongation factor thermo unstable (EF-Tu) and DTD demonstrate that EF-Tu precludes Gly-tRNAGly misediting at normal cellular concentrations. However, even slightly higher DTD levels overcome this protection conferred by EF-Tu, thus resulting in significant depletion of Gly-tRNAGly. Our in vitro observations are substantiated by cell-based studies in Escherichia coli that show that overexpression of DTD causes cellular toxicity, which is largely rescued upon glycine supplementation. Furthermore, we provide direct evidence that DTD is an RNA-based catalyst, since it uses only the terminal 2'-OH of tRNA for catalysis without the involvement of protein side chains. The study therefore provides a unique paradigm of enzyme action for substrate selection/specificity by DTD, and thus explains the underlying cause of DTD's activity on Gly-tRNAGly. It also gives a molecular and functional basis for the necessity and the observed tight regulation of DTD levels, thereby preventing cellular toxicity due to misediting.


Subject(s)
Aminoacyltransferases/chemistry , Aminoacyltransferases/metabolism , Peptide Elongation Factor Tu/metabolism , Alanine/chemistry , Alanine/metabolism , Aminoacyltransferases/genetics , Catalytic Domain , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Escherichia coli/cytology , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Glycine/chemistry , Glycine/metabolism , Hydrolysis , Magnetic Resonance Spectroscopy , Peptide Elongation Factor Tu/genetics , Plasmodium falciparum/enzymology , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/metabolism , Ribosomes/metabolism , Substrate Specificity , Zebrafish Proteins/metabolism
9.
Mitochondrial DNA A DNA Mapp Seq Anal ; 27(5): 3086-8, 2016 09.
Article in English | MEDLINE | ID: mdl-25629498

ABSTRACT

Recently, a growing number of reports had shown the association between mitochondrial DNA (mtDNA) sequence variants and aplastic anemia (AA). Owing to its high mutation rate, mtDNA variant had become biomarker for clinical and molecular diagnosis for AA. However, the relationship between mtDNA variant and AA was largely unknown. In this study, we reanalyzed the possible association between a "pathogenic" mutation A10055G in mt-tRNA(Gly) gene and AA, through the application of bioinformatics tool, we found that this mutation did not alter the secondary structure of tRNA(Gly), the pathogenicity scoring system indicated that the score of this mutation was only two points and belonged to a "neutral polymorphism", suggested that the role of A10055G mutation in clinical expression in AA needed to be further experimentally addressed.


Subject(s)
Alleles , Anemia, Aplastic/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , RNA, Transfer, Gly/genetics , Animals , Computational Biology , Databases, Nucleic Acid , Evolution, Molecular , Genes, Mitochondrial , Humans , Mutation , Nucleic Acid Conformation , Phylogeny , RNA, Transfer, Gly/chemistry
10.
J Biol Chem ; 290(38): 23336-47, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26229106

ABSTRACT

Many amino acid-related genes in Gram-positive bacteria are regulated by the T box riboswitch. The leader RNA of genes in the T box family controls the expression of downstream genes by monitoring the aminoacylation status of the cognate tRNA. Previous studies identified a three-nucleotide codon, termed the "Specifier Sequence," in the riboswitch that corresponds to the amino acid identity of the downstream genes. Pairing of the Specifier Sequence with the anticodon of the cognate tRNA is the primary determinant of specific tRNA recognition. This interaction mimics codon-anticodon pairing in translation but occurs in the absence of the ribosome. The goal of the current study was to determine the effect of a full range of mismatches for comparison with codon recognition in translation. Mutations were individually introduced into the Specifier Sequence of the glyQS leader RNA and tRNA(Gly) anticodon to test the effect of all possible pairing combinations on tRNA binding affinity and antitermination efficiency. The functional role of the conserved purine 3' of the Specifier Sequence was also verifiedin this study. We found that substitutions at the Specifier Sequence resulted in reduced binding, the magnitude of which correlates well with the predicted stability of the RNA-RNA pairing. However, the tolerance for specific mismatches in antitermination was generally different from that during decoding, which reveals a unique tRNA recognition pattern in the T box antitermination system.


Subject(s)
Anticodon/chemistry , Bacillus subtilis/chemistry , Codon/chemistry , RNA, Bacterial/chemistry , RNA, Transfer, Gly/chemistry , Riboswitch/physiology , Anticodon/genetics , Anticodon/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/biosynthesis , Codon/genetics , Codon/metabolism , Protein Biosynthesis/physiology , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Transfer, Gly/genetics , RNA, Transfer, Gly/metabolism
11.
RNA ; 21(10): 1790-806, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26276802

ABSTRACT

In Staphylococcus aureus, a T-box riboswitch exists upstream of the glyS gene to regulate transcription of the sole glycyl-tRNA synthetase, which aminoacylates five tRNA(Gly) isoacceptors bearing GCC or UCC anticodons. Subsequently, the glycylated tRNAs serve as substrates for decoding glycine codons during translation, and also as glycine donors for exoribosomal synthesis of pentaglycine peptides during cell wall formation. Probing of the predicted T-box structure revealed a long stem I, lacking features previously described for similar T-boxes. Moreover, the antiterminator stem includes a 42-nt long intervening sequence, which is staphylococci-specific. Finally, the terminator conformation adopts a rigid two-stem structure, where the intervening sequence forms the first stem followed by the second stem, which includes the more conserved residues. Interestingly, all five tRNA(Gly) isoacceptors interact with S. aureus glyS T-box with different binding affinities and they all induce transcription readthrough at different levels. The ability of both GCC and UCC anticodons to interact with the specifier loop indicates ambiguity during the specifier triplet reading, similar to the unconventional reading of glycine codons during protein synthesis. The S. aureus glyS T-box structure is consistent with the recent crystallographic and NMR studies, despite apparent differences, and highlights the phylogenetic variability of T-boxes when studied in a genome-dependent context. Our data suggest that the S. aureus glyS T-box exhibits differential tRNA selectivity, which possibly contributes toward the regulation and synchronization of ribosomal and exoribosomal peptide synthesis, two essential but metabolically unrelated pathways.


Subject(s)
Proteins/metabolism , RNA, Transfer, Gly/metabolism , Riboswitch , Base Sequence , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Transfer, Gly/chemistry , Sequence Homology, Nucleic Acid , Staphylococcus aureus/genetics , Transcription, Genetic
12.
Mitochondrion ; 21: 49-57, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25615420

ABSTRACT

In this report, we investigate molecular pathogenic mechanism of a diabetes-associated homoplasmic mitochondrial tRNA mutation in a Han Chinese family with maternally transmitted diabetes mellitus. Of 10 adult matrilineal relatives, 5 individuals suffered from diabetes (4 subjects with only diabetes, one subject with both diabetes and hearing impairment), while other five matrilineal relatives (one with hearing loss) had glucose intolerance. The average age at onset of diabetes in matrilineal relatives was 50 years. Molecular analysis of their mitochondrial genomes identified the novel homoplasmic T10003C mutation in the tRNA(Gly) gene belonging to haplogroup M11b. The T10003C mutation is expected to form a base-pairing (13C-22G) at the highly conserved D-stem of tRNA(Gly), thereby affecting secondary structure and function of this tRNA. A tRNA Northern analysis revealed that the T10003C mutation caused ~70% reduction in the steady-state level of tRNA(Gly). An in vivo translation analysis showed ~33% reduction in the rate of mitochondrial translation in mutant cells. Oxygen consumption analysis showed the defects in overall respiratory capacity or the ATP-linked, proton leak, and maximal respiration in mutant cells. As a result, the cellular energy deficiency contributes to the development of diabetes in subjects carrying the T10003C mutation. These data provide the first direct evidence that the tRNA(Gly) mutation might be associated with diabetes. Thus, our findings may provide new insights into the understanding of pathophysiology of maternally inherited diabetes.


Subject(s)
Diabetes Mellitus/genetics , Genes, Mitochondrial , Mitochondrial Diseases/genetics , Mutation , RNA, Transfer, Gly/genetics , Adenosine Triphosphate/metabolism , Adult , Aged , Asian People , Blotting, Northern , Cell Respiration , Energy Metabolism , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Transfer, Gly/chemistry , Young Adult
13.
J Biol Chem ; 289(29): 20359-69, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24898252

ABSTRACT

Aminoacyl-tRNA synthetases are an ancient enzyme family that specifically charges tRNA molecules with cognate amino acids for protein synthesis. Glycyl-tRNA synthetase (GlyRS) is one of the most intriguing aminoacyl-tRNA synthetases due to its divergent quaternary structure and abnormal charging properties. In the past decade, mutations of human GlyRS (hGlyRS) were also found to be associated with Charcot-Marie-Tooth disease. However, the mechanisms of traditional and alternative functions of hGlyRS are poorly understood due to a lack of studies at the molecular basis. In this study we report crystal structures of wild type and mutant hGlyRS in complex with tRNA and with small substrates and describe the molecular details of enzymatic recognition of the key tRNA identity elements in the acceptor stem and the anticodon loop. The cocrystal structures suggest that insertions 1 and 3 work together with the active site in a cooperative manner to facilitate efficient substrate binding. Both the enzyme and tRNA molecules undergo significant conformational changes during glycylation. A working model of multiple conformations for hGlyRS catalysis is proposed based on the crystallographic and biochemical studies. This study provides insights into the catalytic pathway of hGlyRS and may also contribute to our understanding of Charcot-Marie-Tooth disease.


Subject(s)
Glycine-tRNA Ligase/chemistry , RNA, Transfer, Gly/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain/genetics , Charcot-Marie-Tooth Disease/enzymology , Charcot-Marie-Tooth Disease/genetics , Crystallography, X-Ray , Glycine-tRNA Ligase/genetics , Glycine-tRNA Ligase/metabolism , Glycosylation , HeLa Cells , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Models, Molecular , Mutagenesis, Insertional , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleic Acid Conformation , Protein Conformation , RNA, Transfer, Gly/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermus thermophilus/enzymology , Thermus thermophilus/genetics
14.
FEBS Lett ; 587(21): 3495-9, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24036450

ABSTRACT

In Gram-positive bacteria the tRNA-dependent T box riboswitch regulates the expression of many amino acid biosynthetic and aminoacyl-tRNA synthetase genes through a transcription attenuation mechanism. The Specifier domain of the T box riboswitch contains the Specifier sequence that is complementary to the tRNA anticodon and is flanked by a highly conserved purine nucleotide that could result in a fourth base pair involving the invariant U33 of tRNA. We show that the interaction between the T box Specifier domain and tRNA consists of three Watson-Crick base pairs and that U33 confers stability to the complex through intramolecular hydrogen bonding. Enhanced packing within the Specifier domain loop E motif may stabilize the complex and contribute to cognate tRNA selection.


Subject(s)
Anticodon/chemistry , RNA, Transfer, Gly/chemistry , Riboswitch , Anticodon/metabolism , Base Pairing , Base Sequence , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , RNA, Transfer, Gly/metabolism , Solutions
15.
Biochemistry ; 51(17): 3662-74, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22468768

ABSTRACT

Although the fate of most tRNA molecules in the cell is aminoacylation and delivery to the ribosome, some tRNAs are destined to fulfill other functional roles. In addition to their central role in translation, tRNA molecules participate in processes such as regulation of gene expression, bacterial cell wall biosynthesis, viral replication, antibiotic biosynthesis, and suppression of alternative splicing. In bacteria, glycyl-tRNA molecules with anticodon sequences GCC and UCC exhibit multiple extratranslational functions, including transcriptional regulation and cell wall biosynthesis. We have determined the high-resolution structures of three glycyl-tRNA anticodon arms with anticodon sequences GCC and UCC. Two of the tRNA molecules are proteinogenic (tRNA(Gly,GCC) and tRNA(Gly,UCC)), and the third is nonproteinogenic (np-tRNA(Gly,UCC)) and participates in cell wall biosynthesis. The UV-monitored thermal melting curves show that the anticodon arm of tRNA(Gly,UCC) with a loop-closing C-A(+) base pair melts at a temperature 10 °C lower than those of tRNA(Gly,GCC) and np-tRNA(Gly,UCC). U-A and C-G pairs close the loops of the latter two molecules and enhance stem stability. Mg(2+) stabilizes the tRNA(Gly,UCC) anticodon arm and reduces the T(m) differential. The structures of the three tRNA(Gly) anticodon arms exhibit small differences among one another, but none of them form the classical U-turn motif. The anticodon loop of tRNA(Gly,GCC) becomes more dynamic and disordered in the presence of multivalent cations, whereas metal ion coordination in the anticodon loops of tRNA(Gly,UCC) and np-tRNA(Gly,UCC) establishes conformational homogeneity. The conformational similarity of the molecules is greater than their functional differences might suggest. Because aminoacylation of full-length tRNA molecules is accomplished by one tRNA synthetase, the similar structural context of the loop may facilitate efficient recognition of each of the anticodon sequences.


Subject(s)
Anticodon/chemistry , Anticodon/physiology , Protein Biosynthesis , RNA, Transfer, Gly/chemistry , Transcription, Genetic , Aminoacylation/genetics , Cell Wall/chemistry , Cell Wall/genetics , Glycine-tRNA Ligase/chemistry , Glycine-tRNA Ligase/genetics , Nuclear Magnetic Resonance, Biomolecular/methods , Nucleic Acid Conformation , Protein Biosynthesis/genetics , Protein Conformation , Staphylococcus aureus/genetics , Trinucleotide Repeats/genetics
16.
Nucleic Acids Res ; 40(12): 5602-14, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22373920

ABSTRACT

Adaptation to the host cell environment to efficiently take-over the host cell's machinery is crucial in particular for small RNA viruses like picornaviruses that come with only small RNA genomes and replicate exclusively in the cytosol. Their Internal Ribosome Entry Site (IRES) elements are specific RNA structures that facilitate the 5' end-independent internal initiation of translation both under normal conditions and when the cap-dependent host protein synthesis is shut-down in infected cells. A longstanding issue is which host factors play a major role in this internal initiation. Here, we show that the functionally most important domain V of the poliovirus IRES uses tRNA(Gly) anticodon stem-loop mimicry to recruit glycyl-tRNA synthetase (GARS) to the apical part of domain V, adjacent to the binding site of the key initiation factor eIF4G. The binding of GARS promotes the accommodation of the initiation region of the IRES in the mRNA binding site of the ribosome, thereby greatly enhancing the activity of the IRES at the step of the 48S initiation complex formation. Moonlighting functions of GARS that may be additionally needed for other events of the virus-host cell interaction are discussed.


Subject(s)
5' Untranslated Regions , Glycine-tRNA Ligase/metabolism , Peptide Chain Initiation, Translational , Poliovirus/genetics , RNA, Viral/chemistry , Base Sequence , Binding Sites , Glycine-tRNA Ligase/chemistry , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , RNA, Transfer, Gly/chemistry , RNA, Viral/metabolism
17.
Nucleic Acids Res ; 39(17): 7688-701, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21653555

ABSTRACT

Uridine at the wobble position of tRNA is usually modified, and modification is required for accurate and efficient protein translation. In eukaryotes, wobble uridines are modified into 5-methoxycarbonylmethyluridine (mcm(5)U), 5-carbamoylmethyluridine (ncm(5)U) or derivatives thereof. Here, we demonstrate, both by in vitro and in vivo studies, that the Arabidopsis thaliana methyltransferase AT1G31600, denoted by us AtTRM9, is responsible for the final step in mcm(5)U formation, thus representing a functional homologue of the Saccharomyces cerevisiae Trm9 protein. We also show that the enzymatic activity of AtTRM9 depends on either one of two closely related proteins, AtTRM112a and AtTRM112b. Moreover, we demonstrate that AT1G36310, denoted AtALKBH8, is required for hydroxylation of mcm(5)U to (S)-mchm(5)U in tRNA(Gly)(UCC), and has a function similar to the mammalian dioxygenase ALKBH8. Interestingly, atalkbh8 mutant plants displayed strongly increased levels of mcm(5)U, and also of mcm(5)Um, its 2'-O-ribose methylated derivative. This suggests that accumulated mcm(5)U is prone to further ribose methylation by a non-specialized mechanism, and may challenge the notion that the existence of mcm(5)U- and mcm(5)Um-containing forms of the selenocysteine-specific tRNA(Sec) in mammals reflects an important regulatory process. The present study reveals a role in for several hitherto uncharacterized Arabidopsis proteins in the formation of modified wobble uridines.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Dioxygenases/metabolism , Mixed Function Oxygenases/metabolism , Uridine/metabolism , tRNA Methyltransferases/metabolism , AlkB Homolog 8, tRNA Methyltransferase , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Dioxygenases/chemistry , Dioxygenases/genetics , Humans , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Molecular Sequence Data , Mutation , RNA, Transfer/chemistry , RNA, Transfer/metabolism , RNA, Transfer, Gly/chemistry , RNA, Transfer, Gly/metabolism , Sequence Alignment , tRNA Methyltransferases/chemistry , tRNA Methyltransferases/genetics
18.
Nat Commun ; 2: 172, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21285950

ABSTRACT

Mammals have nine different homologues (ALKBH1-9) of the Escherichia coli DNA repair demethylase AlkB. ALKBH2 is a genuine DNA repair enzyme, but the in vivo function of the other ALKBH proteins has remained elusive. It was recently shown that ALKBH8 contains an additional transfer RNA (tRNA) methyltransferase domain, which generates the wobble nucleoside 5-methoxycarbonylmethyluridine (mcm(5)U) from its precursor 5-carboxymethyluridine (cm(5)U). In this study, we report that (R)- and 5-methoxycarbonylhydroxymethyluridine (mchm(5)U), hydroxylated forms of mcm(5)U, are present in mammalian tRNA-Arg(UCG), and tRNA-Gly(UCC), respectively, representing the first example of a diastereomeric pair of modified RNA nucleosides. Through in vitro and in vivo studies, we show that both diastereomers of mchm(5)U are generated from mcm(5)U, and that the AlkB domain of ALKBH8 specifically hydroxylates mcm(5)U into (S)-mchm(5)U in tRNA-Gly(UCC). These findings expand the function of the ALKBH oxygenases beyond nucleic acid repair and increase the current knowledge on mammalian wobble uridine modifications and their biogenesis.


Subject(s)
Codon/genetics , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , RNA, Transfer, Gly/chemistry , Uridine/analogs & derivatives , tRNA Methyltransferases/genetics , AlkB Homolog 8, tRNA Methyltransferase , Animals , Cattle , Chromatography, Liquid , Computational Biology , Mice , Molecular Structure , Species Specificity , Tandem Mass Spectrometry , Uridine/chemistry , Uridine/metabolism , tRNA Methyltransferases/chemistry , tRNA Methyltransferases/metabolism
19.
Biochem Biophys Res Commun ; 404(1): 245-9, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21114959

ABSTRACT

tRNAs are aminoacylated with the correct amino acid by the cognate aminoacyl-tRNA synthetase. The tRNA/synthetase systems can be divided into two classes: class I and class II. Within class I, the tRNA identity elements that enable the specificity consist of complex sequence and structure motifs, whereas in class II the identity elements are assured by few and simple determinants, which are mostly located in the tRNA acceptor stem. The tRNA(Gly)/glycyl-tRNA-synthetase (GlyRS) system is a special case regarding evolutionary aspects. There exist two different types of GlyRS, namely an archaebacterial/human type and an eubacterial type, reflecting the evolutionary divergence within this system. We previously reported the crystal structures of an Escherichia coli and of a human tRNA(Gly) acceptor stem microhelix. Here we present the crystal structure of a thermophilic tRNA(Gly) aminoacyl stem from Thermus thermophilus at 1.6Å resolution and provide insight into the RNA geometry and hydration.


Subject(s)
RNA, Transfer, Gly/chemistry , Thermus thermophilus/metabolism , Base Sequence , Crystallography, X-Ray , Glycine-tRNA Ligase/metabolism , Nucleic Acid Conformation , RNA, Transfer, Gly/metabolism
20.
Nucleic Acids Res ; 37(22): 7638-53, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19793868

ABSTRACT

We analyzed cleavage of precursor tRNAs with an LNA, 2'-OCH(3), 2'-H or 2'-F modification at the canonical (c(0)) site by bacterial RNase P. We infer that the major function of the 2'-substituent at nt -1 during substrate ground state binding is to accept an H-bond. Cleavage of the LNA substrate at the c(0) site by Escherichia coli RNase P RNA demonstrated that the transition state for cleavage can in principle be achieved with a locked C3' -endo ribose and without the H-bond donor function of the 2'-substituent. LNA and 2'-OCH(3) suppressed processing at the major aberrant m(-)(1) site; instead, the m(+1) (nt +1/+2) site was utilized. For the LNA variant, parallel pathways leading to cleavage at the c(0) and m(+1) sites had different pH profiles, with a higher Mg(2+) requirement for c(0) versus m(+1) cleavage. The strong catalytic defect for LNA and 2'-OCH(3) supports a model where the extra methylene (LNA) or methyl group (2'-OCH(3)) causes a steric interference with a nearby bound catalytic Mg(2+) during its recoordination on the way to the transition state for cleavage. The presence of the protein cofactor suppressed the ground state binding defects, but not the catalytic defects.


Subject(s)
Escherichia coli Proteins/metabolism , Oligonucleotides/chemistry , RNA Precursors/chemistry , RNA, Transfer, Gly/chemistry , Ribonuclease P/metabolism , Bacillus subtilis/enzymology , Base Sequence , Biocatalysis , Holoenzymes/metabolism , Hydrogen-Ion Concentration , Kinetics , Magnesium/chemistry , Molecular Sequence Data , Phosphates/chemistry , RNA Precursors/metabolism , RNA, Transfer, Gly/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...