Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nat Rev Mol Cell Biol ; 25(5): 359-378, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38182846

RESUMO

A growing class of small RNAs, known as tRNA-derived RNAs (tdRs), tRNA-derived small RNAs or tRNA-derived fragments, have long been considered mere intermediates of tRNA degradation. These small RNAs have recently been implicated in an evolutionarily conserved repertoire of biological processes. In this Review, we discuss the biogenesis and molecular functions of tdRs in mammals, including tdR-mediated gene regulation in cell metabolism, immune responses, transgenerational inheritance, development and cancer. We also discuss the accumulation of tRNA-derived stress-induced RNAs as a distinct adaptive cellular response to pathophysiological conditions. Furthermore, we highlight new conceptual advances linking RNA modifications with tdR activities and discuss challenges in studying tdR biology in health and disease.


Assuntos
RNA de Transferência , Animais , RNA de Transferência/metabolismo , RNA de Transferência/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Regulação da Expressão Gênica , Pequeno RNA não Traduzido/genética , Pequeno RNA não Traduzido/metabolismo
2.
Nat Immunol ; 23(10): 1433-1444, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36138184

RESUMO

Naive T cells undergo radical changes during the transition from dormant to hyperactive states upon activation, which necessitates de novo protein production via transcription and translation. However, the mechanism whereby T cells globally promote translation remains largely unknown. Here, we show that on exit from quiescence, T cells upregulate transfer RNA (tRNA) m1A58 'writer' proteins TRMT61A and TRMT6, which confer m1A58 RNA modification on a specific subset of early expressed tRNAs. These m1A-modified early tRNAs enhance translation efficiency, enabling rapid and necessary synthesis of MYC and of a specific group of key functional proteins. The MYC protein then guides the exit of naive T cells from a quiescent state into a proliferative state and promotes rapid T cell expansion after activation. Conditional deletion of the Trmt61a gene in mouse CD4+ T cells causes MYC protein deficiency and cell cycle arrest, disrupts T cell expansion upon cognate antigen stimulation and alleviates colitis in a mouse adoptive transfer colitis model. Our study elucidates for the first time, to our knowledge, the in vivo physiological roles of tRNA-m1A58 modification in T cell-mediated pathogenesis and reveals a new mechanism of tRNA-m1A58-controlled T cell homeostasis and signal-dependent translational control of specific key proteins.


Assuntos
Colite , RNA de Transferência , Transferência Adotiva , Animais , Proliferação de Células/genética , Colite/genética , Camundongos , Biossíntese de Proteínas , RNA de Transferência/genética , RNA de Transferência/metabolismo , Linfócitos T/metabolismo
3.
EMBO J ; 41(6): e108544, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-34850409

RESUMO

Since numerous RNAs and RBPs prevalently localize to active chromatin regions, many RNA-binding proteins (RBPs) may be potential transcriptional regulators. RBPs are generally thought to regulate transcription via noncoding RNAs. Here, we describe a distinct, dual mechanism of transcriptional regulation by the previously uncharacterized tRNA-modifying enzyme, hTrmt13. On one hand, hTrmt13 acts in the cytoplasm to catalyze 2'-O-methylation of tRNAs, thus regulating translation in a manner depending on its tRNA-modification activity. On the other hand, nucleus-localized hTrmt13 directly binds DNA as a transcriptional co-activator of key epithelial-mesenchymal transition factors, thereby promoting cell migration independent of tRNA-modification activity. These dual functions of hTrmt13 are mutually exclusive, as it can bind either DNA or tRNA through its CHHC zinc finger domain. Finally, we find that hTrmt13 expression is tightly associated with poor prognosis and survival in diverse cancer patients. Our discovery of the noncatalytic roles of an RNA-modifying enzyme provides a new perspective for understanding epitranscriptomic regulation.


Assuntos
RNA de Transferência , tRNA Metiltransferases , Humanos , Metilação , RNA/metabolismo , Processamento Pós-Transcricional do RNA , RNA de Transferência/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , tRNA Metiltransferases/genética , tRNA Metiltransferases/metabolismo
4.
Nucleic Acids Res ; 52(6): 3291-3309, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38165050

RESUMO

The mechanisms by which the relatively conserved spliceosome manages the enormously large number of splicing events that occur in humans (∼200 000 versus ∼300 in yeast) are poorly understood. Here, we show deposition of one RNA modification-N2-methylguanosine (m2G) on the G72 of U6 snRNA (the catalytic center of the spliceosome) promotes efficient pre-mRNA splicing activity in human cells. This modification was identified to be conserved among vertebrates. Further, THUMPD2 was demonstrated as the methyltransferase responsible for U6 m2G72 by explicitly recognizing the U6-specific sequences and structural elements. The knock-out of THUMPD2 eliminated U6 m2G72 and impaired the pre-mRNA splicing activity, resulting in thousands of changed alternative splicing events of endogenous pre-mRNAs in human cells. Notably, the aberrantly spliced pre-mRNA population elicited the nonsense-mediated mRNA decay pathway. We further show that THUMPD2 was associated with age-related macular degeneration and retinal function. Our study thus demonstrates how an RNA epigenetic modification of the major spliceosome regulates global pre-mRNA splicing and impacts physiology and disease.


Assuntos
Precursores de RNA , Splicing de RNA , Proteínas de Ligação a RNA , Degeneração Retiniana , Animais , Humanos , Metilação , Conformação de Ácido Nucleico , Degeneração Retiniana/metabolismo , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA/genética , RNA Nuclear Pequeno/metabolismo , Saccharomyces cerevisiae/genética , Spliceossomos/genética , Spliceossomos/metabolismo
5.
Genet Med ; 25(9): 100900, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37226891

RESUMO

PURPOSE: 5-methylcytosine RNA modifications are driven by NSUN methyltransferases. Although variants in NSUN2 and NSUN3 were associated with neurodevelopmental diseases, the physiological role of NSUN6 modifications on transfer RNAs and messenger RNAs remained elusive. METHODS: We combined exome sequencing of consanguineous families with functional characterization to identify a new neurodevelopmental disorder gene. RESULTS: We identified 3 unrelated consanguineous families with deleterious homozygous variants in NSUN6. Two of these variants are predicted to be loss-of-function. One maps to the first exon and is predicted to lead to the absence of NSUN6 via nonsense-mediated decay, whereas we showed that the other maps to the last exon and encodes a protein that does not fold correctly. Likewise, we demonstrated that the missense variant identified in the third family has lost its enzymatic activity and is unable to bind the methyl donor S-adenosyl-L-methionine. The affected individuals present with developmental delay, intellectual disability, motor delay, and behavioral anomalies. Homozygous ablation of the NSUN6 ortholog in Drosophila led to locomotion and learning impairment. CONCLUSION: Our data provide evidence that biallelic pathogenic variants in NSUN6 cause one form of autosomal recessive intellectual disability, establishing another link between RNA modification and cognition.


Assuntos
Deficiência Intelectual , Transtornos do Neurodesenvolvimento , Humanos , Deficiência Intelectual/genética , Homozigoto , Transtornos do Neurodesenvolvimento/genética , Metiltransferases/genética , Metiltransferases/metabolismo , RNA , Linhagem , tRNA Metiltransferases/genética , tRNA Metiltransferases/metabolismo
6.
Nucleic Acids Res ; 49(22): 13045-13061, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34871455

RESUMO

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


Assuntos
5-Metilcitosina/metabolismo , Anticódon/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , RNA de Transferência/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Anticódon/genética , Sequência de Bases , DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/genética , Células HEK293 , Células HeLa , Humanos , Inosina/metabolismo , Camundongos , Modelos Moleculares , Células NIH 3T3 , Conformação de Ácido Nucleico , Ligação Proteica , RNA de Transferência/química , RNA de Transferência/genética , RNA de Transferência de Ácido Aspártico/química , RNA de Transferência de Ácido Aspártico/genética , RNA de Transferência de Ácido Aspártico/metabolismo , RNA de Transferência de Glicina/química , RNA de Transferência de Glicina/genética , RNA de Transferência de Glicina/metabolismo , RNA de Transferência de Valina/química , RNA de Transferência de Valina/genética , RNA de Transferência de Valina/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Especificidade por Substrato
7.
Nucleic Acids Res ; 49(20): 11900-11919, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34669960

RESUMO

Post-transcriptional modifications affect tRNA biology and are closely associated with human diseases. However, progress on the functional analysis of tRNA modifications in metazoans has been slow because of the difficulty in identifying modifying enzymes. For example, the biogenesis and function of the prevalent N2-methylguanosine (m2G) at the sixth position of tRNAs in eukaryotes has long remained enigmatic. Herein, using a reverse genetics approach coupled with RNA-mass spectrometry, we identified that THUMP domain-containing protein 3 (THUMPD3) is responsible for tRNA: m2G6 formation in human cells. However, THUMPD3 alone could not modify tRNAs. Instead, multifunctional methyltransferase subunit TRM112-like protein (TRMT112) interacts with THUMPD3 to activate its methyltransferase activity. In the in vitro enzymatic assay system, THUMPD3-TRMT112 could methylate all the 26 tested G6-containing human cytoplasmic tRNAs by recognizing the characteristic 3'-CCA of mature tRNAs. We also showed that m2G7 of tRNATrp was introduced by THUMPD3-TRMT112. Furthermore, THUMPD3 is widely expressed in mouse tissues, with an extremely high level in the testis. THUMPD3-knockout cells exhibited impaired global protein synthesis and reduced growth. Our data highlight the significance of the tRNA: m2G6/7 modification and pave a way for further studies of the role of m2G in sperm tRNA derived fragments.


Assuntos
Metiltransferases/metabolismo , RNA de Transferência/metabolismo , Proteínas de Ligação a RNA/metabolismo , tRNA Metiltransferases/metabolismo , Células HEK293 , Células HeLa , Humanos , Metilação , Metiltransferases/genética , Processamento Pós-Transcricional do RNA , Proteínas de Ligação a RNA/genética , Especificidade por Substrato , tRNA Metiltransferases/genética
8.
EMBO Rep ; 21(8): e50095, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32558197

RESUMO

tRNA modifications at the anti-codon loop are critical for accurate decoding. FTSJ1 was hypothesized to be a human tRNA 2'-O-methyltransferase. tRNAPhe (GAA) from intellectual disability patients with mutations in ftsj1 lacks 2'-O-methylation at C32 and G34 (Cm32 and Gm34). However, the catalytic activity, RNA substrates, and pathogenic mechanism of FTSJ1 remain unknown, owing, in part, to the difficulty in reconstituting enzymatic activity in vitro. Here, we identify an interacting protein of FTSJ1, WDR6. For the first time, we reconstitute the 2'-O-methylation activity of the FTSJ1-WDR6 complex in vitro, which occurs at position 34 of specific tRNAs with m1 G37 as a prerequisite. We find that modifications at positions 32, 34, and 37 are interdependent and occur in a hierarchical order in vivo. We also show that the translation efficiency of the UUU codon, but not the UUC codon decoded by tRNAPhe (GAA), is reduced in ftsj1 knockout cells. Bioinformatics analysis reveals that almost 40% of the high TTT-biased genes are related to brain/nervous functions. Our data potentially enhance our understanding of the relationship between FTSJ1 and nervous system development.


Assuntos
Deficiência Intelectual , Códon , Humanos , Deficiência Intelectual/genética , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Proteínas Nucleares/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , tRNA Metiltransferases/genética , tRNA Metiltransferases/metabolismo
9.
Nucleic Acids Res ; 48(9): 4946-4959, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32232361

RESUMO

Human cytosolic leucyl-tRNA synthetase (hcLRS) is an essential and multifunctional enzyme. Its canonical function is to catalyze the covalent ligation of leucine to tRNALeu, and it may also hydrolyze mischarged tRNAs through an editing mechanism. Together with eight other aminoacyl-tRNA synthetases (AaRSs) and three auxiliary proteins, it forms a large multi-synthetase complex (MSC). Beyond its role in translation, hcLRS has an important moonlight function as a leucine sensor in the rapamycin complex 1 (mTORC1) pathway. Since this pathway is active in cancer development, hcLRS is a potential target for anti-tumor drug development. Moreover, LRS from pathogenic microbes are proven drug targets for developing antibiotics, which however should not inhibit hcLRS. Here we present the crystal structure of hcLRS at a 2.5 Å resolution, the first complete structure of a eukaryotic LRS, and analyze the binding of various compounds that target different sites of hcLRS. We also deduce the assembly mechanism of hcLRS into the MSC through reconstitution of the entire mega complex in vitro. Overall, our study provides the molecular basis for understanding both the multifaceted functions of hcLRS and for drug development targeting these functions.


Assuntos
Leucina-tRNA Ligase/química , Anti-Infecciosos/química , Biocatálise , Domínio Catalítico , Desenho de Fármacos , Humanos , Leucina-tRNA Ligase/efeitos dos fármacos , Leucina-tRNA Ligase/metabolismo , Modelos Moleculares , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Domínios Proteicos , RNA de Transferência de Leucina/metabolismo , Aminoacilação de RNA de Transferência
10.
Nucleic Acids Res ; 47(4): 2041-2055, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30541086

RESUMO

Human NOL1/NOP2/Sun RNA methyltransferase family member 6 (hNSun6) generates 5-methylcytosine (m5C) at C72 of four specific tRNAs, and its homologs are present only in higher eukaryotes and hyperthermophilic archaea. Archaeal NSun6 homologs possess conserved catalytic residues, but have distinct differences in their RNA recognition motifs from eukaryotic NSun6s. Until now, the biochemical properties and functions of archaeal NSun6 homologs were unknown. In archaeon Pyrococcus horikoshii OT3, the gene encoding the NSun6 homolog is PH1991. We demonstrated that the PH1991 protein could catalyze m5C72 formation on some specific PhtRNAs in vitro and was thus named as PhNSun6. Remarkably, PhNSun6 has a much wider range of tRNA substrates than hNSun6, which was attributed to its tRNA substrate specificity. The mechanism was further elucidated using biochemical and crystallographic experiments. Structurally, the binding pocket for nucleotide 73 in PhNSun6 is specific to accommodate U73 or G73-containing PhtRNAs. Furthermore, PhNSun6 lacks the eukaryotic NSun6-specific Lys-rich loop, resulting in the non-recognition of D-stem region by PhNSun6. Functionally, the m5C72 modification could slightly promote the thermal stability of PhtRNAs, but did not affect the amino acid accepting activity of PhtRNAs.


Assuntos
Proteínas Arqueais/química , Pyrococcus horikoshii/enzimologia , RNA de Transferência/genética , tRNA Metiltransferases/química , 5-Metilcitosina/química , Sequência de Aminoácidos/genética , Proteínas Arqueais/genética , Catálise , Cristalografia por Raios X , Humanos , RNA de Transferência/química , Especificidade por Substrato , tRNA Metiltransferases/genética
11.
Nucleic Acids Res ; 45(11): 6684-6697, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28531330

RESUMO

5-methylcytosine (m5C) modifications of RNA are ubiquitous in nature and play important roles in many biological processes such as protein translational regulation, RNA processing and stress response. Aberrant expressions of RNA:m5C methyltransferases are closely associated with various human diseases including cancers. However, no structural information for RNA-bound RNA:m5C methyltransferase was available until now, hindering elucidation of the catalytic mechanism behind RNA:m5C methylation. Here, we have solved the structures of NSun6, a human tRNA:m5C methyltransferase, in the apo form and in complex with a full-length tRNA substrate. These structures show a non-canonical conformation of the bound tRNA, rendering the base moiety of the target cytosine accessible to the enzyme for methylation. Further biochemical assays reveal the critical, but distinct, roles of two conserved cysteine residues for the RNA:m5C methylation. Collectively, for the first time, we have solved the complex structure of a RNA:m5C methyltransferase and addressed the catalytic mechanism of the RNA:m5C methyltransferase family, which may allow for structure-based drug design toward RNA:m5C methyltransferase-related diseases.


Assuntos
tRNA Metiltransferases/química , Sequência de Aminoácidos , Apoenzimas/química , Biocatálise , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , RNA de Transferência/química , Especificidade por Substrato
12.
J Biol Chem ; 291(12): 6507-20, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26811336

RESUMO

Mitochondria require all translational components, including aminoacyl-tRNA synthetases (aaRSs), to complete organelle protein synthesis. Some aaRS mutations cause mitochondrial disorders, including human mitochondrial threonyl-tRNA synthetase (hmtThrRS) (encoded by TARS2), the P282L mutation of which causes mitochondrial encephalomyopathies. However, its catalytic and structural consequences remain unclear. Herein, we cloned TARS2 and purified the wild-type and P282L mutant hmtThrRS. hmtThrRS misactivates non-cognate Ser and uses post-transfer editing to clear erroneously synthesized products. In vitro and in vivo analyses revealed that the mutation induces a decrease in Thr activation, aminoacylation, and proofreading activities and a change in the protein structure and/or stability, which might cause reduced catalytic efficiency. We also identified a splicing variant of TARS2 mRNA lacking exons 8 and 9, the protein product of which is targeted into mitochondria. In HEK293T cells, the variant does not dimerize and cannot complement the ThrRS knock-out strain in yeast, suggesting that the truncated protein is inactive and might have a non-canonical function, as observed for other aaRS fragments. The present study describes the aminoacylation and editing properties of hmtThrRS, clarifies the molecular consequences of the P282L mutation, and shows that the yeast ThrRS-deletion model is suitable to test pathology-associated point mutations or alternative splicing variants of mammalian aaRS mRNAs.


Assuntos
Encefalomiopatias Mitocondriais/genética , Treonina-tRNA Ligase/genética , Monofosfato de Adenosina/química , Processamento Alternativo , Sequência de Aminoácidos , Ativação Enzimática , Estabilidade Enzimática , Teste de Complementação Genética , Células HEK293 , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Mitocôndrias/enzimologia , Modelos Moleculares , Dados de Sequência Molecular , Mutação Puntual , Multimerização Proteica , Transporte Proteico , Saccharomyces cerevisiae/genética , Serina/química , Treonina/química , Treonina-tRNA Ligase/química , Treonina-tRNA Ligase/metabolismo , Aminoacilação de RNA de Transferência
13.
J Biol Chem ; 291(46): 24293-24303, 2016 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-27703015

RESUMO

Human NSun6 is an RNA methyltransferase that catalyzes the transfer of the methyl group from S-adenosyl-l-methionine (SAM) to C72 of tRNAThr and tRNACys In the current study, we used mass spectrometry to demonstrate that human NSun6 indeed introduces 5-methylcytosine (m5C) into tRNA, as expected. To further reveal the tRNA recognition mechanism of human NSun6, we measured the methylation activity of human NSun6 and its kinetic parameters for different tRNA substrates and their mutants. We showed that human NSun6 requires a well folded, full-length tRNA as its substrate. In the acceptor region, the CCA terminus, the target site C72, the discriminator base U73, and the second and third base pairs (2:71 and 3:70) of the acceptor stem are all important RNA recognition elements for human NSun6. In addition, two specific base pairs (11:24 and 12:23) in the D-stem of the tRNA substrate are involved in interacting with human NSun6. Together, our findings suggest that human NSun6 relies on a delicate network for RNA recognition, which involves both the primary sequence and tertiary structure of tRNA substrates.


Assuntos
Dobramento de Proteína , RNA de Transferência de Treonina/química , tRNA Metiltransferases/química , Humanos , Metilação , Domínios Proteicos , RNA de Transferência de Treonina/genética , RNA de Transferência de Treonina/metabolismo , tRNA Metiltransferases/genética , tRNA Metiltransferases/metabolismo
14.
J Biol Chem ; 291(40): 21208-21221, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27542414

RESUMO

Translational fidelity mediated by aminoacyl-tRNA synthetases ensures the generation of the correct aminoacyl-tRNAs, which is critical for most species. Threonyl-tRNA synthetase (ThrRS) contains multiple domains, including an N2 editing domain. Of the ThrRS domains, N1 is the last to be assigned a function. Here, we found that ThrRSs from Mycoplasma species exhibit differences in their domain composition and editing active sites compared with the canonical ThrRSs. The Mycoplasma mobile ThrRS, the first example of a ThrRS naturally lacking the N1 domain, displays efficient post-transfer editing activity. In contrast, the Mycoplasma capricolum ThrRS, which harbors an N1 domain and a degenerate N2 domain, is editing-defective. Only editing-capable ThrRSs were able to support the growth of a yeast thrS deletion strain (ScΔthrS), thus suggesting that ScΔthrS is an excellent tool for studying the in vivo editing of introduced bacterial ThrRSs. On the basis of the presence or absence of an N1 domain, we further revealed the crucial importance of the only absolutely conserved residue within the N1 domain in regulating editing by mediating an N1-N2 domain interaction in Escherichia coli ThrRS. Our results reveal the translational quality control of various ThrRSs and the role of the N1 domain in translational fidelity.


Assuntos
Proteínas de Bactérias , Mycoplasma capricolum , Biossíntese de Proteínas/fisiologia , Treonina-tRNA Ligase , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Deleção de Genes , Teste de Complementação Genética , Mycoplasma capricolum/enzimologia , Mycoplasma capricolum/genética , Domínios Proteicos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Especificidade da Espécie , Treonina-tRNA Ligase/genética , Treonina-tRNA Ligase/metabolismo
15.
Nucleic Acids Res ; 43(15): 7489-503, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26202969

RESUMO

TrmJ proteins from the SPOUT methyltransferase superfamily are tRNA Xm32 modification enzymes that occur in bacteria and archaea. Unlike archaeal TrmJ, bacterial TrmJ require full-length tRNA molecules as substrates. It remains unknown how bacterial TrmJs recognize substrate tRNAs and specifically catalyze a 2'-O modification at ribose 32. Herein, we demonstrate that all six Escherichia coli (Ec) tRNAs with 2'-O-methylated nucleosides at position 32 are substrates of EcTrmJ, and we show that the elbow region of tRNA, but not the amino acid acceptor stem, is needed for the methylation reaction. Our crystallographic study reveals that full-length EcTrmJ forms an unusual dimer in the asymmetric unit, with both the catalytic SPOUT domain and C-terminal extension forming separate dimeric associations. Based on these findings, we used electrophoretic mobility shift assay, isothermal titration calorimetry and enzymatic methods to identify amino acids within EcTrmJ that are involved in tRNA binding. We found that tRNA recognition by EcTrmJ involves the cooperative influences of conserved residues from both the SPOUT and extensional domains, and that this process is regulated by the flexible hinge region that connects these two domains.


Assuntos
Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Metiltransferases/química , RNA de Transferência/metabolismo , tRNA Metiltransferases/química , Motivos de Aminoácidos , Proteínas de Escherichia coli/metabolismo , Metiltransferases/metabolismo , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica , Estrutura Terciária de Proteína , RNA de Transferência/química , S-Adenosil-Homocisteína/química , S-Adenosil-Homocisteína/metabolismo , tRNA Metiltransferases/metabolismo
16.
Antimicrob Agents Chemother ; 60(10): 5817-27, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27431220

RESUMO

The apicomplexan parasites Cryptosporidium and Toxoplasma are serious threats to human health. Cryptosporidiosis is a severe diarrheal disease in malnourished children and immunocompromised individuals, with the only FDA-approved drug treatment currently being nitazoxanide. The existing therapies for toxoplasmosis, an important pathology in immunocompromised individuals and pregnant women, also have serious limitations. With the aim of developing alternative therapeutic options to address these health problems, we tested a number of benzoxaboroles, boron-containing compounds shown to be active against various infectious agents, for inhibition of the growth of Cryptosporidium parasites in mammalian cells. A 3-aminomethyl benzoxaborole, AN6426, with activity in the micromolar range and with activity comparable to that of nitazoxanide, was identified and further characterized using biophysical measurements of affinity and crystal structures of complexes with the editing domain of Cryptosporidium leucyl-tRNA synthetase (LeuRS). The same compound was shown to be active against Toxoplasma parasites, with the activity being enhanced in the presence of norvaline, an amino acid that can be mischarged by LeuRS. Our observations are consistent with AN6426 inhibiting protein synthesis in both Cryptosporidium and Toxoplasma by forming a covalent adduct with tRNA(Leu) in the LeuRS editing active site and suggest that further exploitation of the benzoxaborole scaffold is a valid strategy to develop novel, much needed antiparasitic agents.


Assuntos
Antiprotozoários/farmacologia , Compostos de Boro/farmacologia , Cryptosporidium parvum/efeitos dos fármacos , Leucina-tRNA Ligase/antagonistas & inibidores , Leucina-tRNA Ligase/química , Toxoplasma/efeitos dos fármacos , Animais , Antiprotozoários/química , Antiprotozoários/metabolismo , Compostos de Boro/química , Cristalografia por Raios X , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/parasitologia , Humanos , Leucina-tRNA Ligase/metabolismo , Células Madin Darby de Rim Canino/parasitologia , Simulação de Acoplamento Molecular , Conformação Proteica
17.
Nucleic Acids Res ; 42(22): 13873-86, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25414329

RESUMO

Yeast mitochondria contain a minimalist threonyl-tRNA synthetase (ThrRS) composed only of the catalytic core and tRNA binding domain but lacking the entire editing domain. Besides the usual tRNA(Thr)2, some budding yeasts, such as Saccharomyces cerevisiae, also contain a non-canonical tRNA(Thr)1 with an enlarged 8-nucleotide anticodon loop, reprograming the usual leucine CUN codons to threonine. This raises interesting questions about the aminoacylation fidelity of such ThrRSs and the possible contribution of the two tRNA(Thr)s during editing. Here, we found that, despite the absence of the editing domain, S. cerevisiae mitochondrial ThrRS (ScmtThrRS) harbors a tRNA-dependent pre-transfer editing activity. Remarkably, only the usual tRNA(Thr)2 stimulated pre-transfer editing, thus, establishing the first example of a synthetase exhibiting tRNA-isoacceptor specificity during pre-transfer editing. We also showed that the failure of tRNA(Thr)1 to stimulate tRNA-dependent pre-transfer editing was due to the lack of an editing domain. Using assays of the complementation of a ScmtThrRS gene knockout strain, we showed that the catalytic core and tRNA binding domain of ScmtThrRS co-evolved to recognize the unusual tRNA(Thr)1. In combination, the results provide insights into the tRNA-dependent editing process and suggest that tRNA-dependent pre-transfer editing takes place in the aminoacylation catalytic core.


Assuntos
Mitocôndrias/enzimologia , RNA de Transferência de Treonina/metabolismo , Treonina-tRNA Ligase/metabolismo , Aminoacilação de RNA de Transferência , Anticódon , Evolução Molecular , Deleção de Genes , Estrutura Terciária de Proteína , RNA de Transferência de Treonina/química , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Treonina-tRNA Ligase/química , Treonina-tRNA Ligase/genética
18.
Nucleic Acids Res ; 42(8): 5109-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24500203

RESUMO

Leucyl-tRNA (transfer RNA) synthetase (LeuRS) is a multi-domain enzyme, which is divided into bacterial and archaeal/eukaryotic types. In general, one specific LeuRS, the domains of which are of the same type, exists in a single cell compartment. However, some species, such as the haloalkaliphile Natrialba magadii, encode two cytoplasmic LeuRSs, NmLeuRS1 and NmLeuRS2, which are the first examples of naturally occurring chimeric enzymes with different domains of bacterial and archaeal types. Furthermore, N. magadii encodes typical archaeal tRNA(Leu)s. The tRNA recognition mode, aminoacylation and translational quality control activities of these two LeuRSs are interesting questions to be addressed. Herein, active NmLeuRS1 and NmLeuRS2 were successfully purified after gene expression in Escherichia coli. Under the optimized aminoacylation conditions, we discovered that they distinguished cognate NmtRNA(Leu) in the archaeal mode, whereas the N-terminal region was of the bacterial type. However, NmLeuRS1 exhibited much higher aminoacylation and editing activity than NmLeuRS2, suggesting that NmLeuRS1 is more likely to generate Leu-tRNA(Leu) for protein biosynthesis. Moreover, using NmLeuRS1 as a model, we demonstrated misactivation of several non-cognate amino acids, and accuracy of protein synthesis was maintained mainly via post-transfer editing. This comprehensive study of the NmLeuRS/tRNA(Leu) system provides a detailed understanding of the coevolution of aminoacyl-tRNA synthetases and tRNA.


Assuntos
Halobacteriaceae/enzimologia , Leucina-tRNA Ligase/metabolismo , RNA de Transferência de Leucina/metabolismo , Aminoacilação de RNA de Transferência , Aminoácidos/metabolismo , Bactérias/enzimologia , Halobacteriaceae/genética , Concentração de Íons de Hidrogênio , Leucina-tRNA Ligase/química , Leucina-tRNA Ligase/classificação , Cloreto de Potássio , Estrutura Terciária de Proteína
19.
RNA Biol ; 12(8): 900-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26106808

RESUMO

Post-transcriptional modifications bring chemical diversity to tRNAs, especially at positions 34 and 37 of the anticodon stem-loop (ASL). TrmL is the prokaryotic methyltransferase that catalyzes the transfer of the methyl group from S-adenosyl-L-methionine to the wobble base of tRNA(Leu)CAA and tRNA(Leu)UAA isoacceptors. This Cm34/Um34 modification affects codon-anticodon interactions and is essential for translational fidelity. TrmL-catalyzed 2'-O-methylation requires its homodimerization; however, understanding of the tRNA recognition mechanism by TrmL remains elusive. In the current study, by measuring tRNA methylation by TrmL and performing kinetic analysis of tRNA mutants, we found that TrmL exhibits a fine-tuned tRNA substrate recognition mechanism. Anticodon stem-loop minihelices with an extension of 2 base pairs are the minimal substrate for EcTrmL methylation. A35 is a key residue for TrmL recognition, while A36-A37-A38 are important either via direct interaction with TrmL or due to the necessity for prior isopentenylation (i(6)) at A37. In addition, TrmL only methylates pyrimidines but not purine residues at the wobble position, and the 2'-O-methylation relies on prior N(6)-isopentenyladenosine modification at position 37.


Assuntos
Anticódon/genética , Códon/genética , Proteínas de Escherichia coli/genética , Metiltransferases/genética , RNA de Transferência de Leucina/genética , Alcenos/metabolismo , Anticódon/química , Anticódon/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Biocatálise , Códon/química , Códon/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Cinética , Metilação , Metiltransferases/química , Metiltransferases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Conformação de Ácido Nucleico , Multimerização Proteica , Pirimidinas/metabolismo , RNA de Transferência de Leucina/química , RNA de Transferência de Leucina/metabolismo , S-Adenosilmetionina/metabolismo , Especificidade por Substrato
20.
Nucleic Acids Res ; 41(16): 7828-42, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23804755

RESUMO

Unlike other transfer RNAs (tRNA)-modifying enzymes from the SPOUT methyltransferase superfamily, the tRNA (Um34/Cm34) methyltransferase TrmL lacks the usual extension domain for tRNA binding and consists only of a SPOUT domain. Both the catalytic and tRNA recognition mechanisms of this enzyme remain elusive. By using tRNAs purified from an Escherichia coli strain with the TrmL gene deleted, we found that TrmL can independently catalyze the methyl transfer from S-adenosyl-L-methionine to and isoacceptors without the involvement of other tRNA-binding proteins. We have solved the crystal structures of TrmL in apo form and in complex with S-adenosyl-homocysteine and identified the cofactor binding site and a possible active site. Methyltransferase activity and tRNA-binding affinity of TrmL mutants were measured to identify residues important for tRNA binding of TrmL. Our results suggest that TrmL functions as a homodimer by using the conserved C-terminal half of the SPOUT domain for catalysis, whereas residues from the less-conserved N-terminal half of the other subunit participate in tRNA recognition.


Assuntos
Proteínas de Escherichia coli/química , Metiltransferases/química , RNA de Transferência de Leucina/metabolismo , Sequência de Aminoácidos , Aminoácidos Básicos/química , Domínio Catalítico , Coenzimas/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Metiltransferases/genética , Metiltransferases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Multimerização Proteica , RNA de Transferência de Leucina/química , S-Adenosil-Homocisteína/química , S-Adenosilmetionina/metabolismo , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA