Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Eur J Med Chem ; 187: 111941, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31821989

ABSTRACT

Threonyl-tRNA synthetase (ThrRS) is a key member of the aminoacyl-tRNA synthetase (aaRS) family that plays essential roles in protein biosynthesis, and ThrRS inhibitors have potential in the therapy of multiple diseases, such as microbial infections and cancers. Based on a unique tRNA-amino acid dual-site inhibitory mechanism identified recently with the herb-derived prolyl-tRNA synthetase (ProRS) inhibitor halofuginone (HF), a series of compounds have been designed and synthesized by employing a fragment-based target hopping approach to simultaneously target the tRNAThr and l-threonine binding pockets of ThrRS. Among them, compound 30d showed an IC50 value of 1.4 µM against Salmonella enterica ThrRS (SeThrRS) and MIC values of 16-32 µg/mL against the tested bacterial strains. The cocrystal structure of SeThrRS in complex with 30d was determined at high resolution, revealing that 30d simultaneously occupies both binding pockets for the nucleotide A76 of tRNAThr and l-threonine in an ATP-independent manner, a novel mechanism compared to all other reported ThrRS inhibitors. Our study provides a new class of ThrRS inhibitors, and more importantly, it presents the first experimental evidence that the tRNA-amino acid dual-site inhibitory mechanism could apply to other aaRSs beyond ProRS, thus providing great opportunities for designing new mechanistic inhibitors for aaRS-based therapeutics.


Subject(s)
Drug Discovery , RNA, Transfer, Amino Acid-Specific/pharmacology , Threonine-tRNA Ligase/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , RNA, Transfer, Amino Acid-Specific/chemical synthesis , RNA, Transfer, Amino Acid-Specific/chemistry , Salmonella enterica/enzymology , Structure-Activity Relationship , Threonine-tRNA Ligase/metabolism
2.
Cells ; 8(6)2019 06 11.
Article in English | MEDLINE | ID: mdl-31212706

ABSTRACT

The translation of selenoprotein mRNAs involves a non-canonical ribosomal event in which an in-frame UGA is recoded as a selenocysteine (Sec) codon instead of being read as a stop codon. The recoding machinery is centered around two dedicated RNA components: The selenocysteine insertion sequence (SECIS) located in the 3' UTR of the mRNA and the selenocysteine-tRNA (Sec-tRNA[Ser]Sec). This translational UGA-selenocysteine recoding event by the ribosome is a limiting stage of selenoprotein expression. Its efficiency is controlled by the SECIS, the Sec-tRNA[Ser]Sec and their interacting protein partners. In the present work, we used a recently developed CRISPR strategy based on murine leukemia virus-like particles (VLPs) loaded with Cas9-sgRNA ribonucleoproteins to inactivate the Sec-tRNA[Ser]Sec gene in human cell lines. We showed that these CRISPR-Cas9-VLPs were able to induce efficient genome-editing in Hek293, HepG2, HaCaT, HAP1, HeLa, and LNCaP cell lines and this caused a robust reduction of selenoprotein expression. The alteration of selenoprotein expression was the direct consequence of lower levels of Sec-tRNA[Ser]Sec and thus a decrease in translational recoding efficiency of the ribosome. This novel strategy opens many possibilities to study the impact of selenoprotein deficiency in hard-to-transfect cells, since these CRISPR-Cas9-VLPs have a wide tropism.


Subject(s)
CRISPR-Cas Systems/genetics , Codon, Terminator/genetics , RNA, Transfer, Amino Acid-Specific/genetics , Ribosomes/metabolism , Selenocysteine/metabolism , Virion/metabolism , Base Sequence , Gene Editing , HEK293 Cells , HeLa Cells , Humans , INDEL Mutation/genetics , Nucleic Acid Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer, Amino Acid-Specific/chemistry , Selenium/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism
3.
FEBS Lett ; 592(22): 3759-3768, 2018 11.
Article in English | MEDLINE | ID: mdl-30317559

ABSTRACT

Selenocysteine (Sec) lacks a cognate aminoacyl-tRNA synthetase. Instead, seryl-tRNA synthetase (SerRS) produces Ser-tRNASec , which is subsequently converted by selenocysteine synthase to Sec-tRNASec . Escherichia coli SerRS serylates tRNASec poorly; this may hinder efficient production of designer selenoproteins in vivo. Guided by structural modelling and selection for chloramphenicol acetyltransferase activity, we evolved three SerRS variants capable of improved Ser-tRNASec synthesis. They display 10-, 8-, and 4-fold increased kcat /KM values compared to wild-type SerRS using synthetic tRNASec species as substrates. The enzyme variants also facilitate in vivo read-through of a UAG codon in the position of the critical serine146 of chloramphenicol acetyltransferase. These results indicate that the naturally evolved SerRS is capable of further evolution for increased recognition of a specific tRNA isoacceptor.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , RNA, Transfer, Amino Acid-Specific/genetics , RNA, Transfer, Ser/genetics , Serine-tRNA Ligase/genetics , Base Sequence , Codon, Terminator/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Kinetics , Models, Molecular , Mutation , Nucleic Acid Conformation , Protein Domains , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Amino Acid-Specific/metabolism , RNA, Transfer, Ser/chemistry , RNA, Transfer, Ser/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism , Serine/genetics , Serine/metabolism , Serine-tRNA Ligase/chemistry , Serine-tRNA Ligase/metabolism , Substrate Specificity
4.
Methods Mol Biol ; 1661: 43-60, 2018.
Article in English | MEDLINE | ID: mdl-28917036

ABSTRACT

The selenocysteine (Sec) tRNA[Ser]Sec population consists of two isoforms that differ from each other by a single 2'-O-methylribosyl moiety at position 34 (Um34). These two isoforms, which are encoded in a single gene, Trsp, and modified posttranscriptionally, are involved individually in the synthesis of two subclasses of selenoproteins, designated housekeeping and stress-related selenoproteins. Techniques used in obtaining these isoforms for their characterization include extraction of RNA from mammalian cells and tissues, purifying the tRNA[Ser]Sec population by one or more procedures, and finally resolving the two isoforms from each other. Since some of the older techniques for isolating tRNA[Ser]Sec and resolving the isoforms are used in only a few laboratories, these procedures will be discussed briefly and references provided for more detailed information, while the more recently developed procedures are discussed in detail. In addition, a novel technique that was developed in sequencing tRNA[Ser]Sec for identifying their occurrence in other organisms is also presented.


Subject(s)
RNA, Transfer, Amino Acid-Specific/genetics , Selenoproteins/genetics , Animals , Blotting, Northern , Chromatography, Affinity , Chromatography, Reverse-Phase , Humans , Isotope Labeling , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Transfer, Amino Acid-Specific/chemistry , Selenium Radioisotopes , Selenoproteins/chemistry , Selenoproteins/isolation & purification , Sequence Analysis, RNA
5.
RNA ; 23(11): 1685-1699, 2017 11.
Article in English | MEDLINE | ID: mdl-28808125

ABSTRACT

Seryl-tRNA synthetase (SerRS) attaches L-serine to the cognate serine tRNA (tRNASer) and the noncognate selenocysteine tRNA (tRNASec). The latter activity initiates the anabolic cycle of selenocysteine (Sec), proper decoding of an in-frame Sec UGA codon, and synthesis of selenoproteins across all domains of life. While the accuracy of SerRS is important for overall proteome integrity, it is its substrate promiscuity that is vital for the integrity of the selenoproteome. This raises a question as to what elements in the two tRNA species, harboring different anticodon sequences and adopting distinct folds, facilitate aminoacylation by a common aminoacyl-tRNA synthetase. We sought to answer this question by analyzing the ability of human cytosolic SerRS to bind and act on tRNASer, tRNASec, and 10 mutant and chimeric constructs in which elements of tRNASer were transposed onto tRNASec We show that human SerRS only subtly prefers tRNASer to tRNASec, and that discrimination occurs at the level of the serylation reaction. Surprisingly, the tRNA mutants predicted to adopt either the 7/5 or 8/5 fold are poor SerRS substrates. In contrast, shortening of the acceptor arm of tRNASec by a single base pair yields an improved SerRS substrate that adopts an 8/4 fold. We suggest that an optimal tertiary arrangement of structural elements within tRNASec and tRNASer dictate their utility for serylation. We also speculate that the extended acceptor-TΨC arm of tRNASec evolved as a compromise for productive binding to SerRS while remaining the major recognition element for other enzymes involved in Sec and selenoprotein synthesis.


Subject(s)
RNA, Transfer, Amino Acid-Specific/metabolism , RNA, Transfer, Ser/metabolism , Serine-tRNA Ligase/metabolism , Base Sequence , Binding Sites , Cytosol/enzymology , Humans , Kinetics , Models, Molecular , Mutagenesis , Nucleic Acid Conformation , RNA Folding , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Amino Acid-Specific/genetics , RNA, Transfer, Ser/chemistry , RNA, Transfer, Ser/genetics , Substrate Specificity
6.
Nucleic Acids Res ; 45(5): 2776-2785, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28076288

ABSTRACT

We report the identification of novel tRNA species with 12-base pair amino-acid acceptor branches composed of longer acceptor stem and shorter T-stem. While canonical tRNAs have a 7/5 configuration of the branch, the novel tRNAs have either 8/4 or 9/3 structure. They were found during the search for selenocysteine tRNAs in terabytes of genome, metagenome and metatranscriptome sequences. Certain bacteria and their phages employ the 8/4 structure for serine and histidine tRNAs, while minor cysteine and selenocysteine tRNA species may have a modified 8/4 structure with one bulge nucleotide. In Acidobacteria, tRNAs with 8/4 and 9/3 structures may function as missense and nonsense suppressor tRNAs and/or regulatory noncoding RNAs. In δ-proteobacteria, an additional cysteine tRNA with an 8/4 structure mimics selenocysteine tRNA and may function as opal suppressor. We examined the potential translation function of suppressor tRNA species in Escherichia coli; tRNAs with 8/4 or 9/3 structures efficiently inserted serine, alanine and cysteine in response to stop and sense codons, depending on the identity element and anticodon sequence of the tRNA. These findings expand our view of how tRNA, and possibly the genetic code, is diversified in nature.


Subject(s)
RNA, Bacterial/chemistry , RNA, Transfer/chemistry , Anticodon , Bacteria/genetics , Bacterial Toxins/genetics , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Cys/chemistry , RNA, Transfer, Cys/metabolism
7.
Methods ; 113: 46-55, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27777026

ABSTRACT

In addition to their role in correctly attaching specific amino acids to cognate tRNAs, aminoacyl-tRNA synthetases (aaRS) have been found to possess many alternative functions and often bind to and act on other nucleic acids. In contrast to the well-defined 3D structure of tRNA, the structures of many of the other RNAs recognized by aaRSs have not been solved. Despite advances in the use of X-ray crystallography (XRC), nuclear magnetic resonance (NMR) spectroscopy and cryo-electron microscopy (cryo-EM) for structural characterization of biomolecules, significant challenges to solving RNA structures still exist. Recently, small-angle X-ray scattering (SAXS) has been increasingly employed to characterize the 3D structures of RNAs and RNA-protein complexes. SAXS is capable of providing low-resolution tertiary structure information under physiological conditions and with less intensive sample preparation and data analysis requirements than XRC, NMR and cryo-EM. In this article, we describe best practices involved in the process of RNA and RNA-protein sample preparation, SAXS data collection, data analysis, and structural model building.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Escherichia coli/genetics , RNA Folding , RNA, Transfer, Amino Acid-Specific/chemistry , Transfer RNA Aminoacylation , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/genetics , Chromatography, Gel , Escherichia coli/metabolism , Models, Molecular , Native Polyacrylamide Gel Electrophoresis , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , RNA, Transfer, Amino Acid-Specific/genetics , RNA, Transfer, Amino Acid-Specific/metabolism , Scattering, Small Angle , X-Ray Diffraction
8.
Nature ; 540(7631): 80-85, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27842381

ABSTRACT

In all domains of life, selenocysteine (Sec) is delivered to the ribosome by selenocysteine-specific tRNA (tRNASec) with the help of a specialized translation factor, SelB in bacteria. Sec-tRNASec recodes a UGA stop codon next to a downstream mRNA stem-loop. Here we present the structures of six intermediates on the pathway of UGA recoding in Escherichia coli by single-particle cryo-electron microscopy. The structures explain the specificity of Sec-tRNASec binding by SelB and show large-scale rearrangements of Sec-tRNASec. Upon initial binding of SelB-Sec-tRNASec to the ribosome and codon reading, the 30S subunit adopts an open conformation with Sec-tRNASec covering the sarcin-ricin loop (SRL) on the 50S subunit. Subsequent codon recognition results in a local closure of the decoding site, which moves Sec-tRNASec away from the SRL and triggers a global closure of the 30S subunit shoulder domain. As a consequence, SelB docks on the SRL, activating the GTPase of SelB. These results reveal how codon recognition triggers GTPase activation in translational GTPases.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli/metabolism , GTP Phosphohydrolases/metabolism , Ribosomes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Binding Sites , Codon, Terminator/chemistry , Codon, Terminator/genetics , Codon, Terminator/metabolism , Cryoelectron Microscopy , Endoribonucleases/metabolism , Enzyme Activation , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/ultrastructure , Fungal Proteins/metabolism , GTP Phosphohydrolases/ultrastructure , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Biosynthesis , Protein Domains , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Amino Acid-Specific/genetics , RNA, Transfer, Amino Acid-Specific/metabolism , RNA, Transfer, Amino Acid-Specific/ultrastructure , Ribosome Subunits, Large, Bacterial/chemistry , Ribosome Subunits, Large, Bacterial/metabolism , Ribosome Subunits, Large, Bacterial/ultrastructure , Ribosome Subunits, Small, Bacterial/chemistry , Ribosome Subunits, Small, Bacterial/metabolism , Ribosome Subunits, Small, Bacterial/ultrastructure , Ribosomes/chemistry , Ribosomes/enzymology , Ribosomes/ultrastructure , Ricin/metabolism , Selenocysteine/metabolism
9.
Trends Biochem Sci ; 41(9): 798-814, 2016 09.
Article in English | MEDLINE | ID: mdl-27372401

ABSTRACT

The fidelity of translation depends strongly on the selection of the correct aminoacyl-tRNA that is complementary to the mRNA codon present in the ribosomal decoding center. The ribosome occasionally makes mistakes by selecting the wrong substrate from the pool of aminoacyl-tRNAs. Here, we summarize recent structural advances that may help to clarify the origin of missense errors that occur during decoding. These developments suggest that discrimination between tRNAs is based primarily on steric complementarity and shape acceptance rather than on the number of hydrogen bonds between the molding of the decoding center and the codon-anticodon duplex. They strengthen the hypothesis that spatial mimicry, due either to base tautomerism or ionization, drives infidelity in ribosomal translation.


Subject(s)
Genetic Code/genetics , Mutation, Missense , Protein Biosynthesis , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Amino Acid-Specific/metabolism , Anticodon/genetics , Codon/genetics , Hydrogen Bonding , Mutation, Missense/genetics , Protein Biosynthesis/genetics , RNA, Transfer, Amino Acid-Specific/genetics , Ribosomes/chemistry , Ribosomes/metabolism
10.
Nucleic Acids Res ; 43(21): 10534-45, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26433229

ABSTRACT

Selenocysteine (Sec) is found in the catalytic centers of many selenoproteins and plays important roles in living organisms. Malfunctions of selenoproteins lead to various human disorders including cancer. Known as the 21st amino acid, the biosynthesis of Sec involves unusual pathways consisting of several stages. While the later stages of the pathways are well elucidated, the molecular basis of the first stage-the serylation of Sec-specific tRNA (tRNA(Sec)) catalyzed by seryl-tRNA synthetase (SerRS)-is unclear. Here we present two cocrystal structures of human SerRS bound with tRNA(Sec) in different stoichiometry and confirm the formation of both complexes in solution by various characterization techniques. We discovered that the enzyme mainly recognizes the backbone of the long variable arm of tRNA(Sec) with few base-specific contacts. The N-terminal coiled-coil region works like a long-range lever to precisely direct tRNA 3' end to the other protein subunit for aminoacylation in a conformation-dependent manner. Restraints of the flexibility of the coiled-coil greatly reduce serylation efficiencies. Lastly, modeling studies suggest that the local differences present in the D- and T-regions as well as the characteristic U20:G19:C56 base triple in tRNA(Sec) may allow SerRS to distinguish tRNA(Sec) from closely related tRNA(Ser) substrate.


Subject(s)
RNA, Transfer, Amino Acid-Specific/chemistry , Selenocysteine/biosynthesis , Serine-tRNA Ligase/chemistry , Humans , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , RNA, Transfer, Amino Acid-Specific/metabolism , Serine-tRNA Ligase/metabolism , Substrate Specificity
11.
Genome Biol Evol ; 7(9): 2692-704, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26342139

ABSTRACT

By competing for the acquisition of essential nutrients, Helicobacter pylori has the unique ability to persist in the human stomach, also causing nutritional insufficiencies in the host. Although the H. pylori genome apparently encodes selenocysteine synthase (SelA, HP1513), a key pyridoxal phosphate (PLP)-dependent enzyme for the incorporation of selenium into bacterial proteins, nothing is known about the use of this essential element in protein synthesis by this pathogen. We analyzed the evolution of the complete machinery for incorporation of selenium into proteins and the selenoproteome of several H. pylori strains and related Epsilonproteobacteria. Our searches identified the presence of selenoproteins-including the previously unknown DUF466 family-in various Epsilonproteobacteria, but not in H. pylori. We found that a complete system for selenocysteine incorporation was present in the Helicobacteriaceae ancestor and has been recently lost before the split of Helicobacter acinonychis and H. pylori. Our results indicate that H. pylori, at variance with other gastric and enterohepatic Helicobacter, does not use selenocysteine in protein synthesis and does not use selenium for tRNA wobble base modification. However, selA has survived as a functional gene, having lost the domain for the binding of selenocysteine tRNA, but maintaining the ability to bind the PLP cofactor. The evolutionary modifications described for the SelA protein of H. pylori find parallels in other bacterial and archaeal species, suggesting that an alternative enzymatic function is hidden in many proteins annotated as selenocysteinyl-tRNA synthase.


Subject(s)
Epsilonproteobacteria/genetics , Evolution, Molecular , Helicobacter pylori/enzymology , Selenoproteins/genetics , Transferases/genetics , Amino Acid Sequence , Conserved Sequence , Helicobacter pylori/genetics , Proteome/genetics , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Amino Acid-Specific/genetics , Sequence Alignment , Transferases/chemistry
12.
Nucleic Acids Res ; 43(18): 9028-38, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26304550

ABSTRACT

Selenocysteine (Sec), the 21(st) amino acid in translation, uses its specific tRNA (tRNA(Sec)) to recognize the UGA codon. The Sec-specific elongation factor SelB brings the selenocysteinyl-tRNA(Sec) (Sec-tRNA(Sec)) to the ribosome, dependent on both an in-frame UGA and a Sec-insertion sequence (SECIS) in the mRNA. The bacterial SelB binds mRNA through its C-terminal region, for which crystal structures have been reported. In this study, we determined the crystal structure of the full-length SelB from the bacterium Aquifex aeolicus, in complex with a GTP analog, at 3.2-Å resolution. SelB consists of three EF-Tu-like domains (D1-3), followed by four winged-helix domains (WHD1-4). The spacer region, connecting the N- and C-terminal halves, fixes the position of WHD1 relative to D3. The binding site for the Sec moiety of Sec-tRNA(Sec) is located on the interface between D1 and D2, where a cysteine molecule from the crystallization solution is coordinated by Arg residues, which may mimic Sec binding. The Sec-binding site is smaller and more exposed than the corresponding site of EF-Tu. Complex models of Sec-tRNA(Sec), SECIS RNA, and the 70S ribosome suggest that the unique secondary structure of tRNA(Sec) allows SelB to specifically recognize tRNA(Sec) and characteristically place it at the ribosomal A-site.


Subject(s)
Bacterial Proteins/chemistry , Peptide Elongation Factors/chemistry , RNA, Transfer, Amino Acid-Specific/chemistry , Selenocysteine/chemistry , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Models, Molecular , Peptide Elongation Factors/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Transfer, Amino Acid-Specific/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Selenocysteine/metabolism
13.
FEBS Lett ; 589(17): 2194-9, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26160755

ABSTRACT

Incorporation of selenocysteine (Sec) in bacteria requires a UGA codon that is reassigned to Sec by the Sec-specific elongation factor SelB and a conserved mRNA motif (SECIS element). These requirements severely restrict the engineering of selenoproteins. Earlier, a synthetic tRNASec was reported that allowed canonical Sec incorporation by EF-Tu; however, serine misincorporation limited its scope. We report a superior tRNASec variant (tRNAUTuX) that facilitates EF-Tu dependent stoichiometric Sec insertion in response to UAG both in vivo in Escherichia coli and in vitro in a cellfree protein synthesis system. We also demonstrate recoding of several sense codons in a SelB supplemented cell-free system. These advances in Sec incorporation will aid rational design and directed evolution of selenoproteins.


Subject(s)
Peptide Elongation Factor Tu/metabolism , Protein Biosynthesis , RNA, Transfer, Amino Acid-Specific/metabolism , Selenocysteine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Codon, Terminator/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Amino Acid-Specific/genetics , RNA, Transfer, Amino Acyl/chemistry , RNA, Transfer, Amino Acyl/genetics , RNA, Transfer, Amino Acyl/metabolism , Selenoproteins/genetics , Selenoproteins/metabolism
14.
Orig Life Evol Biosph ; 44(4): 269-77, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25585798

ABSTRACT

The self-referential genetic encoding starts with glycine and serine, in the realm of one-carbon units of metabolism. It is proposed that the prototRNA dimer-directed mechanism of protein synthesis and encoding promotes a sink dynamics that corresponds to the driving 'force' for the fixation of the supporting metabolic pathways. A succession of processes is delineated, ending up in reproduction, which accomplished the function of reinforcing the protein synthesis metabolic sink mechanism.


Subject(s)
Genetic Code , Glycine/chemistry , Models, Genetic , Origin of Life , RNA, Transfer, Amino Acid-Specific/chemistry , Serine/chemistry , Biological Evolution , Carbon/metabolism , Cell Division , Dimerization , Glycine/metabolism , Glycine Hydroxymethyltransferase/chemistry , Glycine Hydroxymethyltransferase/metabolism , Metabolic Networks and Pathways , Protein Biosynthesis , RNA, Transfer, Amino Acid-Specific/metabolism , Reproduction , Serine/metabolism
15.
Orig Life Evol Biosph ; 44(4): 287-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25585805

ABSTRACT

The origin of a genetic code made it possible to create ordered sequences of amino acids. In this article we provide two perspectives on code origin by carrying out simulations of code-sequence coevolution in finite populations with the aim of examining how the standard genetic code may have evolved from more primitive code(s) encoding a small number of amino acids. We determine the efficacy of the physico-chemical hypothesis of code origin in the absence and presence of horizontal gene transfer (HGT) by allowing a diverse collection of code-sequence sets to compete with each other. We find that in the absence of horizontal gene transfer, natural selection between competing codes distinguished by differences in the degree of physico-chemical optimization is unable to explain the structure of the standard genetic code. However, for certain probabilities of the horizontal transfer events, a universal code emerges having a structure that is consistent with the standard genetic code.


Subject(s)
Amino Acyl-tRNA Synthetases/chemistry , Codon/chemistry , Genetic Code , Origin of Life , RNA, Messenger/chemistry , RNA, Transfer, Amino Acid-Specific/chemistry , Amino Acids/chemistry , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Codon/metabolism , Evolution, Molecular , Gene Transfer, Horizontal , Genes , Models, Genetic , Probability , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Transfer, Amino Acid-Specific/metabolism , Selection, Genetic
16.
Orig Life Evol Biosph ; 44(4): 293-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25585807

ABSTRACT

It is proposed that the prebiotic ordering of nucleic acid and peptide sequences was a cooperative process in which nearly random populations of both kinds of polymers went through a codependent series of self-organisation events that simultaneously refined not only the accuracy of genetic replication and coding but also the functional specificity of protein catalysts, especially nascent aminoacyl-tRNA synthetase "urzymes".


Subject(s)
Amino Acids/chemistry , Amino Acyl-tRNA Synthetases/chemistry , Origin of Life , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Transfer, Amino Acid-Specific/chemistry , Amino Acid Sequence , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Biocatalysis , Energy Transfer , Genetic Code , Information Theory , Molecular Sequence Data , RNA, Catalytic/chemistry , RNA, Catalytic/metabolism , RNA, Messenger/metabolism , RNA, Transfer, Amino Acid-Specific/metabolism , Thermodynamics
17.
Orig Life Evol Biosph ; 44(4): 299-302, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25592392

ABSTRACT

RNA world hypothesis is widely accepted still now, as an idea by which the origin of life might be explained. But, there are many weak points in the hypothesis. In contrast, I have proposed a more reasonable [GADV]-protein world hypothesis or GADV hypothesis, suggesting that life originated from the protein world, which was formed by pseudo-replication of [GADV]-proteins. In this communication, I will discuss about the origin of life from the point of view of the GADV hypothesis.


Subject(s)
Genetic Code , Models, Genetic , Origin of Life , Protein Biosynthesis , RNA, Transfer, Amino Acid-Specific/chemistry , Alanine/chemistry , Alanine/metabolism , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Biological Evolution , Carbon/metabolism , Glycine/chemistry , Glycine/metabolism , Protein Conformation , Protein Folding , RNA, Transfer, Amino Acid-Specific/metabolism , Valine/chemistry , Valine/metabolism
18.
RNA Biol ; 11(12): 1540-54, 2014.
Article in English | MEDLINE | ID: mdl-25616362

ABSTRACT

Pseudouridine (Ψ) is the most abundant of >150 nucleoside modifications in RNA. Although Ψ was discovered as the first modified nucleoside more than half a century ago, neither the enzymatic mechanism of its formation, nor the function of this modification are fully elucidated. We present the consistent picture of Ψ synthases, their substrates and their substrate positions in model organisms of all domains of life as it has emerged to date and point out the challenges that remain concerning higher eukaryotes and the elucidation of the enzymatic mechanism.


Subject(s)
Intramolecular Transferases/metabolism , Pseudouridine/metabolism , RNA Processing, Post-Transcriptional , RNA, Guide, Kinetoplastida/metabolism , RNA, Transfer, Amino Acid-Specific/metabolism , Uridine/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Intramolecular Transferases/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Nucleic Acid Conformation , RNA/genetics , RNA/metabolism , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics , RNA, Mitochondrial , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Transfer, Amino Acid-Specific/chemistry , RNA, Transfer, Amino Acid-Specific/genetics , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Ribosomes/chemistry , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
19.
Chemistry ; 19(47): 15872-8, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24127424

ABSTRACT

The twenty first amino acid, selenocysteine (Sec), is the only amino acid that is synthesized on its cognate transfer RNA (tRNA(Sec)) in all domains of life. The multistep pathway involves O-phosphoseryl-tRNA:selenocysteinyl-tRNA synthase (SepSecS), an enzyme that catalyzes the terminal chemical reaction during which the phosphoseryl-tRNA(Sec) intermediate is converted into selenocysteinyl-tRNA(Sec). The SepSecS architecture and the mode of tRNA(Sec) recognition have been recently determined at atomic resolution. The crystal structure provided valuable insights that gave rise to mechanistic proposals that could not be validated because of the lack of appropriate molecular probes. To further improve our understanding of the mechanism of the biosynthesis of selenocysteine in general and the mechanism of SepSecS in particular, stable tRNA(Sec) substrates carrying aminoacyl moieties that mimic particular reaction intermediates are needed. Here, we report on the accurate synthesis of methylated, phosphorylated, and phosphonated serinyl-derived tRNA(Sec) mimics that contain a hydrolysis-resistant ribose 3'-amide linkage instead of the natural ester bond. The procedures introduced allow for efficient site-specific methylation and/or phosphorylation directly on the solid support utilized in the automated RNA synthesis. For the preparation of (S)-2-amino-4-phosphonobutyric acid-oligoribonucleotide conjugates, a separate solid support was generated. Furthermore, we developed a three-strand enzymatic ligation protocol to obtain the corresponding full-length tRNA(Sec) derivatives. Finally, we developed an electrophoretic mobility shift assay (EMSA) for rapid, qualitative characterization of the SepSecS-tRNA interactions. The novel tRNA(Sec) mimics are promising candidates for further elucidation of the biosynthesis of selenocysteine by X-ray crystallography and other biochemical approaches, and could be attractive for similar studies on other tRNA-dependent enzymes.


Subject(s)
Biocompatible Materials/chemical synthesis , Phosphorous Acids/chemistry , RNA, Transfer, Amino Acid-Specific/chemistry , Aminobutyrates/chemistry , Base Sequence , Biocompatible Materials/chemistry , Methylation , Nucleic Acid Conformation , Phosphorylation , RNA, Transfer, Amino Acid-Specific/chemical synthesis , Solid-Phase Synthesis Techniques
20.
Nucleic Acids Res ; 41(21): 9800-11, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23982514

ABSTRACT

Selenoprotein expression in Escherichia coli redefines specific single UGA codons from translational termination to selenocysteine (Sec) insertion. This process requires the presence of a Sec Insertion Sequence (SECIS) in the mRNA, which forms a secondary structure that binds a unique Sec-specific elongation factor that catalyzes Sec insertion at the predefined UGA instead of release factor 2-mediated termination. During overproduction of recombinant selenoproteins, this process nonetheless typically results in expression of UGA-truncated products together with the production of recombinant selenoproteins. Here, we found that premature termination can be fully avoided through a SECIS-dependent Sec-mediated suppression of UGG, thereby yielding either tryptophan or Sec insertion without detectable premature truncation. The yield of recombinant selenoprotein produced with this method approached that obtained with a classical UGA codon for Sec insertion. Sec-mediated suppression of UGG thus provides a novel method for selenoprotein production, as here demonstrated with rat thioredoxin reductase. The results also reveal that the E. coli selenoprotein synthesis machinery has the inherent capability to promote wobble decoding.


Subject(s)
Codon , Escherichia coli/genetics , Selenocysteine/metabolism , Selenoproteins/biosynthesis , Animals , Anticodon , RNA, Transfer, Amino Acid-Specific/chemistry , Rats , Recombinant Proteins/biosynthesis , Selenoproteins/genetics , Thioredoxin Reductase 1/biosynthesis , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL