Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 853
Filter
1.
Elife ; 122024 Jul 30.
Article in English | MEDLINE | ID: mdl-39076160

ABSTRACT

Current methods to quantify the fraction of aminoacylated tRNAs, also known as the tRNA charge, are limited by issues with either low throughput, precision, and/or accuracy. Here, we present an optimized charge transfer RNA sequencing (tRNA-Seq) method that combines previous developments with newly described approaches to establish a protocol for precise and accurate tRNA charge measurements. We verify that this protocol provides robust quantification of tRNA aminoacylation and we provide an end-to-end method that scales to hundreds of samples including software for data processing. Additionally, we show that this method supports measurements of relative tRNA expression levels and can be used to infer tRNA modifications through reverse transcription misincorporations, thereby supporting multipurpose applications in tRNA biology.


Subject(s)
RNA, Transfer , RNA, Transfer/genetics , RNA, Transfer/metabolism , Transfer RNA Aminoacylation , Sequence Analysis, RNA/methods , Aminoacylation/genetics
2.
Nucleic Acids Res ; 52(7): 3938-3949, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38477328

ABSTRACT

In the hypothetical RNA world, ribozymes could have acted as modern aminoacyl-tRNA synthetases (ARSs) to charge tRNAs, thus giving rise to the peptide synthesis along with the evolution of a primitive translation apparatus. We previously reported a T-boxzyme, Tx2.1, which selectively charges initiator tRNA with N-biotinyl-phenylalanine (BioPhe) in situ in a Flexible In-vitro Translation (FIT) system to produce BioPhe-initiating peptides. Here, we performed in vitro selection of elongation-capable T-boxzymes (elT-boxzymes), using para-azido-l-phenylalanine (PheAZ) as an acyl-donor. We implemented a new strategy to enrich elT-boxzyme-tRNA conjugates that self-aminoacylated on the 3'-terminus selectively. One of them, elT32, can charge PheAZ onto tRNA in trans in response to its cognate anticodon. Further evolution of elT32 resulted in elT49, with enhanced aminoacylation activity. We have demonstrated the translation of a PheAZ-containing peptide in an elT-boxzyme-integrated FIT system, revealing that elT-boxzymes are able to generate the PheAZ-tRNA in response to the cognate anticodon in situ of a custom-made translation system. This study, together with Tx2.1, illustrates a scenario where a series of ribozymes could have overseen aminoacylation and co-evolved with a primitive RNA-based translation system.


Subject(s)
Anticodon , Protein Biosynthesis , RNA, Catalytic , RNA, Transfer, Amino Acyl , RNA, Catalytic/metabolism , RNA, Catalytic/genetics , Anticodon/genetics , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Amino Acyl/genetics , Phenylalanine/metabolism , Phenylalanine/analogs & derivatives , Amino Acyl-tRNA Synthetases/metabolism , Amino Acyl-tRNA Synthetases/genetics , Transfer RNA Aminoacylation , Aminoacylation , Peptide Chain Elongation, Translational
3.
Chem Commun (Camb) ; 60(12): 1607-1610, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38230513

ABSTRACT

Extensive research has focused on genetic code reprogramming using flexizymes (Fxs), ribozymes enabling diverse tRNA acylation. Here we describe a nucleoside-modification strategy for the preparation of flexizyme variants derived from 2'-OMe, 2'-F, and 2'-MOE modifications with unique and versatile activities, enabling the charging of tRNAs with a broad range of substrates. This innovative strategy holds promise for synthetic biology applications, offering a robust pathway to expand the genetic code for diverse substrate incorporation.


Subject(s)
RNA, Catalytic , Transfer RNA Aminoacylation , Nucleosides/metabolism , RNA, Transfer/metabolism , Genetic Code , RNA, Catalytic/metabolism
4.
Nucleic Acids Res ; 52(2): 513-524, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38100361

ABSTRACT

Protein translation is orchestrated through tRNA aminoacylation and ribosomal elongation. Among the highly conserved structure of tRNAs, they have distinguishing features which promote interaction with their cognate aminoacyl tRNA synthetase (aaRS). These key features are referred to as identity elements. In our study, we investigated the tRNA:aaRS pair that installs the 22nd amino acid, pyrrolysine (tRNAPyl:PylRS). Pyrrolysyl-tRNA synthetases (PylRSs) are naturally encoded in some archaeal and bacterial genomes to acylate tRNAPyl with pyrrolysine. Their large amino acid binding pocket and poor recognition of the tRNA anticodon have been instrumental in incorporating >200 noncanonical amino acids. PylRS enzymes can be divided into three classes based on their genomic structure. Two classes contain both an N-terminal and C-terminal domain, however the third class (ΔpylSn) lacks the N-terminal domain. In this study we explored the tRNA identity elements for a ΔpylSn tRNAPyl from Candidatus Methanomethylophilus alvus which drives the orthogonality seen with its cognate PylRS (MaPylRS). From aminoacylation and translation assays we identified five key elements in ΔpylSn tRNAPyl necessary for MaPylRS activity. The absence of a base (position 8) and a G-U wobble pair (G28:U42) were found to affect the high-resolution structure of the tRNA, while molecular dynamic simulations led us to acknowledge the rigidity imparted from the G-C base pairs (G3:C70 and G5:C68).


Enzymes known as PylRS offer the remarkable ability to expand the natural genetic code of a living cell with unnatural amino acids. Currently, over 200 unnatural amino acids can be genetically encoded with the help of PylRS and its partner tRNAPyl, enabling us to endow proteins with novel properties, or regulate protein activity using light or inducible cross-linking. One intriguing feature of PylRS enzymes is their ability to avoid cross-reactivity when two PylRS homologs from different organisms-such as those from the archaea Methanosarcina mazei and Methanomethylophilus alvus-are co-expressed in a single cell. This makes it possible to simultaneously encode two unnatural amino acids in a single protein. This study illuminates the elusive mechanism of PylRS specificity by using cryo-electron microscopy, biochemistry and molecular simulations. The interaction of PylRS from M. alvus with its tRNAPyl is best described as two pieces of a jigsaw puzzle; in which PylRS recognizes the unique shape of its cognate tRNA instead of specific nucleotides in the tRNA sequence like other tRNA-binding enzymes. This finding may streamline the rational design of tools for simultaneous genetic incorporation of multiple unnatural amino acids, thereby facilitating the development of valuable proteins for research, medicine, and biotechnology.


Subject(s)
Amino Acyl-tRNA Synthetases , Archaea , Gastrointestinal Microbiome , Humans , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/isolation & purification , Amino Acyl-tRNA Synthetases/metabolism , Archaea/enzymology , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Transfer RNA Aminoacylation
5.
Trends Genet ; 40(3): 276-290, 2024 03.
Article in English | MEDLINE | ID: mdl-38123442

ABSTRACT

In the past decade tRNA sequencing (tRNA-seq) has attracted considerable attention as an important tool for the development of novel approaches to quantify highly modified tRNA species and to propel tRNA research aimed at understanding the cellular physiology and disease and development of tRNA-based therapeutics. Many methods are available to quantify tRNA abundance while accounting for modifications and tRNA charging/acylation. Advances in both library preparation methods and bioinformatic workflows have enabled developments in next-generation sequencing (NGS) workflows. Other approaches forgo NGS applications in favor of hybridization-based approaches. In this review we provide a brief comparative overview of various tRNA quantification approaches, focusing on the advantages and disadvantages of these methods, which together facilitate reliable tRNA quantification.


Subject(s)
High-Throughput Nucleotide Sequencing , RNA, Transfer , RNA, Transfer/genetics , High-Throughput Nucleotide Sequencing/methods , Computational Biology , Transfer RNA Aminoacylation
6.
Nat Commun ; 14(1): 5764, 2023 09 16.
Article in English | MEDLINE | ID: mdl-37717009

ABSTRACT

The expanded hexanucleotide GGGGCC repeat mutation in the C9orf72 gene is the main genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Under one disease mechanism, sense and antisense transcripts of the repeat are predicted to bind various RNA-binding proteins, compromise their function and cause cytotoxicity. Here we identify phenylalanine-tRNA synthetase (FARS) subunit alpha (FARSA) as the main interactor of the CCCCGG antisense repeat RNA in cytosol. The aminoacylation of tRNAPhe by FARS is inhibited by antisense RNA, leading to decreased levels of charged tRNAPhe. Remarkably, this is associated with global reduction of phenylalanine incorporation in the proteome and decrease in expression of phenylalanine-rich proteins in cellular models and patient tissues. In conclusion, this study reveals functional inhibition of FARSA in the presence of antisense RNA repeats. Compromised aminoacylation of tRNA could lead to impairments in protein synthesis and further contribute to C9orf72 mutation-associated pathology.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Humans , Transfer RNA Aminoacylation , Aminoacylation , Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , C9orf72 Protein/genetics , Phenylalanine/genetics , RNA, Transfer, Phe , RNA, Antisense
7.
Nucleic Acids Res ; 51(15): 8070-8084, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37470821

ABSTRACT

Leucyl-tRNA synthetase (LeuRS) is a Class I aminoacyl-tRNA synthetase (aaRS) that synthesizes leucyl-tRNAleu for codon-directed protein synthesis. Two signature sequences, HxGH and KMSKS help stabilize transition-states for amino acid activation and tRNA aminoacylation by all Class I aaRS. Separate alanine mutants of each signature, together with the double mutant, behave in opposite ways in Pyrococcus horikoshii LeuRS and the 129-residue urzyme ancestral model generated from it (LeuAC). Free energy coupling terms, Δ(ΔG‡), for both reactions are large and favourable for LeuRS, but unfavourable for LeuAC. Single turnover assays with 32Pα-ATP show correspondingly different internal products. These results implicate domain motion in catalysis by full-length LeuRS. The distributed thermodynamic cycle of mutational changes authenticates LeuAC urzyme catalysis far more convincingly than do single point mutations. Most importantly, the evolutionary gain of function induced by acquiring the anticodon-binding (ABD) and multiple insertion modules in the catalytic domain appears to be to coordinate the catalytic function of the HxGH and KMSKS signature sequences. The implication that backbone elements of secondary structures achieve a major portion of the overall transition-state stabilization by LeuAC is also consistent with coevolution of the genetic code and metabolic pathways necessary to produce histidine and lysine sidechains.


Subject(s)
Amino Acyl-tRNA Synthetases , Leucine-tRNA Ligase , Amino Acyl-tRNA Synthetases/metabolism , Anticodon , Transfer RNA Aminoacylation , Genetic Code , Leucine-tRNA Ligase/metabolism , Catalysis
8.
Nucleic Acids Res ; 51(12): 5911-5930, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37224536

ABSTRACT

In Escherichia coli, inconsistencies between in vitro tRNA aminoacylation measurements and in vivo protein synthesis demands were postulated almost 40 years ago, but have proven difficult to confirm. Whole-cell modeling can test whether a cell behaves in a physiologically correct manner when parameterized with in vitro measurements by providing a holistic representation of cellular processes in vivo. Here, a mechanistic model of tRNA aminoacylation, codon-based polypeptide elongation, and N-terminal methionine cleavage was incorporated into a developing whole-cell model of E. coli. Subsequent analysis confirmed the insufficiency of aminoacyl-tRNA synthetase kinetic measurements for cellular proteome maintenance, and estimated aminoacyl-tRNA synthetase kcats that were on average 7.6-fold higher. Simulating cell growth with perturbed kcats demonstrated the global impact of these in vitro measurements on cellular phenotypes. For example, an insufficient kcat for HisRS caused protein synthesis to be less robust to the natural variability in aminoacyl-tRNA synthetase expression in single cells. More surprisingly, insufficient ArgRS activity led to catastrophic impacts on arginine biosynthesis due to underexpressed N-acetylglutamate synthase, where translation depends on repeated CGG codons. Overall, the expanded E. coli model deepens understanding of how translation operates in an in vivo context.


Subject(s)
Amino Acyl-tRNA Synthetases , Arginine , Escherichia coli , Amino Acyl-tRNA Synthetases/metabolism , Aminoacylation , Arginine/biosynthesis , Escherichia coli/metabolism , Feedback , Transfer RNA Aminoacylation
9.
Wiley Interdiscip Rev RNA ; 14(5): e1789, 2023.
Article in English | MEDLINE | ID: mdl-37042417

ABSTRACT

Aminoacyl-tRNA synthetases form the protein family that controls the interpretation of the genetic code, with tRNA aminoacylation being the key chemical step during which an amino acid is assigned to a corresponding sequence of nucleic acids. In consequence, aminoacyl-tRNA synthetases have been studied in their physiological context, in disease states, and as tools for synthetic biology to enable the expansion of the genetic code. Here, we review the fundamentals of aminoacyl-tRNA synthetase biology and classification, with a focus on mammalian cytoplasmic enzymes. We compile evidence that the localization of aminoacyl-tRNA synthetases can be critical in health and disease. In addition, we discuss evidence from synthetic biology which made use of the importance of subcellular localization for efficient manipulation of the protein synthesis machinery. This article is categorized under: RNA Processing Translation > Translation Regulation RNA Processing > tRNA Processing RNA Export and Localization > RNA Localization.


Subject(s)
Amino Acyl-tRNA Synthetases , Transfer RNA Aminoacylation , Animals , RNA, Transfer/genetics , Genetic Code , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Mammals/genetics , Mammals/metabolism
10.
Methods Mol Biol ; 2620: 93-99, 2023.
Article in English | MEDLINE | ID: mdl-37010753

ABSTRACT

This chapter describes the preparation of tRNAArg by in vitro transcription. tRNA produced by this method can be efficiently utilized for in vitro arginylation assays, following aminoacylation with Arg-tRNA synthetase, either directly during the arginylation reaction or separately to produce the purified preparation of Arg-tRNAArg. tRNA charging is described in other chapters of this book.


Subject(s)
Arginine-tRNA Ligase , RNA, Transfer, Arg , RNA, Transfer, Arg/genetics , RNA, Transfer, Arg/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Arginine-tRNA Ligase/genetics , Arginine-tRNA Ligase/metabolism , Transfer RNA Aminoacylation
11.
Methods Mol Biol ; 2620: 107-111, 2023.
Article in English | MEDLINE | ID: mdl-37010755

ABSTRACT

This chapter describes the preparation of pre-charged Arg-tRNA that can be used in arginylation reaction. While in a typical arginylation reaction arginyl-tRNA synthetase (RARS) is normally included as a component of the reaction and continually charges tRNA during arginylation, it is sometimes necessary to separate the charging and the arginylation step, in order to perform each reaction under controlled conditions, e.g., for measuring the kinetics or determining the effect of different compounds and chemicals on the reaction. In such cases, tRNAArg can be pre-charged with Arg and purified away from the RARS enzyme prior to arginylation.


Subject(s)
Amino Acyl-tRNA Synthetases , Arginine-tRNA Ligase , Arginine-tRNA Ligase/chemistry , Arginine-tRNA Ligase/genetics , Arginine-tRNA Ligase/metabolism , RNA, Transfer, Arg/chemistry , RNA, Transfer, Arg/genetics , RNA, Transfer, Arg/metabolism , Aminoacylation , RNA, Transfer/genetics , Transfer RNA Aminoacylation , Kinetics , Amino Acyl-tRNA Synthetases/metabolism
12.
Nucleic Acids Res ; 51(8): 3988-3999, 2023 05 08.
Article in English | MEDLINE | ID: mdl-36951109

ABSTRACT

High fidelity tRNA aminoacylation by aminoacyl-tRNA synthetases is essential for cell viability. ProXp-ala is a trans-editing protein that is present in all three domains of life and is responsible for hydrolyzing mischarged Ala-tRNAPro and preventing mistranslation of proline codons. Previous studies have shown that, like bacterial prolyl-tRNA synthetase, Caulobacter crescentus ProXp-ala recognizes the unique C1:G72 terminal base pair of the tRNAPro acceptor stem, helping to ensure deacylation of Ala-tRNAPro but not Ala-tRNAAla. The structural basis for C1:G72 recognition by ProXp-ala is still unknown and was investigated here. NMR spectroscopy, binding, and activity assays revealed two conserved residues, K50 and R80, that likely interact with the first base pair, stabilizing the initial protein-RNA encounter complex. Modeling studies are consistent with direct interaction between R80 and the major groove of G72. A third key contact between A76 of tRNAPro and K45 of ProXp-ala was essential for binding and accommodating the CCA-3' end in the active site. We also demonstrated the essential role that the 2'OH of A76 plays in catalysis. Eukaryotic ProXp-ala proteins recognize the same acceptor stem positions as their bacterial counterparts, albeit with different nucleotide base identities. ProXp-ala is encoded in some human pathogens; thus, these results have the potential to inform new antibiotic drug design.


Subject(s)
Amino Acyl-tRNA Synthetases , RNA, Transfer, Pro , Humans , RNA, Transfer, Pro/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Proline/chemistry , Transfer RNA Aminoacylation , Codon , Catalytic Domain
13.
Nucleic Acids Res ; 51(4): 1528-1570, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36744444

ABSTRACT

tRNAs are key partners in ribosome-dependent protein synthesis. This process is highly dependent on the fidelity of tRNA aminoacylation by aminoacyl-tRNA synthetases and relies primarily on sets of identities within tRNA molecules composed of determinants and antideterminants preventing mischarging by non-cognate synthetases. Such identity sets were discovered in the tRNAs of a few model organisms, and their properties were generalized as universal identity rules. Since then, the panel of identity elements governing the accuracy of tRNA aminoacylation has expanded considerably, but the increasing number of reported functional idiosyncrasies has led to some confusion. In parallel, the description of other processes involving tRNAs, often well beyond aminoacylation, has progressed considerably, greatly expanding their interactome and uncovering multiple novel identities on the same tRNA molecule. This review highlights key findings on the mechanistics and evolution of tRNA and tRNA-like identities. In addition, new methods and their results for searching sets of multiple identities on a single tRNA are discussed. Taken together, this knowledge shows that a comprehensive understanding of the functional role of individual and collective nucleotide identity sets in tRNA molecules is needed for medical, biotechnological and other applications.


Subject(s)
Amino Acyl-tRNA Synthetases , Transfer RNA Aminoacylation , Aminoacylation , Biotechnology , RNA, Transfer
14.
Nucleic Acids Res ; 50(22): 12951-12968, 2022 12 09.
Article in English | MEDLINE | ID: mdl-36503967

ABSTRACT

Mitochondrial RNA metabolism is suggested to occur in identified compartmentalized foci, i.e. mitochondrial RNA granules (MRGs). Mitochondrial aminoacyl-tRNA synthetases (mito aaRSs) catalyze tRNA charging and are key components in mitochondrial gene expression. Mutations of mito aaRSs are associated with various human disorders. However, the suborganelle distribution, interaction network and regulatory mechanism of mito aaRSs remain largely unknown. Here, we found that all mito aaRSs partly colocalize with MRG, and this colocalization is likely facilitated by tRNA-binding capacity. A fraction of human mitochondrial AlaRS (hmtAlaRS) and hmtSerRS formed a direct complex via interaction between catalytic domains in vivo. Aminoacylation activities of both hmtAlaRS and hmtSerRS were fine-tuned upon complex formation in vitro. We further established a full spectrum of interaction networks via immunoprecipitation and mass spectrometry for all mito aaRSs and discovered interactions between hmtSerRS and hmtAsnRS, between hmtSerRS and hmtTyrRS and between hmtThrRS and hmtArgRS. The activity of hmtTyrRS was also influenced by the presence of hmtSerRS. Notably, hmtSerRS utilized the same catalytic domain in mediating several interactions. Altogether, our results systematically analyzed the suborganelle localization and interaction network of mito aaRSs and discovered several mito aaRS-containing complexes, deepening our understanding of the functional and regulatory mechanisms of mito aaRSs.


Subject(s)
Amino Acyl-tRNA Synthetases , Transfer RNA Aminoacylation , Humans , Amino Acyl-tRNA Synthetases/metabolism , Cytoplasmic Ribonucleoprotein Granules/metabolism , RNA, Mitochondrial/metabolism , RNA, Transfer/metabolism
15.
Genes (Basel) ; 13(12)2022 12 09.
Article in English | MEDLINE | ID: mdl-36553587

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are highly conserved essential enzymes that charge tRNA with cognate amino acids-the first step of protein synthesis. Of the 37 nuclear-encoded human ARS genes, 17 encode enzymes are exclusively targeted to the mitochondria (mt-ARSs). Mutations in nuclear mt-ARS genes are associated with rare, recessive human diseases with a broad range of clinical phenotypes. While the hypothesized disease mechanism is a loss-of-function effect, there is significant clinical heterogeneity among patients that have mutations in different mt-ARS genes and also among patients that have mutations in the same mt-ARS gene. This observation suggests that additional factors are involved in disease etiology. In this review, we present our current understanding of diseases caused by mutations in the genes encoding mt-ARSs and propose explanations for the observed clinical heterogeneity.


Subject(s)
Amino Acyl-tRNA Synthetases , Transfer RNA Aminoacylation , Humans , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Phenotype , RNA, Transfer/genetics , RNA, Transfer/metabolism
16.
Cell Chem Biol ; 29(7): 1071-1112, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35413283

ABSTRACT

A critical step in repurposing the cellular translation machinery for the synthesis of polymeric products is the acylation of transfer RNA (tRNA) with unnatural monomers. Toward this goal, flexizymes, ribozymes capable of aminoacylation, have emerged as a uniquely adept tool for charging tRNA with ever increasingly diverse substrates. In this review, we present a library of monomer substrates that have been tested for tRNA acylation with the flexizyme system. From this mile-high view, we provide insights for understanding the chemical factors that influence flexizyme-mediated tRNA acylation. We conclude that flexizymes are primitive esterification catalysts that display a modest binding affinity to the monomer's aromatic recognition element. Together, these robust, yet flexible, flexizyme systems provide researchers with unprecedented access for preparing unnatural acyl-tRNA and the opportunity to repurpose the translation machinery for the synthesis of novel biologically derived structures beyond native proteins and peptides.


Subject(s)
RNA, Catalytic , Transfer RNA Aminoacylation , Acylation , Catalysis , Peptides/metabolism , RNA, Catalytic/chemistry , RNA, Transfer/metabolism
18.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Article in English | MEDLINE | ID: mdl-35140183

ABSTRACT

Aminoacylated transfer RNAs, which harbor a covalent linkage between amino acids and RNA, are a universally conserved feature of life. Because they are essential substrates for ribosomal translation, aminoacylated oligonucleotides must have been present in the RNA world prior to the evolution of the ribosome. One possibility we are exploring is that the aminoacyl ester linkage served another function before being recruited for ribosomal protein synthesis. The nonenzymatic assembly of ribozymes from short RNA oligomers under realistic conditions remains a key challenge in demonstrating a plausible pathway from prebiotic chemistry to the RNA world. Here, we show that aminoacylated RNAs can undergo template-directed assembly into chimeric amino acid-RNA polymers that are active ribozymes. We demonstrate that such chimeric polymers can retain the enzymatic function of their all-RNA counterparts by generating chimeric hammerhead, RNA ligase, and aminoacyl transferase ribozymes. Amino acids with diverse side chains form linkages that are well tolerated within the RNA backbone and, in the case of an aminoacyl transferase, even in its catalytic center, potentially bringing novel functionalities to ribozyme catalysis. Our work suggests that aminoacylation chemistry may have played a role in primordial ribozyme assembly. Increasing the efficiency of this process provides an evolutionary rationale for the emergence of sequence and amino acid-specific aminoacyl-RNA synthetase ribozymes, which could then have generated the substrates for ribosomal protein synthesis.


Subject(s)
RNA, Catalytic/metabolism , Transfer RNA Aminoacylation/physiology , Base Sequence , DNA , Nucleic Acid Conformation , RNA, Transfer/genetics , RNA, Transfer/metabolism
19.
Nucleic Acids Res ; 49(22): 13108-13121, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34878141

ABSTRACT

Mutations in genes encoding mitochondrial aminoacyl-tRNA synthetases are linked to diverse diseases. However, the precise mechanisms by which these mutations affect mitochondrial function and disease development are not fully understood. Here, we develop a Drosophila model to study the function of dFARS2, the Drosophila homologue of the mitochondrial phenylalanyl-tRNA synthetase, and further characterize human disease-associated FARS2 variants. Inactivation of dFARS2 in Drosophila leads to developmental delay and seizure. Biochemical studies reveal that dFARS2 is required for mitochondrial tRNA aminoacylation, mitochondrial protein stability, and assembly and enzyme activities of OXPHOS complexes. Interestingly, by modeling FARS2 mutations associated with human disease in Drosophila, we provide evidence that expression of two human FARS2 variants, p.G309S and p.D142Y, induces seizure behaviors and locomotion defects, respectively. Together, our results not only show the relationship between dysfunction of mitochondrial aminoacylation system and pathologies, but also illustrate the application of Drosophila model for functional analysis of human disease-causing variants.


Subject(s)
Developmental Disabilities/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Mitochondrial Proteins/genetics , Mutation , Phenylalanine-tRNA Ligase/genetics , RNA, Transfer/genetics , Seizures/genetics , Animals , Cell Line , Developmental Disabilities/enzymology , Disease Models, Animal , Drosophila Proteins/deficiency , Drosophila melanogaster/enzymology , Gene Knockdown Techniques , Humans , Microscopy, Electron, Transmission , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/deficiency , Oxidative Phosphorylation , Phenylalanine-tRNA Ligase/deficiency , RNA, Transfer/metabolism , Seizures/enzymology , Transfer RNA Aminoacylation
20.
Science ; 374(6570): 955-960, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34793227

ABSTRACT

Viruses require multifunctional structured RNAs to hijack their host's biochemistry, but their mechanisms can be obscured by the difficulty of solving conformationally dynamic RNA structures. Using cryo­electron microscopy (cryo-EM), we visualized the structure of the mysterious viral transfer RNA (tRNA)­like structure (TLS) from the brome mosaic virus, which affects replication, translation, and genome encapsidation. Structures in isolation and those bound to tyrosyl-tRNA synthetase (TyrRS) show that this ~55-kilodalton purported tRNA mimic undergoes large conformational rearrangements to bind TyrRS in a form that differs substantially from that of tRNA. Our study reveals how viral RNAs can use a combination of static and dynamic RNA structures to bind host machinery through highly noncanonical interactions, and we highlight the utility of cryo-EM for visualizing small, conformationally dynamic structured RNAs.


Subject(s)
Bromovirus/genetics , RNA, Transfer/chemistry , RNA, Viral/chemistry , Tyrosine-tRNA Ligase/metabolism , Bromovirus/physiology , Cryoelectron Microscopy , Genome, Viral , Models, Molecular , Molecular Mimicry , Nucleic Acid Conformation , Phaseolus/enzymology , Phaseolus/virology , Protein Binding , Protein Conformation , RNA, Transfer/metabolism , RNA, Viral/metabolism , Transfer RNA Aminoacylation , Tyrosine-tRNA Ligase/chemistry , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL