Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(4): 114098, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38625793

ABSTRACT

Developing an effective mRNA therapeutic often requires maximizing protein output per delivered mRNA molecule. We previously found that coding sequence (CDS) design can substantially affect protein output, with mRNA variants containing more optimal codons and higher secondary structure yielding the highest protein outputs due to their slow rates of mRNA decay. Here, we demonstrate that CDS-dependent differences in translation initiation and elongation rates lead to differences in translation- and deadenylation-dependent mRNA decay rates, thus explaining the effect of CDS on mRNA half-life. Surprisingly, the most stable and highest-expressing mRNAs in our test set have modest initiation/elongation rates and ribosome loads, leading to minimal translation-dependent mRNA decay. These findings are of potential interest for optimization of protein output from therapeutic mRNAs, which may be achieved by attenuating rather than maximizing ribosome load.


Subject(s)
Protein Biosynthesis , RNA Stability , RNA, Messenger , Ribosomes , Ribosomes/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Humans
2.
J Biol Chem ; 299(10): 105246, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37703991

ABSTRACT

Long viewed as an intermediary in protein translation, there is a growing awareness that tRNAs are capable of myriad other biological functions linked to human health and disease. These emerging roles could be tapped to leverage tRNAs as diagnostic biomarkers, therapeutic targets, or even as novel medicines. Furthermore, the growing array of tRNA-derived fragments, which modulate an increasingly broad spectrum of cellular pathways, is expanding this opportunity. Together, these molecules offer drug developers the chance to modulate the impact of mutations and to alter cell homeostasis. Moreover, because a single therapeutic tRNA can facilitate readthrough of a genetic mutation shared across multiple genes, such medicines afford the opportunity to define patient populations not based on their clinical presentation or mutated gene but rather on the mutation itself. This approach could potentially transform the treatment of patients with rare and ultrarare diseases. In this review, we explore the diverse biology of tRNA and its fragments, examining the past and present challenges to provide a comprehensive understanding of the molecules and their therapeutic potential.

3.
J Biol Chem ; 293(39): 15021-15032, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30087118

ABSTRACT

N-Formylation of the Met-tRNAMet by the nuclearly encoded mitochondrial methionyl-tRNA formyltransferase (MTFMT) has been found to be a key determinant of protein synthesis initiation in mitochondria. In humans, mutations in the MTFMT gene result in Leigh syndrome, a progressive and severe neurometabolic disorder. However, the absolute requirement of formylation of Met-tRNAMet for protein synthesis in mammalian mitochondria is still debated. Here, we generated a Mtfmt-KO mouse fibroblast cell line and demonstrated that N-formylation of the first methionine via fMet-tRNAMet by MTFMT is not an absolute requirement for initiation of protein synthesis. However, it differentially affected the efficiency of synthesis of mtDNA-coded polypeptides. Lack of methionine N-formylation did not compromise the stability of these individual subunits but had a marked effect on the assembly and stability of the OXPHOS complexes I and IV and on their supercomplexes. In summary, N-formylation is not essential for mitochondrial protein synthesis but is critical for efficient synthesis of several mitochondrially encoded peptides and for OXPHOS complex stability and assembly into supercomplexes.


Subject(s)
Hydroxymethyl and Formyl Transferases/genetics , Methionine/genetics , Mitochondria/genetics , Protein Biosynthesis/genetics , Animals , DNA, Mitochondrial/genetics , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Mutation , Oxidative Phosphorylation , RNA, Transfer, Amino Acyl/genetics
4.
Nucleic Acid Ther ; 28(5): 285-296, 2018 10.
Article in English | MEDLINE | ID: mdl-30088967

ABSTRACT

The advent of therapeutic mRNAs significantly increases the possibilities of protein-based biologics beyond those that can be synthesized by recombinant technologies (eg, monoclonal antibodies, extracellular enzymes, and cytokines). In addition to their application in the areas of vaccine development, immune-oncology, and protein replacement therapies, one exciting possibility is to use therapeutic mRNAs to program undesired, diseased cells to synthesize a toxic intracellular protein, causing cells to self-destruct. For this approach to work, however, methods are needed to limit toxic protein expression to the intended cell type. Here, we show that inclusion of microRNA target sites in therapeutic mRNAs encoding apoptotic proteins, Caspase or PUMA, can prevent their expression in healthy hepatocytes while triggering apoptosis in hepatocellular carcinoma cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Caspases/genetics , Gene Expression Regulation, Neoplastic/genetics , HeLa Cells , Hepatocytes/metabolism , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , MicroRNAs/therapeutic use , Primary Cell Culture , Proto-Oncogene Proteins/genetics , RAW 264.7 Cells , RNA, Messenger/therapeutic use
5.
Biochem Biophys Res Commun ; 484(3): 612-617, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28153719

ABSTRACT

The product of the human C21orf57 (huYBEY) gene is predicted to be a homologue of the highly conserved YbeY proteins found in nearly all bacteria. We show that, like its bacterial and chloroplast counterparts, the HuYbeY protein is an RNase and that it retains sufficient function in common with bacterial YbeY proteins to partially suppress numerous aspects of the complex phenotype of an Escherichia coli ΔybeY mutant. Expression of HuYbeY in Saccharomyces cerevisiae, which lacks a YbeY homologue, results in a severe growth phenotype. This observation suggests that the function of HuYbeY in human cells is likely regulated through specific interactions with partner proteins similarly to the way YbeY is regulated in bacteria.


Subject(s)
Chloroplasts/chemistry , Chloroplasts/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Evolution, Molecular , Metalloproteins/chemistry , Metalloproteins/genetics , Ribonucleases/chemistry , Ribonucleases/genetics , Sequence Homology, Amino Acid , Amino Acid Sequence , Base Sequence , Conserved Sequence/genetics , Molecular Sequence Data
6.
mBio ; 7(6)2016 11 08.
Article in English | MEDLINE | ID: mdl-27834201

ABSTRACT

YbeY is part of a core set of RNases in Escherichia coli and other bacteria. This highly conserved endoribonuclease has been implicated in several important processes such as 16S rRNA 3' end maturation, 70S ribosome quality control, and regulation of mRNAs and small noncoding RNAs, thereby affecting cellular viability, stress tolerance, and pathogenic and symbiotic behavior of bacteria. Thus, YbeY likely interacts with numerous protein or RNA partners that are involved in various aspects of cellular physiology. Using a bacterial two-hybrid system, we identified several proteins that interact with YbeY, including ribosomal protein S11, the ribosome-associated GTPases Era and Der, YbeZ, and SpoT. In particular, the interaction of YbeY with S11 and Era provides insight into YbeY's involvement in the 16S rRNA maturation process. The three-way association between YbeY, S11, and Era suggests that YbeY is recruited to the ribosome where it could cleave the 17S rRNA precursor endonucleolytically at or near the 3' end maturation site. Analysis of YbeY missense mutants shows that a highly conserved beta-sheet in YbeY-and not amino acids known to be important for YbeY's RNase activity-functions as the interface between YbeY and S11. This protein-interacting interface of YbeY is needed for correct rRNA maturation and stress regulation, as missense mutants show significant phenotypic defects. Additionally, structure-based in silico prediction of putative interactions between YbeY and the Era-30S complex through protein docking agrees well with the in vivo results. IMPORTANCE: Ribosomes are ribonucleoprotein complexes responsible for a key cellular function, protein synthesis. Their assembly is a highly coordinated process of RNA cleavage, RNA posttranscriptional modification, RNA conformational changes, and protein-binding events. Many open questions remain after almost 5 decades of study, including which RNase is responsible for final processing of the 16S rRNA 3' end. The highly conserved RNase YbeY, belonging to a core set of RNases essential in many bacteria, was previously shown to participate in 16S rRNA processing and ribosome quality control. However, detailed mechanistic insight into YbeY's ribosome-associated function has remained elusive. This work provides the first evidence that YbeY is recruited to the ribosome through interaction with proteins involved in ribosome biogenesis (i.e., ribosomal protein S11, Era). In addition, we identified key residues of YbeY involved in the interaction with S11 and propose a possible binding mode of YbeY to the ribosome using in silico docking.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Metalloproteins/genetics , Metalloproteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal, 16S/metabolism , Ribosomes/metabolism , Stress, Physiological , Escherichia coli/genetics , Escherichia coli Proteins/isolation & purification , GTP-Binding Proteins/genetics , GTP-Binding Proteins/isolation & purification , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Bacterial , Molecular Docking Simulation , Mutation, Missense , Protein Binding , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , RNA, Bacterial/metabolism , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/isolation & purification , RNA-Binding Proteins/genetics , RNA-Binding Proteins/isolation & purification , RNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
7.
Nucleic Acids Res ; 44(20): 9965-9976, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27638883

ABSTRACT

Queuosine (Q) was discovered in the wobble position of a transfer RNA (tRNA) 47 years ago, yet the final biosynthetic enzyme responsible for Q-maturation, epoxyqueuosine (oQ) reductase (QueG), was only recently identified. QueG is a cobalamin (Cbl)-dependent, [4Fe-4S] cluster-containing protein that produces the hypermodified nucleoside Q in situ on four tRNAs. To understand how QueG is able to perform epoxide reduction, an unprecedented reaction for a Cbl-dependent enzyme, we have determined a series of high resolution structures of QueG from Bacillus subtilis Our structure of QueG bound to a tRNATyr anticodon stem loop shows how this enzyme uses a HEAT-like domain to recognize the appropriate anticodons and position the hypermodified nucleoside into the enzyme active site. We find Q bound directly above the Cbl, consistent with a reaction mechanism that involves the formation of a covalent Cbl-tRNA intermediate. Using protein film electrochemistry, we show that two [4Fe-4S] clusters adjacent to the Cbl have redox potentials in the range expected for Cbl reduction, suggesting how Cbl can be activated for nucleophilic attack on oQ. Together, these structural and electrochemical data inform our understanding of Cbl dependent nucleic acid modification.


Subject(s)
RNA Processing, Post-Transcriptional , RNA, Transfer/chemistry , RNA, Transfer/genetics , Vitamin B 12/chemistry , Anticodon , Bacillus subtilis/genetics , Hydrogen Bonding , Iron/chemistry , Models, Molecular , Molecular Conformation , Nucleic Acid Conformation , Nucleoside Q/analogs & derivatives , Nucleoside Q/chemistry , Protein Binding , RNA Stability , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Transfer/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism , Sulfur/chemistry , Vitamin B 12/metabolism
9.
Mol Microbiol ; 98(6): 1199-221, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26337258

ABSTRACT

Threonylcarbamoyladenosine (t(6)A) is a modified nucleoside universally conserved in tRNAs in all three kingdoms of life. The recently discovered genes for t(6)A synthesis, including tsaC and tsaD, are essential in model prokaryotes but not essential in yeast. These genes had been identified as antibacterial targets even before their functions were known. However, the molecular basis for this prokaryotic-specific essentiality has remained a mystery. Here, we show that t(6)A is a strong positive determinant for aminoacylation of tRNA by bacterial-type but not by eukaryotic-type isoleucyl-tRNA synthetases and might also be a determinant for the essential enzyme tRNA(Ile)-lysidine synthetase. We confirm that t(6)A is essential in Escherichia coli and a survey of genome-wide essentiality studies shows that genes for t(6)A synthesis are essential in most prokaryotes. This essentiality phenotype is not universal in Bacteria as t(6)A is dispensable in Deinococcus radiodurans, Thermus thermophilus, Synechocystis PCC6803 and Streptococcus mutans. Proteomic analysis of t(6)A(-) D. radiodurans strains revealed an induction of the proteotoxic stress response and identified genes whose translation is most affected by the absence of t(6)A in tRNAs. Thus, although t(6)A is universally conserved in tRNAs, its role in translation might vary greatly between organisms.


Subject(s)
Adenosine/analogs & derivatives , Deinococcus/genetics , Escherichia coli/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism , Adenosine/genetics , Adenosine/metabolism , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Aminoacylation/genetics , Conserved Sequence , Deinococcus/metabolism , Escherichia coli/metabolism , Molecular Sequence Data , Prokaryotic Cells , Proteomics , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Saccharomyces cerevisiae/genetics
10.
Nat Commun ; 6: 7520, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26138142

ABSTRACT

Dominant mutations in five tRNA synthetases cause Charcot-Marie-Tooth (CMT) neuropathy, suggesting that altered aminoacylation function underlies the disease. However, previous studies showed that loss of aminoacylation activity is not required to cause CMT. Here we present a Drosophila model for CMT with mutations in glycyl-tRNA synthetase (GARS). Expression of three CMT-mutant GARS proteins induces defects in motor performance and motor and sensory neuron morphology, and shortens lifespan. Mutant GARS proteins display normal subcellular localization but markedly reduce global protein synthesis in motor and sensory neurons, or when ubiquitously expressed in adults, as revealed by FUNCAT and BONCAT. Translational slowdown is not attributable to altered tRNA(Gly) aminoacylation, and cannot be rescued by Drosophila Gars overexpression, indicating a gain-of-toxic-function mechanism. Expression of CMT-mutant tyrosyl-tRNA synthetase also impairs translation, suggesting a common pathogenic mechanism. Finally, genetic reduction of translation is sufficient to induce CMT-like phenotypes, indicating a causal contribution of translational slowdown to CMT.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Glycine-tRNA Ligase/genetics , Motor Neurons/metabolism , Movement , Protein Biosynthesis/genetics , Sensory Receptor Cells/metabolism , Tyrosine-tRNA Ligase/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila , Humans , Life Expectancy , Motor Neurons/pathology , Mutagenesis, Site-Directed , Mutation , Neuromuscular Junction/pathology , Phenotype , Sensory Receptor Cells/pathology
11.
Proc Natl Acad Sci U S A ; 112(19): 6015-20, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25918386

ABSTRACT

Bacterial strains carrying nonsense suppressor tRNA genes played a crucial role in early work on bacterial and bacterial viral genetics. In eukaryotes as well, suppressor tRNAs have played important roles in the genetic analysis of yeast and worms. Surprisingly, little is known about genetic suppression in archaea, and there has been no characterization of suppressor tRNAs or identification of nonsense mutations in any of the archaeal genes. Here, we show, using the ß-gal gene as a reporter, that amber, ochre, and opal suppressors derived from the serine and tyrosine tRNAs of the archaeon Haloferax volcanii are active in suppression of their corresponding stop codons. Using a promoter for tRNA expression regulated by tryptophan, we also show inducible and regulatable suppression of all three stop codons in H. volcanii. Additionally, transformation of a ΔpyrE2 H. volcanii strain with plasmids carrying the genes for a pyrE2 amber mutant and the serine amber suppressor tRNA yielded transformants that grow on agar plates lacking uracil. Thus, an auxotrophic amber mutation in the pyrE2 gene can be complemented by expression of the amber suppressor tRNA. These results pave the way for generating archaeal strains carrying inducible suppressor tRNA genes on the chromosome and their use in archaeal and archaeviral genetics. We also provide possible explanations for why suppressor tRNAs have not been identified in archaea.


Subject(s)
Archaea/genetics , Codon, Terminator , Haloferax volcanii/genetics , RNA, Transfer/metabolism , Suppression, Genetic , Archaea/metabolism , Base Sequence , Codon, Nonsense , Escherichia coli/metabolism , Genes, Suppressor , Haloferax volcanii/metabolism , Molecular Sequence Data , Novobiocin/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Serine/chemistry , Thymidine/chemistry , Tryptophan/chemistry , Uracil/chemistry , beta-Galactosidase/metabolism
12.
J Biol Chem ; 289(47): 32729-41, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25288793

ABSTRACT

N-Formylation of initiator methionyl-tRNA (Met-tRNA(Met)) by methionyl-tRNA formyltransferase (MTF) is important for translation initiation in bacteria, mitochondria, and chloroplasts. Unlike all other translation systems, the metazoan mitochondrial system is unique in using a single methionine tRNA (tRNA(Met)) for both initiation and elongation. A portion of Met-tRNA(Met) is formylated for initiation, whereas the remainder is used for elongation. Recently, we showed that compound heterozygous mutations within the nuclear gene encoding human mitochondrial MTF (mt-MTF) significantly reduced mitochondrial translation efficiency, leading to combined oxidative phosphorylation deficiency and Leigh syndrome in two unrelated patients. Patient P1 has a stop codon mutation in one of the MTF genes and an S209L mutation in the other MTF gene. P2 has a S125L mutation in one of the MTF genes and the same S209L mutation as P1 in the other MTF gene. Here, we have investigated the effect of mutations at Ser-125 and Ser-209 on activities of human mt-MTF and of the corresponding mutations, Ala-89 or Ala-172, respectively, on activities of Escherichia coli MTF. The S125L mutant has 653-fold lower activity, whereas the S209L mutant has 36-fold lower activity. Thus, both patients depend upon residual activity of the S209L mutant to support low levels of mitochondrial protein synthesis. We discuss the implications of these and other results for whether the effect of the S209L mutation on mitochondrial translational efficiency is due to reduced activity of the mutant mt-MTF and/or reduced levels of the mutant mt-MTF.


Subject(s)
Hydroxymethyl and Formyl Transferases/genetics , Leigh Disease/genetics , Mitochondrial Proteins/genetics , Mutation , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Hydroxymethyl and Formyl Transferases/metabolism , Immunoblotting , Leigh Disease/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Protein Biosynthesis/genetics , RNA, Transfer, Met/genetics , RNA, Transfer, Met/metabolism , Sequence Homology, Amino Acid , Serine/genetics , Serine/metabolism
13.
PLoS Pathog ; 10(6): e1004175, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24901994

ABSTRACT

YbeY, a highly conserved protein, is an RNase in E. coli and plays key roles in both processing of the critical 3' end of 16 S rRNA and in 70 S ribosome quality control under stress. These central roles account for YbeY's inclusion in the postulated minimal bacterial genome. However, YbeY is not essential in E. coli although loss of ybeY severely sensitizes it to multiple physiological stresses. Here, we show that YbeY is an essential endoribonuclease in Vibrio cholerae and is crucial for virulence, stress regulation, RNA processing and ribosome quality control, and is part of a core set of RNases essential in most representative pathogens. To understand its function, we analyzed the rRNA and ribosome profiles of a V. cholerae strain partially depleted for YbeY and other RNase mutants associated with 16 S rRNA processing; our results demonstrate that YbeY is also crucial for 16 S rRNA 3' end maturation in V. cholerae and that its depletion impedes subunit assembly into 70 S ribosomes. YbeY's importance to V. cholerae pathogenesis was demonstrated by the complete loss of mice colonization and biofilm formation, reduced cholera toxin production, and altered expression levels of virulence-associated small RNAs of a V. cholerae strain partially depleted for YbeY. Notably, the ybeY genes of several distantly related pathogens can fully complement an E. coli ΔybeY strain under various stress conditions, demonstrating the high conservation of YbeY's activity in stress regulation. Taken together, this work provides the first comprehensive exploration of YbeY's physiological role in a human pathogen, showing its conserved function across species in essential cellular processes.


Subject(s)
Bacterial Proteins/metabolism , Endoribonucleases/metabolism , RNA 3' End Processing , RNA, Bacterial/metabolism , RNA, Ribosomal/metabolism , Stress, Physiological , Vibrio cholerae/enzymology , Amino Acid Sequence , Animals , Animals, Newborn , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biofilms/growth & development , Cholera/enzymology , Cholera/immunology , Cholera/metabolism , Cholera/microbiology , Cholera Toxin/biosynthesis , Conserved Sequence , Endoribonucleases/chemistry , Endoribonucleases/genetics , Gene Expression Regulation, Bacterial , Immunity, Mucosal , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mutation , Phylogeny , Vibrio cholerae/immunology , Vibrio cholerae/pathogenicity , Vibrio cholerae/physiology , Virulence , Virulence Factors/biosynthesis
14.
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
15.
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
16.
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
17.
Mol Cell ; 49(3): 427-38, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23273979

ABSTRACT

Quality control of ribosomes is critical for cellular function since protein mistranslation leads to severe physiological consequences. We report evidence of a previously unrecognized ribosome quality control system in bacteria that operates at the level of 70S to remove defective ribosomes. YbeY, a previously unidentified endoribonuclease, and the exonuclease RNase R act together by a process mediated specifically by the 30S ribosomal subunit, to degrade defective 70S ribosomes but not properly matured 70S ribosomes or individual subunits. Furthermore, there is essentially no fully matured 16S rRNA in a ΔybeY mutant at 45°C, making YbeY the only endoribonuclease to be implicated in the critically important processing of the 16S rRNA 3' terminus. These key roles in ribosome quality control and maturation indicate why YbeY is a member of the minimal bacterial gene set and suggest that it could be a potential target for antibacterial drugs.


Subject(s)
Conserved Sequence , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Metalloproteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal, 16S/metabolism , Ribosomes/metabolism , Arginine/metabolism , Base Sequence , Escherichia coli/drug effects , Escherichia coli Proteins/chemistry , Exoribonucleases/metabolism , Histidine/metabolism , Hot Temperature , Metalloproteins/chemistry , Metals/pharmacology , Models, Biological , Molecular Sequence Data , Mutation/genetics , Protein Biosynthesis/drug effects , RNA Processing, Post-Transcriptional/drug effects , RNA, Ribosomal, 16S/genetics , Ribosome Subunits, Small, Bacterial/metabolism , Ribosomes/drug effects , Stress, Physiological/drug effects , Substrate Specificity/drug effects
18.
Cell Metab ; 14(3): 428-34, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21907147

ABSTRACT

The metazoan mitochondrial translation machinery is unusual in having a single tRNA(Met) that fulfills the dual role of the initiator and elongator tRNA(Met). A portion of the Met-tRNA(Met) pool is formylated by mitochondrial methionyl-tRNA formyltransferase (MTFMT) to generate N-formylmethionine-tRNA(Met) (fMet-tRNA(met)), which is used for translation initiation; however, the requirement of formylation for initiation in human mitochondria is still under debate. Using targeted sequencing of the mtDNA and nuclear exons encoding the mitochondrial proteome (MitoExome), we identified compound heterozygous mutations in MTFMT in two unrelated children presenting with Leigh syndrome and combined OXPHOS deficiency. Patient fibroblasts exhibit severe defects in mitochondrial translation that can be rescued by exogenous expression of MTFMT. Furthermore, patient fibroblasts have dramatically reduced fMet-tRNA(Met) levels and an abnormal formylation profile of mitochondrially translated COX1. Our findings demonstrate that MTFMT is critical for efficient human mitochondrial translation and reveal a human disorder of Met-tRNA(Met) formylation.


Subject(s)
Cyclooxygenase 1/metabolism , DNA, Mitochondrial/chemistry , Fibroblasts/metabolism , Leigh Disease/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Protein Biosynthesis , RNA, Transfer, Met/metabolism , Cells, Cultured , Child , Cyclooxygenase 1/genetics , DNA, Mitochondrial/genetics , Fibroblasts/pathology , Heterozygote , Humans , Hydroxymethyl and Formyl Transferases , Immunoblotting , Leigh Disease/metabolism , Leigh Disease/pathology , Lentivirus , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mutation , Protein Biosynthesis/genetics , Sequence Analysis, DNA , Transduction, Genetic , Virion
19.
Mol Microbiol ; 78(2): 506-18, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20807199

ABSTRACT

The UPF0054 protein family is highly conserved with homologues present in nearly every sequenced bacterium. In some bacteria, the respective gene is essential, while in others its loss results in a highly pleiotropic phenotype. Despite detailed structural studies, a cellular role for this protein family has remained unknown. We report here that deletion of the Escherichia coli homologue, YbeY, causes striking defects that affect ribosome activity, translational fidelity and ribosome assembly. Mapping of 16S, 23S and 5S rRNA termini reveals that YbeY influences the maturation of all three rRNAs, with a particularly strong effect on maturation at both the 5'- and 3'-ends of 16S rRNA as well as maturation of the 5'-termini of 23S and 5S rRNAs. Furthermore, we demonstrate strong genetic interactions between ybeY and rnc (encoding RNase III), ybeY and rnr (encoding RNase R), and ybeY and pnp (encoding PNPase), further suggesting a role for YbeY in rRNA maturation. Mutation of highly conserved amino acids in YbeY, allowed the identification of two residues (H114, R59) that were found to have a significant effect in vivo. We discuss the implications of these findings for rRNA maturation and ribosome assembly in bacteria.


Subject(s)
Escherichia coli Proteins/metabolism , Metalloproteins/metabolism , RNA, Bacterial/metabolism , RNA, Ribosomal/metabolism , Amino Acid Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gene Deletion , Metalloproteins/genetics , Molecular Sequence Data , Prokaryotic Initiation Factors/metabolism , Protein Binding , Ribosomes/metabolism , Sequence Alignment
20.
Proc Natl Acad Sci U S A ; 107(7): 2872-7, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133752

ABSTRACT

Modification of the cytidine in the first anticodon position of the AUA decoding tRNA(Ile) (tRNA2(Ile)) of bacteria and archaea is essential for this tRNA to read the isoleucine codon AUA and to differentiate between AUA and the methionine codon AUG. To identify the modified cytidine in archaea, we have purified this tRNA species from Haloarcula marismortui, established its codon reading properties, used liquid chromatography-mass spectrometry (LC-MS) to map RNase A and T1 digestion products onto the tRNA, and used LC-MS/MS to sequence the oligonucleotides in RNase A digests. These analyses revealed that the modification of cytidine in the anticodon of tRNA2(Ile) adds 112 mass units to its molecular mass and makes the glycosidic bond unusually labile during mass spectral analyses. Accurate mass LC-MS and LC-MS/MS analysis of total nucleoside digests of the tRNA2(Ile) demonstrated the absence in the modified cytidine of the C2-oxo group and its replacement by agmatine (decarboxy-arginine) through a secondary amine linkage. We propose the name agmatidine, abbreviation C(+), for this modified cytidine. Agmatidine is also present in Methanococcus maripaludis tRNA2(Ile) and in Sulfolobus solfataricus total tRNA, indicating its probable occurrence in the AUA decoding tRNA(Ile) of euryarchaea and crenarchaea. The identification of agmatidine shows that bacteria and archaea have developed very similar strategies for reading the isoleucine codon AUA while discriminating against the methionine codon AUG.


Subject(s)
Anticodon/genetics , Base Pairing/genetics , Cytidine/chemistry , Haloarcula marismortui/chemistry , RNA, Transfer, Ile/chemistry , Agmatine/chemistry , Chromatography, Liquid , Methanococcus/chemistry , Molecular Structure , RNA, Transfer, Ile/genetics , Sulfolobus solfataricus/chemistry , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...