Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Nature ; 613(7943): 383-390, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36599982

RESUMEN

Specific, regulated modification of RNAs is important for proper gene expression1,2. tRNAs are rich with various chemical modifications that affect their stability and function3,4. 7-Methylguanosine (m7G) at tRNA position 46 is a conserved modification that modulates steady-state tRNA levels to affect cell growth5,6. The METTL1-WDR4 complex generates m7G46 in humans, and dysregulation of METTL1-WDR4 has been linked to brain malformation and multiple cancers7-22. Here we show how METTL1 and WDR4 cooperate to recognize RNA substrates and catalyse methylation. A crystal structure of METTL1-WDR4 and cryo-electron microscopy structures of METTL1-WDR4-tRNA show that the composite protein surface recognizes the tRNA elbow through shape complementarity. The cryo-electron microscopy structures of METTL1-WDR4-tRNA with S-adenosylmethionine or S-adenosylhomocysteine along with METTL1 crystal structures provide additional insights into the catalytic mechanism by revealing the active site in multiple states. The METTL1 N terminus couples cofactor binding with conformational changes in the tRNA, the catalytic loop and the WDR4 C terminus, acting as the switch to activate m7G methylation. Thus, our structural models explain how post-translational modifications of the METTL1 N terminus can regulate methylation. Together, our work elucidates the core and regulatory mechanisms underlying m7G modification by METTL1, providing the framework to understand its contribution to biology and disease.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de Unión al GTP , Metilación , Metiltransferasas , Procesamiento Postranscripcional del ARN , ARN de Transferencia , Humanos , Dominio Catalítico , Cristalografía por Rayos X , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/ultraestructura , Metiltransferasas/química , Metiltransferasas/metabolismo , Metiltransferasas/ultraestructura , ARN de Transferencia/química , ARN de Transferencia/metabolismo , ARN de Transferencia/ultraestructura , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Especificidad por Sustrato , Biocatálisis
2.
Nucleic Acids Res ; 50(18): 10201-10211, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-35882385

RESUMEN

Ribosomes are remarkable in their malleability to accept diverse aminoacyl-tRNA substrates from both the same organism and other organisms or domains of life. This is a critical feature of the ribosome that allows the use of orthogonal translation systems for genetic code expansion. Optimization of these orthogonal translation systems generally involves focusing on the compatibility of the tRNA, aminoacyl-tRNA synthetase, and a non-canonical amino acid with each other. As we expand the diversity of tRNAs used to include non-canonical structures, the question arises as to the tRNA suitability on the ribosome. Specifically, we investigated the ribosomal translation of allo-tRNAUTu1, a uniquely shaped (9/3) tRNA exploited for site-specific selenocysteine insertion, using single-molecule fluorescence. With this technique we identified ribosomal disassembly occurring from translocation of allo-tRNAUTu1 from the A to the P site. Using cryo-EM to capture the tRNA on the ribosome, we pinpointed a distinct tertiary interaction preventing fluid translocation. Through a single nucleotide mutation, we disrupted this tertiary interaction and relieved the translation roadblock. With the continued diversification of genetic code expansion, our work highlights a targeted approach to optimize translation by distinct tRNAs as they move through the ribosome.


Continued expansion of the genetic code has required the use of synthetic tRNAs for decoding. Some of these synthetic tRNAs have unique structural features that are not observed in canonical tRNAs. Here, the authors applied single-molecule, biochemical and structural methods to determine whether these distinct features were deleterious for efficient protein translation on the ribosome. With a focus on selenocysteine insertion, the authors explored an allo-tRNA with a 9/3 acceptor domain. They observed a translational roadblock that occurred in A to P site tRNA translocation. This block was mediated by a tertiary interaction across the tRNA core, directing the variable arm position into an unfavorable conformation. A single-nucleotide mutation disrupted this interaction, providing flexibility in the variable arm and promoting efficient protein production.


Asunto(s)
Biosíntesis de Proteínas , ARN de Transferencia/ultraestructura , Ribosomas/ultraestructura , Aminoácidos/genética , Aminoacil-ARNt Sintetasas/genética , Nucleótidos/metabolismo , ARN de Transferencia/metabolismo , Ribosomas/metabolismo , Selenocisteína/química
3.
Nucleic Acids Res ; 49(14): 8309-8323, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34268557

RESUMEN

tRNAs harbor the most diverse posttranscriptional modifications. The 3-methylcytidine (m3C) is widely distributed at position C32 (m3C32) of eukaryotic tRNAThr and tRNASer species. m3C32 is decorated by the single methyltransferase Trm140 in budding yeasts; however, two (Trm140 and Trm141 in fission yeasts) or three enzymes (METTL2A, METTL2B and METTL6 in mammals) are involved in its biogenesis. The rationale for the existence of multiple m3C32 methyltransferases and their substrate discrimination mechanism is hitherto unknown. Here, we revealed that both METTL2A and METTL2B are expressed in vivo. We purified human METTL2A, METTL2B, and METTL6 to high homogeneity. We successfully reconstituted m3C32 modification activity for tRNAThr by METT2A and for tRNASer(GCU) by METTL6, assisted by seryl-tRNA synthetase (SerRS) in vitro. Compared with METTL2A, METTL2B exhibited dramatically lower activity in vitro. Both G35 and t6A at position 37 (t6A37) are necessary but insufficient prerequisites for tRNAThr m3C32 formation, while the anticodon loop and the long variable arm, but not t6A37, are key determinants for tRNASer(GCU) m3C32 biogenesis, likely being recognized synergistically by METTL6 and SerRS, respectively. Finally, we proposed a mutually exclusive substrate selection model to ensure correct discrimination among multiple tRNAs by multiple m3C32 methyltransferases.


Asunto(s)
Conformación de Ácido Nucleico , ARN de Transferencia/genética , ARNt Metiltransferasas/genética , Anticodón/genética , Citidina/análogos & derivados , Citidina/genética , Humanos , ARN/genética , ARN de Transferencia/ultraestructura , Serina-ARNt Ligasa/genética , Especificidad por Sustrato
4.
Nucleic Acids Res ; 49(14): 8247-8260, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34244755

RESUMEN

Transfer RNAs (tRNAs) are key players in protein synthesis. To be fully active, tRNAs undergo extensive post-transcriptional modifications, including queuosine (Q), a hypermodified 7-deaza-guanosine present in the anticodon of several tRNAs in bacteria and eukarya. Here, molecular and biochemical approaches revealed that in the protozoan parasite Trypanosoma brucei, Q-containing tRNAs have a preference for the U-ending codons for asparagine, aspartate, tyrosine and histidine, analogous to what has been described in other systems. However, since a lack of tRNA genes in T. brucei mitochondria makes it essential to import a complete set from the cytoplasm, we surprisingly found that Q-modified tRNAs are preferentially imported over their unmodified counterparts. In turn, their absence from mitochondria has a pronounced effect on organellar translation and affects function. Although Q modification in T. brucei is globally important for codon selection, it is more so for mitochondrial protein synthesis. These results provide a unique example of the combined regulatory effect of codon usage and wobble modifications on protein synthesis; all driven by tRNA intracellular transport dynamics.


Asunto(s)
Mitocondrias/genética , Conformación de Ácido Nucleico , Nucleósido Q/genética , ARN de Transferencia/genética , Anticodón/genética , Núcleo Celular/genética , Núcleo Celular/ultraestructura , Codón/genética , Citoplasma/genética , Citoplasma/ultraestructura , Guanosina/genética , Biosíntesis de Proteínas/genética , Procesamiento Postranscripcional del ARN/genética , ARN de Transferencia/ultraestructura , Trypanosoma brucei brucei/genética
5.
FEBS Lett ; 595(7): 913-924, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33460451

RESUMEN

The mechanism and evolution of the recognition scheme between key components of the translation system, that is, tRNAs, synthetases, and elongation factors, are fundamental issues in understanding the translation of genetic information into proteins. Statistical analysis of bacterial tRNA sequences reveals that for six amino acids, a string of 10 nucleotides preceding the tRNA 3' end carries cognate coding triplets to nearly full extent. The triplets conserved in positions 63-67 are implicated in the recognition by the elongation factor EF-Tu, and those conserved in positions 68-72, in the identification of cognate tRNAs, and their derived minihelices by class IIa synthetases. These coding triplets are suggested to have primordial origin, being engaged in aminoacylation of prebiotic tRNAs and in the establishment of the canonical codon set.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Evolución Molecular , Factor Tu de Elongación Peptídica/genética , ARN de Transferencia/genética , Aminoácidos/genética , Aminoacil-ARNt Sintetasas/ultraestructura , Aminoacilación/genética , Codón/genética , Escherichia coli/genética , Código Genético/genética , Conformación de Ácido Nucleico , Factor Tu de Elongación Peptídica/ultraestructura , ARN de Transferencia/ultraestructura
6.
Elife ; 92020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32924932

RESUMEN

Using cryo-electron microscopy (cryo-EM), we determined the structure of the Escherichia coli 70S ribosome with a global resolution of 2.0 Å. The maps reveal unambiguous positioning of protein and RNA residues, their detailed chemical interactions, and chemical modifications. Notable features include the first examples of isopeptide and thioamide backbone substitutions in ribosomal proteins, the former likely conserved in all domains of life. The maps also reveal extensive solvation of the small (30S) ribosomal subunit, and interactions with A-site and P-site tRNAs, mRNA, and the antibiotic paromomycin. The maps and models of the bacterial ribosome presented here now allow a deeper phylogenetic analysis of ribosomal components including structural conservation to the level of solvation. The high quality of the maps should enable future structural analyses of the chemical basis for translation and aid the development of robust tools for cryo-EM structure modeling and refinement.


Inside cells, proteins are produced by complex molecular machines called ribosomes. Techniques that allow scientists to visualize ribosomes at the atomic level, such as cryogenic electron microscopy (cryo-EM), help shed light on the structure of these molecular machines, revealing details of how they build proteins. Understanding how ribosomes work has many benefits, including the development of new antibiotics that can kill bacteria without affecting animal cells. Watson et al. used cryo-EM techniques with increased resolution to examine the ribosomes of the bacterium Escherichia coli in a higher level of detail than has been seen before. The results revealed two chemical modifications in proteins that form the ribosome that had not been observed in ribosomes previously. Additionally, a protein segment with a previously undescribed structure was identified close to the site where the ribosome reads the genetic instructions needed to make proteins. Further genetic analyses suggested these structures are in many related species, and may play important roles in how the ribosome works. Watson et al. were also able to see how paromomycin, an antibiotic used to treat parasitic infections, is positioned in the ribosome. The antibiotic interacts with a site near where the genetic code is read out, which might explain why certain changes to the antibiotic can interfere with its potency. Finally, the new ribosome structure reveals thousands of water molecules and metal ions that help keep the ribosome together as it produces proteins. This study shows the value of advances in cryo-EM technology and illustrates the importance of applying these techniques to other cell components. The results also reveal details of the ribosome useful for further research into this essential molecular machine.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Escherichia coli/ultraestructura , ARN Bacteriano/ultraestructura , Proteínas Ribosómicas/ultraestructura , Ribosomas/ultraestructura , Microscopía por Crioelectrón , ARN Mensajero/ultraestructura , ARN de Transferencia/ultraestructura
7.
RNA ; 26(12): 1755-1766, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32826323

RESUMEN

Ribonucleic acids (RNAs) play essential roles in living cells. Many of them fold into defined three-dimensional (3D) structures to perform functions. Recent advances in single-particle cryo-electron microscopy (cryo-EM) have enabled structure determinations of RNA to atomic resolutions. However, most RNA molecules are structurally flexible, limiting the resolution of their structures solved by cryo-EM. In modeling these molecules, several computational methods are limited by the requirement of massive computational resources and/or the low efficiency in exploring large-scale structural variations. Here we use hierarchical natural move Monte Carlo (HNMMC), which takes advantage of collective motions for groups of nucleic acid residues, to refine RNA structures into their cryo-EM maps, preserving atomic details in the models. After validating the method on a simulated density map of tRNA, we applied it to objectively obtain the model of the folding intermediate for the specificity domain of ribonuclease P from Bacillus subtilis and refine a flexible ribosomal RNA (rRNA) expansion segment from the Mycobacterium tuberculosis (Mtb) ribosome in different conformational states. Finally, we used HNMMC to model atomic details and flexibility for two distinct conformations of the complete genomic RNA (gRNA) inside MS2, a single-stranded RNA virus, revealing multiple pathways for its capsid assembly.


Asunto(s)
Método de Montecarlo , Virus ARN/ultraestructura , ARN Ribosómico/ultraestructura , ARN de Transferencia/ultraestructura , ARN/ultraestructura , Ribosomas/ultraestructura , Bacillus subtilis/enzimología , Proteínas de la Cápside/genética , Proteínas de la Cápside/ultraestructura , Modelos Moleculares , ARN/genética , Virus ARN/genética , ARN Ribosómico/genética , ARN de Transferencia/genética , Ribonucleasa P/genética , Ribonucleasa P/ultraestructura , Ribosomas/genética
8.
Nature ; 584(7822): 640-645, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32612237

RESUMEN

Ribosomes accurately decode mRNA by proofreading each aminoacyl-tRNA that is delivered by the elongation factor EF-Tu1. To understand the molecular mechanism of this proofreading step it is necessary to visualize GTP-catalysed elongation, which has remained a challenge2-4. Here we use time-resolved cryogenic electron microscopy to reveal 33 ribosomal states after the delivery of aminoacyl-tRNA by EF-Tu•GTP. Instead of locking cognate tRNA upon initial recognition, the ribosomal decoding centre dynamically monitors codon-anticodon interactions before and after GTP hydrolysis. GTP hydrolysis enables the GTPase domain of EF-Tu to extend away, releasing EF-Tu from tRNA. The 30S subunit then locks cognate tRNA in the decoding centre and rotates, enabling the tRNA to bypass 50S protrusions during accommodation into the peptidyl transferase centre. By contrast, the decoding centre fails to lock near-cognate tRNA, enabling the dissociation of near-cognate tRNA both during initial selection (before GTP hydrolysis) and proofreading (after GTP hydrolysis). These findings reveal structural similarity between ribosomes in initial selection states5,6 and in proofreading states, which together govern the efficient rejection of incorrect tRNA.


Asunto(s)
Microscopía por Crioelectrón , Guanosina Trifosfato/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Ribosomas/metabolismo , Ribosomas/ultraestructura , Escherichia coli , GTP Fosfohidrolasas/metabolismo , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/química , Hidrólisis , Modelos Moleculares , Factor Tu de Elongación Peptídica/química , Factor Tu de Elongación Peptídica/ultraestructura , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Transferencia/química , ARN de Transferencia/ultraestructura , Ribosomas/química , Rotación
9.
Nucleic Acids Res ; 48(12): 6906-6918, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32459340

RESUMEN

The universal L-shaped tertiary structure of tRNAs is maintained with the help of nucleotide modifications within the D- and T-loops, and these modifications are most extensive within hyperthermophilic species. The obligate-commensal Nanoarchaeum equitans and its phylogenetically-distinct host Ignicoccus hospitalis grow physically coupled under identical hyperthermic conditions. We report here two fundamentally different routes by which these archaea modify the key conserved nucleotide U54 within their tRNA T-loops. In N. equitans, this nucleotide is methylated by the S-adenosylmethionine-dependent enzyme NEQ053 to form m5U54, and a recombinant version of this enzyme maintains specificity for U54 in Escherichia coli. In N. equitans, m5U54 is subsequently thiolated to form m5s2U54. In contrast, I. hospitalis isomerizes U54 to pseudouridine prior to methylating its N1-position and thiolating the O4-position of the nucleobase to form the previously uncharacterized nucleotide m1s4Ψ. The methyl and thiol groups in m1s4Ψ and m5s2U are presented within the T-loop in a spatially identical manner that stabilizes the 3'-endo-anti conformation of nucleotide-54, facilitating stacking onto adjacent nucleotides and reverse-Hoogsteen pairing with nucleotide m1A58. Thus, two distinct structurally-equivalent solutions have evolved independently and convergently to maintain the tertiary fold of tRNAs under extreme hyperthermic conditions.


Asunto(s)
Desulfurococcaceae/genética , Nanoarchaeota/genética , Conformación de Ácido Nucleico , ARN de Transferencia/ultraestructura , Archaea/genética , Archaea/ultraestructura , Escherichia coli/genética , Metilación , Filogenia , ARN de Transferencia/genética , ARNt Metiltransferasas/genética , ARNt Metiltransferasas/ultraestructura
10.
RNA ; 26(3): 278-289, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31848215

RESUMEN

Ubiquitous across all domains of life, tRNAs constitute an essential component of cellular physiology, carry out an indispensable role in protein synthesis, and have been historically the subject of a wide range of biochemical and biophysical studies as prototypical folded RNA molecules. Although conformational flexibility is a well-established characteristic of tRNA structure, it is typically regarded as an adaptive property exhibited in response to an inducing event, such as the binding of a tRNA synthetase or the accommodation of an aminoacyl-tRNA into the ribosome. In this study, we present crystallographic data of a tRNA molecule to expand on this paradigm by showing that structural flexibility and plasticity are intrinsic properties of tRNAs, apparent even in the absence of other factors. Based on two closely related conformations observed within the same crystal, we posit that unbound tRNAs by themselves are flexible and dynamic molecules. Furthermore, we demonstrate that the formation of the T-loop conformation by the tRNA TΨC stem-loop, a well-characterized and classic RNA structural motif, is possible even in the absence of important interactions observed in fully folded tRNAs.


Asunto(s)
Conformación de Ácido Nucleico , Aminoacil-ARN de Transferencia/ultraestructura , ARN de Transferencia/ultraestructura , Anticodón/química , Anticodón/genética , Cristalografía , Escherichia coli/química , Escherichia coli/ultraestructura , Motivos de Nucleótidos/genética , ARN de Transferencia/química , Aminoacil-ARN de Transferencia/química , Ribosomas/genética , Ribosomas/ultraestructura
12.
Nat Struct Mol Biol ; 26(12): 1094-1105, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31740854

RESUMEN

Amino acid availability in Gram-positive bacteria is monitored by T-box riboswitches. T-boxes directly bind tRNAs, assess their aminoacylation state, and regulate the transcription or translation of downstream genes to maintain nutritional homeostasis. Here, we report cocrystal and cryo-EM structures of Geobacillus kaustophilus and Bacillus subtilis T-box-tRNA complexes, detailing their multivalent, exquisitely selective interactions. The T-box forms a U-shaped molecular vise that clamps the tRNA, captures its 3' end using an elaborate 'discriminator' structure, and interrogates its aminoacylation state using a steric filter fashioned from a wobble base pair. In the absence of aminoacylation, T-boxes clutch tRNAs and form a continuously stacked central spine, permitting transcriptional readthrough or translation initiation. A modeled aminoacyl disrupts tRNA-T-box stacking, severing the central spine and blocking gene expression. Our data establish a universal mechanism of amino acid sensing on tRNAs and gene regulation by T-box riboswitches and exemplify how higher-order RNA-RNA interactions achieve multivalency and specificity.


Asunto(s)
Aminoácidos/metabolismo , Bacillus subtilis/metabolismo , Geobacillus/metabolismo , ARN Bacteriano/metabolismo , ARN de Transferencia/metabolismo , Riboswitch , Aminoacilación , Bacillus subtilis/química , Microscopía por Crioelectrón , Cristalografía por Rayos X , Geobacillus/química , Modelos Moleculares , Conformación de Ácido Nucleico , ARN Bacteriano/química , ARN Bacteriano/ultraestructura , ARN de Transferencia/química , ARN de Transferencia/ultraestructura
13.
EMBO J ; 38(21): e102226, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31609474

RESUMEN

Colony collapse disorder (CCD) is a multi-faceted syndrome decimating bee populations worldwide, and a group of viruses of the widely distributed Dicistroviridae family have been identified as a causing agent of CCD. This family of viruses employs non-coding RNA sequences, called internal ribosomal entry sites (IRESs), to precisely exploit the host machinery for viral protein production. Using single-particle cryo-electron microscopy (cryo-EM), we have characterized how the IRES of Israeli acute paralysis virus (IAPV) intergenic region captures and redirects translating ribosomes toward viral RNA messages. We reconstituted two in vitro reactions targeting a pre-translocation and a post-translocation state of the IAPV-IRES in the ribosome, allowing us to identify six structures using image processing classification methods. From these, we reconstructed the trajectory of IAPV-IRES from the early small subunit recruitment to the final post-translocated state in the ribosome. An early commitment of IRES/ribosome complexes for global pre-translocation mimicry explains the high efficiency observed for this IRES. Efforts directed toward fighting CCD by targeting the IAPV-IRES using RNA-interference technology are underway, and the structural framework presented here may assist in further refining these approaches.


Asunto(s)
Biomimética , Dicistroviridae/fisiología , Sitios Internos de Entrada al Ribosoma/genética , Biosíntesis de Proteínas , ARN de Transferencia/genética , ARN Viral/genética , Ribosomas/metabolismo , Microscopía por Crioelectrón , Dicistroviridae/ultraestructura , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , ARN de Transferencia/ultraestructura , Ribosomas/ultraestructura
14.
Nat Commun ; 10(1): 2617, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31197137

RESUMEN

Ribonuclease P (RNase P) is an essential ribozyme responsible for tRNA 5' maturation. Here we report the cryo-EM structures of Methanocaldococcus jannaschii (Mja) RNase P holoenzyme alone and in complex with a tRNA substrate at resolutions of 4.6 Å and 4.3 Å, respectively. The structures reveal that the subunits of MjaRNase P are strung together to organize the holoenzyme in a dimeric conformation required for efficient catalysis. The structures also show that archaeal RNase P is a functional chimera of bacterial and eukaryal RNase Ps that possesses bacterial-like two RNA-based anchors and a eukaryal-like protein-aided stabilization mechanism. The 3'-RCCA sequence of tRNA, which is a key recognition element for bacterial RNase P, is dispensable for tRNA recognition by MjaRNase P. The overall organization of MjaRNase P, particularly within the active site, is similar to those of bacterial and eukaryal RNase Ps, suggesting a universal catalytic mechanism for all RNase Ps.


Asunto(s)
Proteínas Arqueales/ultraestructura , Ribonucleasa P/ultraestructura , Proteínas Arqueales/metabolismo , Biocatálisis , Microscopía por Crioelectrón , Holoenzimas/ultraestructura , Methanocaldococcus/metabolismo , ARN de Transferencia/metabolismo , ARN de Transferencia/ultraestructura , Ribonucleasa P/metabolismo
15.
Biochem Biophys Res Commun ; 493(1): 240-245, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28911863

RESUMEN

The wyosine hypermodification found exclusively at G37 of tRNAPhe in eukaryotes and archaea is a very complicated process involving multiple steps and enzymes, and the derivatives are essential for the maintenance of the reading frame during translation. In the archaea Pyrococcus abyssi, two key enzymes from the Trm5 family, named PaTrm5a and PaTrm5b respectively, start the process by forming N1-methylated guanosine (m1G37). In addition, PaTrm5a catalyzes the further methylation of C7 on 4-demethylwyosine (imG-14) to produce isowyosine (imG2) at the same position. The structural basis of the distinct methylation capacities and possible conformational changes during catalysis displayed by the Trm5 enzymes are poorly studied. Here we report the 3.3 Å crystal structure of the mono-functional PaTrm5b, which shares 32% sequence identity with PaTrm5a. Interestingly, structural superposition reveals that the PaTrm5b protein exhibits an extended conformation similar to that of tRNA-bound Trm5b from Methanococcus jannaschii (MjTrm5b), but quite different from the open conformation of apo-PaTrm5a or well folded apo-MjTrm5b reported previously. Truncation of the N-terminal D1 domain leads to reduced tRNA binding as well as the methyltransfer activity of PaTrm5b. The differential positioning of the D1 domains from three reported Trm5 structures were rationalized, which could be attributable to the dissimilar inter-domain interactions and crystal packing patterns. This study expands our understanding on the methylation mechanism of the Trm5 enzymes and wyosine hypermodification.


Asunto(s)
Proteínas Arqueales/química , Proteínas Arqueales/ultraestructura , Methanococcus/enzimología , Metiltransferasas/química , Metiltransferasas/ultraestructura , Pyrococcus abyssi/enzimología , Sitios de Unión , Simulación por Computador , Activación Enzimática , Guanosina/análogos & derivados , Guanosina/química , Modelos Químicos , Modelos Moleculares , Unión Proteica , Conformación Proteica , ARN de Transferencia/química , ARN de Transferencia/ultraestructura , Especificidad de la Especie , Relación Estructura-Actividad
16.
Biomaterials ; 112: 44-61, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27744220

RESUMEN

Traditional therapeutic interventions against abnormal gene expression in disease states at the level of expressed proteins are becoming increasingly difficult due to poor selectivity, off-target effects and associated toxicity. Upstream catalytic targeting of specific RNA sequences offers an alternative platform for drug discovery to achieve more potent and selective treatment through antisense interference with disease-relevant RNAs. We report a novel class of catalytic biomaterials, comprising amphipathic RNA-cleaving peptides placed between two RNA recognition motifs, here demonstrated to target the TΨC loop and 3'- acceptor stem of tRNAPhe. These unique peptidyl-oligonucleotide 'dual' conjugates (DCs) were created by phosphoramidate or thiol-maleimide conjugation chemistry of a TΨC-targeting oligonucleotide to the N-terminus of the amphipathic peptide sequence, followed by amide coupling of a 3'-acceptor stem-targeting oligonucleotide to the free C-terminal carboxylic acid functionality of the same peptide. Hybridization of the DCs bearing two spatially-separated recognition motifs with the target tRNAPhe placed the peptide adjacent to a single-stranded RNA region and promoted cleavage within the 'action radius' of the catalytic peptide. Up to 100% cleavage of the target tRNAPhe was achieved by the best candidate (i.e. DC6) within 4 h, when conformational flexibility was introduced into the linker regions between the peptide and oligonucleotide components. This study provides the strong position for future development of highly selective RNA-targeting agents that can potentially be used for disease-selective treatment at the level of messenger, micro, and genomic viral RNA.


Asunto(s)
Marcación de Gen/métodos , Nanoconjugados/química , Nanoconjugados/ultraestructura , Péptidos/química , ARN de Transferencia/química , ARN de Transferencia/genética , Sitios de Unión , Catálisis , Reactivos de Enlaces Cruzados/química , Diseño de Fármacos , Unión Proteica , Conformación Proteica , ARN de Transferencia/ultraestructura , Relación Estructura-Actividad
17.
Proc Natl Acad Sci U S A ; 113(43): 12198-12201, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27729525

RESUMEN

The recent developments in cryo-EM have revolutionized our access to previously refractory structures. In particular, such studies of mammalian mitoribosomes have confirmed the absence of any 5S rRNA species and revealed the unexpected presence of a mitochondrially encoded tRNA (mt-tRNA) that usurps this position. Although the cryo-EM structures resolved the conundrum of whether mammalian mitoribosomes contain a 5S rRNA, they introduced a new dilemma: Why do human and porcine mitoribosomes integrate contrasting mt-tRNAs? Human mitoribosomes have been shown to integrate mt-tRNAVal compared with the porcine use of mt-tRNAPhe We have explored this observation further. Our studies examine whether a range of mt-tRNAs are used by different mammals, or whether the mt-tRNA selection is strictly limited to only these two species of the 22 tRNAs encoded by the mitochondrial genome (mtDNA); whether there is tissue-specific variation within a single organism; and what happens to the human mitoribosome when levels of the mt-tRNAVal are depleted. Our data demonstrate that only mt-tRNAVal or mt-tRNAPhe are found in the mitoribosomes of five different mammals, each mammal favors the same mt-tRNA in all tissue types, and strikingly, when steady-state levels of mt-tRNAVal are reduced, human mitoribosome biogenesis displays an adaptive response by switching to the incorporation of mt-tRNAPhe to generate translationally competent machinery.


Asunto(s)
Ribosomas Mitocondriales/química , Conformación de Ácido Nucleico , Biosíntesis de Proteínas/genética , ARN de Transferencia/ultraestructura , Animales , Microscopía por Crioelectrón , ADN Mitocondrial/química , ADN Mitocondrial/genética , Genoma Mitocondrial/genética , Humanos , Ribosomas Mitocondriales/ultraestructura , Procesamiento Postranscripcional del ARN/genética , ARN Ribosómico 5S/genética , ARN de Transferencia/genética , Porcinos
18.
Elife ; 52016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27434674

RESUMEN

Stringent response is a conserved bacterial stress response underlying virulence and antibiotic resistance. RelA/SpoT-homolog proteins synthesize transcriptional modulators (p)ppGpp, allowing bacteria to adapt to stress. RelA is activated during amino-acid starvation, when cognate deacyl-tRNA binds to the ribosomal A (aminoacyl-tRNA) site. We report four cryo-EM structures of E. coli RelA bound to the 70S ribosome, in the absence and presence of deacyl-tRNA accommodating in the 30S A site. The boomerang-shaped RelA with a wingspan of more than 100 Å wraps around the A/R (30S A-site/RelA-bound) tRNA. The CCA end of the A/R tRNA pins the central TGS domain against the 30S subunit, presenting the (p)ppGpp-synthetase domain near the 30S spur. The ribosome and A/R tRNA are captured in three conformations, revealing hitherto elusive states of tRNA engagement with the ribosomal decoding center. Decoding-center rearrangements are coupled with the step-wise 30S-subunit 'closure', providing insights into the dynamics of high-fidelity tRNA decoding.


Asunto(s)
Escherichia coli/fisiología , Ligasas/metabolismo , Ligasas/ultraestructura , ARN de Transferencia/metabolismo , ARN de Transferencia/ultraestructura , Ribosomas/metabolismo , Ribosomas/ultraestructura , Microscopía por Crioelectrón , Unión Proteica , Estrés Fisiológico
19.
Nature ; 534(7606): 277-280, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27279228

RESUMEN

In order to survive, bacteria continually sense, and respond to, environmental fluctuations. Stringent control represents a key bacterial stress response to nutrient starvation that leads to rapid and comprehensive reprogramming of metabolic and transcriptional patterns. In general, transcription of genes for growth and proliferation is downregulated, while those important for survival and virulence are upregulated. Amino acid starvation is sensed by depletion of the aminoacylated tRNA pools, and this results in accumulation of ribosomes stalled with non-aminoacylated (uncharged) tRNA in the ribosomal A site. RelA is recruited to stalled ribosomes and activated to synthesize a hyperphosphorylated guanosine analogue, (p)ppGpp, which acts as a pleiotropic secondary messenger. However, structural information about how RelA recognizes stalled ribosomes and discriminates against aminoacylated tRNAs is missing. Here we present the cryo-electron microscopy structure of RelA bound to the bacterial ribosome stalled with uncharged tRNA. The structure reveals that RelA utilizes a distinct binding site compared to the translational factors, with a multi-domain architecture that wraps around a highly distorted A-site tRNA. The TGS (ThrRS, GTPase and SpoT) domain of RelA binds the CCA tail to orient the free 3' hydroxyl group of the terminal adenosine towards a ß-strand, such that an aminoacylated tRNA at this position would be sterically precluded. The structure supports a model in which association of RelA with the ribosome suppresses auto-inhibition to activate synthesis of (p)ppGpp and initiate the stringent response. Since stringent control is responsible for the survival of pathogenic bacteria under stress conditions, and contributes to chronic infections and antibiotic tolerance, RelA represents a good target for the development of novel antibacterial therapeutics.


Asunto(s)
Aminoácidos/deficiencia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , GTP Pirofosfoquinasa/metabolismo , ARN de Transferencia/metabolismo , Ribosomas/metabolismo , Estrés Fisiológico , Adenosina/metabolismo , Sitios de Unión , Microscopía por Crioelectrón , Escherichia coli/metabolismo , Escherichia coli/patogenicidad , Escherichia coli/ultraestructura , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestructura , GTP Pirofosfoquinasa/antagonistas & inhibidores , GTP Pirofosfoquinasa/genética , GTP Pirofosfoquinasa/ultraestructura , Regulación Bacteriana de la Expresión Génica , Guanosina Tetrafosfato/química , Guanosina Tetrafosfato/metabolismo , Redes y Vías Metabólicas , Modelos Moleculares , Fosforilación , Biosíntesis de Proteínas , Conformación Proteica , Estructura Terciaria de Proteína , ARN de Transferencia/química , ARN de Transferencia/genética , ARN de Transferencia/ultraestructura , Aminoacil-ARN de Transferencia/química , Aminoacil-ARN de Transferencia/genética , Aminoacil-ARN de Transferencia/metabolismo , Ribosomas/química , Ribosomas/ultraestructura , Sistemas de Mensajero Secundario
20.
Proc Natl Acad Sci U S A ; 113(18): 4994-9, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27092003

RESUMEN

During translation, a plethora of protein factors bind to the ribosome and regulate protein synthesis. Many of those factors are guanosine triphosphatases (GTPases), proteins that catalyze the hydrolysis of guanosine 5'-triphosphate (GTP) to promote conformational changes. Despite numerous studies, the function of elongation factor 4 (EF-4/LepA), a highly conserved translational GTPase, has remained elusive. Here, we present the crystal structure at 2.6-Å resolution of the Thermus thermophilus 70S ribosome bound to EF-4 with a nonhydrolyzable GTP analog and A-, P-, and E-site tRNAs. The structure reveals the interactions of EF-4 with the A-site tRNA, including contacts between the C-terminal domain (CTD) of EF-4 and the acceptor helical stem of the tRNA. Remarkably, EF-4 induces a distortion of the A-site tRNA, allowing it to interact simultaneously with EF-4 and the decoding center of the ribosome. The structure provides insights into the tRNA-remodeling function of EF-4 on the ribosome and suggests that the displacement of the CCA-end of the A-site tRNA away from the peptidyl transferase center (PTC) is functionally significant.


Asunto(s)
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestructura , Factores de Iniciación de Péptidos/química , Factores de Iniciación de Péptidos/ultraestructura , ARN Bacteriano/química , ARN Bacteriano/ultraestructura , ARN de Transferencia/química , ARN de Transferencia/ultraestructura , Sitios de Unión , Simulación por Computador , Simulación del Acoplamiento Molecular , Conformación de Ácido Nucleico , Unión Proteica , Conformación Proteica , Motivos de Unión al ARN , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/ultraestructura , Ribosomas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA