Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
RNA ; 30(2): 105-112, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38071475

ABSTRACT

Ribosomal RNA contains many posttranscriptionally modified nucleosides, particularly in the functional parts of the ribosome. The distribution of these modifications varies from one organism to another. In Bacillus subtilis, the model organism for Gram-positive bacteria, mass spectrometry experiments revealed the presence of 7-methylguanosine (m7G) at position 2574 of the 23S rRNA, which lies in the A-site of the peptidyl transferase center of the large ribosomal subunit. Testing several m7G methyltransferase candidates allowed us to identify the RlmQ enzyme, encoded by the ywbD open reading frame, as the MTase responsible for this modification. The enzyme methylates free RNA and not ribosomal 50S or 70S particles, suggesting that modification occurs in the early steps of ribosome biogenesis.


Subject(s)
Peptidyl Transferases , Peptidyl Transferases/genetics , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 23S/chemistry , Bacillus subtilis/genetics , RNA/chemistry , Methyltransferases/genetics
2.
RNA ; 28(9): 1185-1196, 2022 09.
Article in English | MEDLINE | ID: mdl-35710145

ABSTRACT

A previous bioinformatic analysis predicted that the ysgA open reading frame of Bacillus subtilis encodes an RNA methyltransferase of the SPOUT superfamily. Here we show that YsgA is the 2'-O-methyltransferase that targets position G2553 (Escherichia coli numbering) of the A-loop of 23S rRNA. This was shown by a combination of biochemical and mass spectrometry approaches using both rRNA extracted from B. subtilis wild-type or ΔysgA cells and in vitro synthesized rRNA. When the target G2553 is mutated, YsgA is able to methylate the ribose of adenosine. However, it cannot methylate cytidine nor uridine. The enzyme modifies free 23S rRNA but not the fully assembled ribosome nor the 50S subunit, suggesting that the modification occurs early during ribosome biogenesis. Nevertheless, ribosome subunits assembly is unaffected in a B. subtilis ΔysgA mutant strain. The crystal structure of the recombinant YsgA protein, combined with mutagenesis data, outlined in this article highlights a typical SPOUT fold preceded by an L7Ae/L30 (eL8/eL30 in a new nomenclature) amino-terminal domain.


Subject(s)
Methyltransferases , RNA, Ribosomal, 23S , AAA Domain , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Escherichia coli/metabolism , Guanosine/analogs & derivatives , Methylation , Methyltransferases/metabolism , Open Reading Frames , RNA, Ribosomal, 23S/chemistry
3.
Yeast ; 39(9): 493-507, 2022 09.
Article in English | MEDLINE | ID: mdl-35942513

ABSTRACT

Nitrogen catabolite repression (NCR) is a major transcriptional control pathway governing nitrogen use in yeast, with several hundred of target genes identified to date. Early and extensive studies on NCR led to the identification of the 4 GATA zinc finger transcription factors, but the primary mechanism initiating NCR is still unclear up till now. To identify novel players of NCR, we have undertaken a genetic screen in an NCR-relieved gdh1Δ mutant, which led to the identification of four genes directly linked to protein ubiquitylation. Ubiquitylation is an important way of regulating amino acid transporters and our observations being specifically observed in glutamine-containing media, we hypothesized that glutamine transport could be involved in establishing NCR. Stabilization of Gap1 at the plasma membrane restored NCR in gdh1Δ cells and AGP1 (but not GAP1) deletion could relieve repression in the ubiquitylation mutants isolated during the screen. Altogether, our results suggest that deregulated glutamine transporter function in all three weak nitrogen derepressed (wnd) mutants restores the repression of NCR-sensitive genes consecutive to GDH1 deletion.


Subject(s)
Amino Acid Transport Systems, Neutral , Catabolite Repression , Saccharomyces cerevisiae Proteins , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , GATA Transcription Factors/chemistry , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Gene Expression Regulation, Fungal , Glutamine/genetics , Glutamine/metabolism , Nitrogen/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nucleic Acids Res ; 47(9): 4736-4750, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30931478

ABSTRACT

1-Methyladenosine (m1A) is a modified nucleoside found at positions 9, 14, 22 and 58 of tRNAs, which arises from the transfer of a methyl group onto the N1-atom of adenosine. The yqfN gene of Bacillus subtilis encodes the methyltransferase TrmK (BsTrmK) responsible for the formation of m1A22 in tRNA. Here, we show that BsTrmK displays a broad substrate specificity, and methylates seven out of eight tRNA isoacceptor families of B. subtilis bearing an A22. In addition to a non-Watson-Crick base-pair between the target A22 and a purine at position 13, the formation of m1A22 by BsTrmK requires a full-length tRNA with intact tRNA elbow and anticodon stem. We solved the crystal structure of BsTrmK showing an N-terminal catalytic domain harbouring the typical Rossmann-like fold of Class-I methyltransferases and a C-terminal coiled-coil domain. We used NMR chemical shift mapping to drive the docking of BstRNASer to BsTrmK in complex with its methyl-donor cofactor S-adenosyl-L-methionine (SAM). In this model, validated by methyltransferase activity assays on BsTrmK mutants, both domains of BsTrmK participate in tRNA binding. BsTrmK recognises tRNA with very few structural changes in both partner, the non-Watson-Crick R13-A22 base-pair positioning the A22 N1-atom close to the SAM methyl group.


Subject(s)
Bacillus subtilis/chemistry , RNA Recognition Motif Proteins/chemistry , S-Adenosylmethionine/chemistry , tRNA Methyltransferases/chemistry , Anticodon/chemistry , Anticodon/genetics , Bacillus subtilis/enzymology , Catalytic Domain/genetics , Crystallography, X-Ray , Methylation , Protein Conformation , RNA Recognition Motif Proteins/genetics , RNA, Transfer/chemistry , RNA, Transfer/genetics , Substrate Specificity , tRNA Methyltransferases/genetics
5.
RNA ; 24(8): 1080-1092, 2018 08.
Article in English | MEDLINE | ID: mdl-29848639

ABSTRACT

tRNA molecules get heavily modified post-transcriptionally. The N-1 methylation of purines at position 9 of eukaryal and archaeal tRNA is catalyzed by the SPOUT methyltranferase Trm10. Remarkably, while certain Trm10 orthologs are specific for either guanosine or adenosine, others show a dual specificity. Structural and functional studies have been performed on guanosine- and adenosine-specific enzymes. Here we report the structure and biochemical analysis of the dual-specificity enzyme from Thermococcus kodakaraensis (TkTrm10). We report the first crystal structure of a construct of this enzyme, consisting of the N-terminal domain and the catalytic SPOUT domain. Moreover, crystal structures of the SPOUT domain, either in the apo form or bound to S-adenosyl-l-methionine or S-adenosyl-l-homocysteine reveal the conformational plasticity of two active site loops upon substrate binding. Kinetic analysis shows that TkTrm10 has a high affinity for its tRNA substrates, while the enzyme on its own has a very low methyltransferase activity. Mutation of either of two active site aspartate residues (Asp206 and Asp245) to Asn or Ala results in only modest effects on the N-1 methylation reaction, with a small shift toward a preference for m1G formation over m1A formation. Only a double D206A/D245A mutation severely impairs activity. These results are in line with the recent finding that the single active-site aspartate was dispensable for activity in the guanosine-specific Trm10 from yeast, and suggest that also dual-specificity Trm10 orthologs use a noncanonical tRNA methyltransferase mechanism without residues acting as general base catalysts.


Subject(s)
Adenosine/chemistry , Guanosine/chemistry , RNA Processing, Post-Transcriptional/physiology , Thermococcus/enzymology , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism , Binding Sites , Catalysis , Catalytic Domain/physiology , Crystallography, X-Ray , Models, Molecular , Molecular Docking Simulation , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Substrate Specificity/genetics , Thermococcus/metabolism
6.
J Biol Chem ; 293(33): 12862-12876, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29880640

ABSTRACT

Mitochondrial tRNAs are transcribed as long polycistronic transcripts of precursor tRNAs and undergo posttranscriptional modifications such as endonucleolytic processing and methylation required for their correct structure and function. Among them, 5'-end processing and purine 9 N1-methylation of mitochondrial tRNA are catalyzed by two proteinaceous complexes with overlapping subunit composition. The Mg2+-dependent RNase P complex for 5'-end cleavage comprises the methyltransferase domain-containing protein tRNA methyltransferase 10C, mitochondrial RNase P subunit (TRMT10C/MRPP1), short-chain oxidoreductase hydroxysteroid 17ß-dehydrogenase 10 (HSD17B10/MRPP2), and metallonuclease KIAA0391/MRPP3. An MRPP1-MRPP2 subcomplex also catalyzes the formation of 1-methyladenosine/1-methylguanosine at position 9 using S-adenosyl-l-methionine as methyl donor. However, a lack of structural information has precluded insights into how these complexes methylate and process mitochondrial tRNA. Here, we used a combination of X-ray crystallography, interaction and activity assays, and small angle X-ray scattering (SAXS) to gain structural insight into the two tRNA modification complexes and their components. The MRPP1 N terminus is involved in tRNA binding and monomer-monomer self-interaction, whereas the C-terminal SPOUT fold contains key residues for S-adenosyl-l-methionine binding and N1-methylation. The entirety of MRPP1 interacts with MRPP2 to form the N1-methylation complex, whereas the MRPP1-MRPP2-MRPP3 RNase P complex only assembles in the presence of precursor tRNA. This study proposes low-resolution models of the MRPP1-MRPP2 and MRPP1-MRPP2-MRPP3 complexes that suggest the overall architecture, stoichiometry, and orientation of subunits and tRNA substrates.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/chemistry , Methyltransferases/chemistry , Models, Molecular , Multienzyme Complexes/chemistry , RNA, Mitochondrial/chemistry , RNA, Transfer/chemistry , Ribonuclease P/chemistry , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Crystallography, X-Ray , Humans , Methyltransferases/metabolism , Multienzyme Complexes/metabolism , RNA, Mitochondrial/metabolism , RNA, Transfer/metabolism , Ribonuclease P/metabolism , Scattering, Small Angle
7.
Amino Acids ; 50(12): 1647-1661, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30238253

ABSTRACT

In all organisms, carbamoylphosphate (CP) is a precursor common to the synthesis of arginine and pyrimidines. In Escherichia coli and most other Gram-negative bacteria, CP is produced by a single enzyme, carbamoylphosphate synthase (CPSase), encoded by the carAB operon. This particular situation poses a question of basic physiological interest: what are the metabolic controls coordinating the synthesis and distribution of this high-energy substance in view of the needs of both pathways? The study of the mechanisms has revealed unexpected moonlighting gene regulatory activities of enzymes and functional links between mechanisms as diverse as gene regulation and site-specific DNA recombination. At the level of enzyme production, various regulatory mechanisms were found to cooperate in a particularly intricate transcriptional control of a pair of tandem promoters. Transcription initiation is modulated by an interplay of several allosteric DNA-binding transcription factors using effector molecules from three different pathways (arginine, pyrimidines, purines), nucleoid-associated factors (NAPs), trigger enzymes (enzymes with a second unlinked gene regulatory function), DNA remodeling (bending and wrapping), UTP-dependent reiterative transcription initiation, and stringent control by the alarmone ppGpp. At the enzyme level, CPSase activity is tightly controlled by allosteric effectors originating from different pathways: an inhibitor (UMP) and two activators (ornithine and IMP) that antagonize the inhibitory effect of UMP. Furthermore, it is worth noticing that all reaction intermediates in the production of CP are extremely reactive and unstable, and protected by tunneling through a 96 Å long internal channel.


Subject(s)
Carbamyl Phosphate/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Ligases/genetics , Arginine/biosynthesis , Escherichia coli/genetics , Promoter Regions, Genetic , Pyrimidines/biosynthesis
8.
Nucleic Acids Res ; 44(2): 940-53, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26673726

ABSTRACT

Purine nucleosides on position 9 of eukaryal and archaeal tRNAs are frequently modified in vivo by the post-transcriptional addition of a methyl group on their N1 atom. The methyltransferase Trm10 is responsible for this modification in both these domains of life. While certain Trm10 orthologues specifically methylate either guanosine or adenosine at position 9 of tRNA, others have a dual specificity. Until now structural information about this enzyme family was only available for the catalytic SPOUT domain of Trm10 proteins that show specificity toward guanosine. Here, we present the first crystal structure of a full length Trm10 orthologue specific for adenosine, revealing next to the catalytic SPOUT domain also N- and C-terminal domains. This structure hence provides crucial insights in the tRNA binding mechanism of this unique monomeric family of SPOUT methyltransferases. Moreover, structural comparison of this adenosine-specific Trm10 orthologue with guanosine-specific Trm10 orthologues suggests that the N1 methylation of adenosine relies on additional catalytic residues.


Subject(s)
Adenosine/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , RNA, Transfer/metabolism , Sulfolobus acidocaldarius/enzymology , tRNA Methyltransferases/metabolism , Adenosine/chemistry , Archaeal Proteins/genetics , Catalytic Domain , Crystallography, X-Ray , Methylation , Models, Molecular , Molecular Docking Simulation , Protein Structure, Tertiary , RNA, Transfer/chemistry , RNA, Transfer, Met/chemistry , RNA, Transfer, Met/metabolism , Scattering, Small Angle , X-Ray Diffraction , tRNA Methyltransferases/chemistry , tRNA Methyltransferases/genetics
9.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3294-3302, 2017 12.
Article in English | MEDLINE | ID: mdl-28888424

ABSTRACT

MRPP2 (also known as HSD10/SDR5C1) is a multifunctional protein that harbours both catalytic and non-catalytic functions. The protein belongs to the short-chain dehydrogenase/reductases (SDR) family and is involved in the catabolism of isoleucine in vivo and steroid metabolism in vitro. MRPP2 also moonlights in a complex with the MRPP1 (also known as TRMT10C) protein for N1-methylation of purines at position 9 of mitochondrial tRNA, and in a complex with MRPP1 and MRPP3 (also known as PRORP) proteins for 5'-end processing of mitochondrial precursor tRNA. Inherited mutations in the HSD17B10 gene encoding MRPP2 protein lead to a childhood disorder characterised by progressive neurodegeneration, cardiomyopathy or both. Here we report two patients with novel missense mutations in the HSD17B10 gene (c.34G>C and c.526G>A), resulting in the p.V12L and p.V176M substitutions. Val12 and Val176 are highly conserved residues located at different regions of the MRPP2 structure. Recombinant mutant proteins were expressed and characterised biochemically to investigate their effects towards the functions of MRPP2 and associated complexes in vitro. Both mutant proteins showed significant reduction in the dehydrogenase, methyltransferase and tRNA processing activities compared to wildtype, associated with reduced stability for protein with p.V12L, whereas the protein carrying p.V176M showed impaired kinetics and complex formation. This study therefore identified two distinctive molecular mechanisms to explain the biochemical defects for the novel missense patient mutations.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Mitochondria/metabolism , RNA, Transfer/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/chemistry , Female , Gene Expression , Humans , Infant , Male , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Molecular , Mutation, Missense , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonuclease P/genetics , Ribonuclease P/metabolism
10.
RNA ; 20(8): 1257-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24951554

ABSTRACT

The 2'-O-methylation of the nucleoside at position 32 of tRNA is found in organisms belonging to the three domains of life. Unrelated enzymes catalyzing this modification in Bacteria (TrmJ) and Eukarya (Trm7) have already been identified, but until now, no information is available for the archaeal enzyme. In this work we have identified the methyltransferase of the archaeon Sulfolobus acidocaldarius responsible for the 2'-O-methylation at position 32. This enzyme is a homolog of the bacterial TrmJ. Remarkably, both enzymes have different specificities for the nature of the nucleoside at position 32. While the four canonical nucleosides are substrates of the Escherichia coli enzyme, the archaeal TrmJ can only methylate the ribose of a cytidine. Moreover, the two enzymes recognize their tRNA substrates in a different way. We have solved the crystal structure of the catalytic domain of both enzymes to gain better understanding of these differences at a molecular level.


Subject(s)
RNA, Transfer/metabolism , tRNA Methyltransferases/metabolism , Archaea/genetics , Archaea/metabolism , Bacterial Proteins/metabolism , Catalysis , Escherichia coli/genetics , Escherichia coli/metabolism , Inverted Repeat Sequences , Models, Molecular , Mutation , Nucleic Acid Conformation , Nucleosides/chemistry , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , RNA, Transfer/chemistry , RNA, Transfer/genetics , Reproducibility of Results , Substrate Specificity , tRNA Methyltransferases/chemistry
11.
RNA ; 18(4): 815-24, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22337946

ABSTRACT

N(2)-methylguanosine (m(2)G) is found at position 6 in the acceptor stem of Thermus thermophilus tRNA(Phe). In this article, we describe the cloning, expression, and characterization of the T. thermophilus HB27 methyltransferase (MTase) encoded by the TTC1157 open reading frame that catalyzes the formation of this modified nucleoside. S-adenosyl-L-methionine is used as donor of the methyl group. The enzyme behaves as a monomer in solution. It contains an N-terminal THUMP domain predicted to bind RNA and contains a C-terminal Rossmann-fold methyltransferase (RFM) domain predicted to be responsible for catalysis. We propose to rename the TTC1157 gene trmN and the corresponding protein TrmN, according to the bacterial nomenclature of tRNA methyltransferases. Inactivation of the trmN gene in the T. thermophilus HB27 chromosome led to a total absence of m(2)G in tRNA but did not affect cell growth or the formation of other modified nucleosides in tRNA(Phe). Archaeal homologs of TrmN were identified and characterized. These proteins catalyze the same reaction as TrmN from T. thermophilus. Individual THUMP and RFM domains of PF1002 from Pyrococcus furiosus were produced. These separate domains were inactive and did not bind tRNA, reinforcing the idea that the THUMP domain acts in concert with the catalytic domain to target a particular position of the tRNA molecule.


Subject(s)
Methyltransferases/genetics , RNA, Transfer/genetics , Thermus thermophilus/genetics , Open Reading Frames , Thermus thermophilus/enzymology
12.
Nucleic Acids Res ; 40(11): 5149-61, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362751

ABSTRACT

Methyltransferases (MTases) form a major class of tRNA-modifying enzymes needed for the proper functioning of tRNA. Recently, RNA MTases from the TrmN/Trm14 family that are present in Archaea, Bacteria and Eukaryota have been shown to specifically modify tRNA(Phe) at guanosine 6 in the tRNA acceptor stem. Here, we report the first X-ray crystal structures of the tRNA m(2)G6 (N(2)-methylguanosine) MTase (TTC)TrmN from Thermus thermophilus and its ortholog (Pf)Trm14 from Pyrococcus furiosus. Structures of (Pf)Trm14 were solved in complex with the methyl donor S-adenosyl-l-methionine (SAM or AdoMet), as well as the reaction product S-adenosyl-homocysteine (SAH or AdoHcy) and the inhibitor sinefungin. (TTC)TrmN and (Pf)Trm14 consist of an N-terminal THUMP domain fused to a catalytic Rossmann-fold MTase (RFM) domain. These results represent the first crystallographic structure analysis of proteins containing both THUMP and RFM domain, and hence provide further insight in the contribution of the THUMP domain in tRNA recognition and catalysis. Electrostatics and conservation calculations suggest a main tRNA binding surface in a groove between the THUMP domain and the MTase domain. This is further supported by a docking model of TrmN in complex with tRNA(Phe) of T. thermophilus and via site-directed mutagenesis.


Subject(s)
Archaeal Proteins/chemistry , Bacterial Proteins/chemistry , tRNA Methyltransferases/chemistry , Amino Acid Sequence , Archaeal Proteins/metabolism , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Guanosine/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Pyrococcus furiosus/enzymology , Sequence Alignment , Thermus thermophilus/enzymology , tRNA Methyltransferases/metabolism
13.
Nucleic Acids Res ; 38(19): 6533-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20525789

ABSTRACT

Two archaeal tRNA methyltransferases belonging to the SPOUT superfamily and displaying unexpected activities are identified. These enzymes are orthologous to the yeast Trm10p methyltransferase, which catalyses the formation of 1-methylguanosine at position 9 of tRNA. In contrast, the Trm10p orthologue from the crenarchaeon Sulfolobus acidocaldarius forms 1-methyladenosine at the same position. Even more surprisingly, the Trm10p orthologue from the euryarchaeon Thermococcus kodakaraensis methylates the N(1)-atom of either adenosine or guanosine at position 9 in different tRNAs. This is to our knowledge the first example of a tRNA methyltransferase with a broadened nucleoside recognition capability. The evolution of tRNA methyltransferases methylating the N(1) atom of a purine residue is discussed.


Subject(s)
Adenosine/analogs & derivatives , Archaeal Proteins/metabolism , Guanosine/analogs & derivatives , tRNA Methyltransferases/metabolism , Adenosine/metabolism , Archaeal Proteins/classification , Biocatalysis , Escherichia coli/genetics , Guanosine/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Sulfolobus acidocaldarius/enzymology , Thermococcus/enzymology , tRNA Methyltransferases/classification
14.
Nucleic Acids Res ; 38(18): 6206-18, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20483913

ABSTRACT

The S-adenosyl-L-methionine dependent methylation of adenine 58 in the T-loop of tRNAs is essential for cell growth in yeast or for adaptation to high temperatures in thermophilic organisms. In contrast to bacterial and eukaryotic tRNA m(1)A58 methyltransferases that are site-specific, the homologous archaeal enzyme from Pyrococcus abyssi catalyzes the formation of m(1)A also at the adjacent position 57, m(1)A57 being a precursor of 1-methylinosine. We report here the crystal structure of P. abyssi tRNA m(1)A57/58 methyltransferase ((Pab)TrmI), in complex with S-adenosyl-L-methionine or S-adenosyl-L-homocysteine in three different space groups. The fold of the monomer and the tetrameric architecture are similar to those of the bacterial enzymes. However, the inter-monomer contacts exhibit unique features. In particular, four disulfide bonds contribute to the hyperthermostability of the archaeal enzyme since their mutation lowers the melting temperature by 16.5°C. His78 in conserved motif X, which is present only in TrmIs from the Thermococcocales order, lies near the active site and displays two alternative conformations. Mutagenesis indicates His78 is important for catalytic efficiency of (Pab)TrmI. When A59 is absent in tRNA(Asp), only A57 is modified. Identification of the methylated positions in tRNAAsp by mass spectrometry confirms that (Pab)TrmI methylates the first adenine of an AA sequence.


Subject(s)
Adenine/metabolism , Archaeal Proteins/chemistry , Pyrococcus abyssi/enzymology , RNA, Transfer, Asp/metabolism , tRNA Methyltransferases/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dimerization , Disulfides/chemistry , Enzyme Stability , Histidine/chemistry , Models, Molecular , Mutation , RNA, Transfer, Asp/chemistry , S-Adenosylmethionine/chemistry , tRNA Methyltransferases/genetics , tRNA Methyltransferases/metabolism
15.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 11): 1432-5, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22102250

ABSTRACT

Methyltransferases form a major class of tRNA-modifying enzymes that are needed for the proper functioning of tRNA. Here, the expression, purification and crystallization of two related putative tRNA methyltransferases from two kingdoms of life are reported. The protein encoded by the gene pf1002 from the archaeon Pyrococcus furiosus was crystallized in the monoclinic space group P2(1). A complete data set was collected to 2.2 Å resolution. The protein encoded by the gene ttc1157 from the eubacterium Thermus thermophilus was crystallized in the trigonal space group P3(2)21. A complete data set was collected to 2.05 Å resolution.


Subject(s)
Pyrococcus furiosus/enzymology , Thermus thermophilus/enzymology , tRNA Methyltransferases/chemistry , Crystallization , Crystallography, X-Ray
16.
Genes (Basel) ; 12(2)2021 01 22.
Article in English | MEDLINE | ID: mdl-33499018

ABSTRACT

The high conservation of nucleotides of the T-loop, including their chemical identity, are hallmarks of tRNAs from organisms belonging to the three Domains of Life. These structural characteristics allow the T-loop to adopt a peculiar intraloop conformation able to interact specifically with other conserved residues of the D-loop, which ultimately folds the mature tRNA in a unique functional canonical L-shaped architecture. Paradoxically, despite the high conservation of modified nucleotides in the T-loop, enzymes catalyzing their formation depend mostly on the considered organism, attesting for an independent but convergent evolution of the post-transcriptional modification processes. The driving force behind this is the preservation of a native conformation of the tRNA elbow that underlies the various interactions of tRNA molecules with different cellular components.


Subject(s)
Base Sequence , Conserved Sequence , Evolution, Molecular , Nucleic Acid Conformation , RNA Processing, Post-Transcriptional , RNA, Transfer/genetics , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Humans , Microbiology , RNA, Transfer/chemistry , Structure-Activity Relationship
17.
Nucleic Acids Res ; 36(10): 3252-62, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18420655

ABSTRACT

N(1)-methylation of adenosine to m(1)A occurs in several different positions in tRNAs from various organisms. A methyl group at position N(1) prevents Watson-Crick-type base pairing by adenosine and is therefore important for regulation of structure and stability of tRNA molecules. Thus far, only one family of genes encoding enzymes responsible for m(1)A methylation at position 58 has been identified, while other m(1)A methyltransferases (MTases) remain elusive. Here, we show that Bacillus subtilis open reading frame yqfN is necessary and sufficient for N(1)-adenosine methylation at position 22 of bacterial tRNA. Thus, we propose to rename YqfN as TrmK, according to the traditional nomenclature for bacterial tRNA MTases, or TrMet(m(1)A22) according to the nomenclature from the MODOMICS database of RNA modification enzymes. tRNAs purified from a DeltatrmK strain are a good substrate in vitro for the recombinant TrmK protein, which is sufficient for m(1)A methylation at position 22 as are tRNAs from Escherichia coli, which natively lacks m(1)A22. TrmK is conserved in Gram-positive bacteria and present in some Gram-negative bacteria, but its orthologs are apparently absent from archaea and eukaryota. Protein structure prediction indicates that the active site of TrmK does not resemble the active site of the m(1)A58 MTase TrmI, suggesting that these two enzymatic activities evolved independently.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/chemistry , tRNA Methyltransferases/chemistry , Amino Acid Sequence , Bacillus subtilis/growth & development , Bacterial Proteins/classification , Bacterial Proteins/genetics , Base Sequence , Models, Molecular , Molecular Sequence Data , Phylogeny , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Sequence Homology, Amino Acid , tRNA Methyltransferases/classification , tRNA Methyltransferases/genetics
18.
Proteins ; 71(4): 2076-85, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18186482

ABSTRACT

MnmC catalyses the last two steps in the biosynthesis of 5-methylaminomethyl-2-thiouridine (mnm(5)s(2)U) in tRNA. Previously, we reported that this bifunctional enzyme is encoded by the yfcK open reading frame in the Escherichia coli K12 genome. However, the mechanism of its activity, in particular the potential structural and functional dependence of the domains responsible for catalyzing the two modification reactions, remains unknown. With the aid of the protein fold-recognition method, we constructed a structural model of MnmC in complex with the ligands and target nucleosides and studied the role of individual amino acids and entire domains by site-directed and deletion mutagenesis, respectively. We found out that the N-terminal domain contains residues responsible for binding of the S-adenosylmethionine cofactor and catalyzing the methylation of nm(5)s(2)U to form mnm(5)s(2)U, while the C-terminal domain contains residues responsible for binding of the FAD cofactor. Further, point mutants with compromised activity of either domain can complement each other to restore a fully functional enzyme. Thus, in the conserved fusion protein MnmC, the individual domains retain independence as enzymes. Interestingly, the N-terminal domain is capable of independent folding, while the isolated C-terminal domain is incapable of folding on its own, a situation similar to the one reported recently for the rRNA modification enzyme RsmC.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , RNA, Transfer/biosynthesis , Thiouridine/analogs & derivatives , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Catalysis , Computational Biology/methods , Conserved Sequence , Escherichia coli Proteins/analysis , Escherichia coli Proteins/genetics , Flavin-Adenine Dinucleotide/metabolism , Genetic Complementation Test , Ligands , Methylation , Models, Molecular , Molecular Sequence Data , Multienzyme Complexes/analysis , Multienzyme Complexes/genetics , Protein Binding , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Bacterial/biosynthesis , Sequence Analysis , Sequence Deletion , Sequence Homology, Amino Acid , Static Electricity , Structure-Activity Relationship , Thiouridine/metabolism
19.
Nucleic Acids Res ; 34(15): 4293-301, 2006.
Article in English | MEDLINE | ID: mdl-16920741

ABSTRACT

Pseudouridine (Psi) located at position 55 in tRNA is a nearly universally conserved RNA modification found in all three domains of life. This modification is catalyzed by TruB in bacteria and by Pus4 in eukaryotes, but so far the Psi55 synthase has not been identified in archaea. In this work, we report the ability of two distinct pseudouridine synthases from the hyperthermophilic archaeon Pyrococcus furiosus to specifically modify U55 in tRNA in vitro. These enzymes are (pfu)Cbf5, a protein known to play a role in RNA-guided modification of rRNA, and (pfu)PsuX, a previously uncharacterized enzyme that is not a member of the TruB/Pus4/Cbf5 family of pseudouridine synthases. (pfu)PsuX is hereafter renamed (pfu)Pus10. Both enzymes specifically modify tRNA U55 in vitro but exhibit differences in substrate recognition. In addition, we find that in a heterologous in vivo system, (pfu)Pus10 efficiently complements an Escherichia coli strain deficient in the bacterial Psi55 synthase TruB. These results indicate that it is probable that (pfu)Cbf5 or (pfu)Pus10 (or both) is responsible for the introduction of pseudouridine at U55 in tRNAs in archaea. While we cannot unequivocally assign the function from our results, both possibilities represent unexpected functions of these proteins as discussed herein.


Subject(s)
Archaea/genetics , Pseudouridine/genetics , RNA, Transfer, Phe/chemistry , RNA, Transfer/genetics , Base Sequence , Molecular Sequence Data , Nucleic Acid Conformation
20.
Methods Enzymol ; 425: 55-101, 2007.
Article in English | MEDLINE | ID: mdl-17673079

ABSTRACT

The presence of modified ribonucleotides derived from adenosine, guanosine, cytidine, and uridine is a hallmark of almost all cellular RNA, and especially tRNA. The objective of this chapter is to describe a few simple methods that can be used to identify the presence or absence of a modified nucleotide in tRNA and to reveal the enzymatic activity of particular tRNA-modifying enzymes in vitro and in vivo. The procedures are based on analysis of prelabeled or postlabeled nucleotides (mainly with [(32)P] but also with [(35)S], [(14)C] or [(3)H]) generated after complete digestion with selected nucleases of modified tRNA isolated from cells or incubated in vitro with modifying enzyme(s). Nucleotides of the tRNA digests are separated by two-dimensional (2D) thin-layer chromatography on cellulose plates (TLC), which allows establishment of base composition and identification of the nearest neighbor nucleotide of a given modified nucleotide in the tRNA sequence. This chapter provides useful maps for identification of migration of approximately 70 modified nucleotides on TLC plates by use of two different chromatographic systems. The methods require only a few micrograms of purified tRNA and can be run at low cost in any laboratory.


Subject(s)
Enzymes/analysis , RNA Processing, Post-Transcriptional/physiology , RNA, Transfer/metabolism , Radioisotopes , Animals , Humans , RNA, Transfer/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL