Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Cell ; 186(25): 5517-5535.e24, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-37992713

RESUMEN

Transfer RNA (tRNA) modifications are critical for protein synthesis. Queuosine (Q), a 7-deaza-guanosine derivative, is present in tRNA anticodons. In vertebrate tRNAs for Tyr and Asp, Q is further glycosylated with galactose and mannose to generate galQ and manQ, respectively. However, biogenesis and physiological relevance of Q-glycosylation remain poorly understood. Here, we biochemically identified two RNA glycosylases, QTGAL and QTMAN, and successfully reconstituted Q-glycosylation of tRNAs using nucleotide diphosphate sugars. Ribosome profiling of knockout cells revealed that Q-glycosylation slowed down elongation at cognate codons, UAC and GAC (GAU), respectively. We also found that galactosylation of Q suppresses stop codon readthrough. Moreover, protein aggregates increased in cells lacking Q-glycosylation, indicating that Q-glycosylation contributes to proteostasis. Cryo-EM of human ribosome-tRNA complex revealed the molecular basis of codon recognition regulated by Q-glycosylations. Furthermore, zebrafish qtgal and qtman knockout lines displayed shortened body length, implying that Q-glycosylation is required for post-embryonic growth in vertebrates.


Asunto(s)
ARN de Transferencia , Animales , Humanos , Ratas , Anticodón , Línea Celular , Codón , Glicosilación , Nucleósido Q/química , Nucleósido Q/genética , Nucleósido Q/metabolismo , ARN de Transferencia/química , ARN de Transferencia/metabolismo , Porcinos , Pez Cebra/metabolismo , Conformación de Ácido Nucleico
2.
Cell ; 164(5): 962-73, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26919431

RESUMEN

PIWI-interacting RNAs (piRNAs) play a crucial role in transposon silencing in animal germ cells. In piRNA biogenesis, single-stranded piRNA intermediates are loaded into PIWI-clade proteins and cleaved by Zucchini/MitoPLD, yielding precursor piRNAs (pre-piRNAs). Pre-piRNAs that are longer than the mature piRNA length are then trimmed at their 3' ends. Although recent studies implicated the Tudor domain protein Papi/Tdrkh in pre-piRNA trimming, the identity of Trimmer and its relationship with Papi/Tdrkh remain unknown. Here, we identified PNLDC1, an uncharacterized 3'-5' exonuclease, as Trimmer in silkworms. Trimmer is enriched in the mitochondrial fraction and binds to Papi/Tdrkh. Depletion of Trimmer and Papi/Tdrkh additively inhibits trimming, causing accumulation of ∼35-40-nt pre-piRNAs that are impaired for target cleavage and prone to degradation. Our results highlight the cooperative action of Trimmer and Papi/Tdrkh in piRNA maturation.


Asunto(s)
Bombyx/enzimología , Bombyx/genética , Proteínas de Insectos/metabolismo , Procesamiento Postranscripcional del ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Bombyx/metabolismo , Mitocondrias/metabolismo
3.
Nature ; 616(7956): 390-397, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37020030

RESUMEN

The class 2 type V CRISPR effector Cas12 is thought to have evolved from the IS200/IS605 superfamily of transposon-associated TnpB proteins1. Recent studies have identified TnpB proteins as miniature RNA-guided DNA endonucleases2,3. TnpB associates with a single, long RNA (ωRNA) and cleaves double-stranded DNA targets complementary to the ωRNA guide. However, the RNA-guided DNA cleavage mechanism of TnpB and its evolutionary relationship with Cas12 enzymes remain unknown. Here we report the cryo-electron microscopy (cryo-EM) structure of Deinococcus radiodurans ISDra2 TnpB in complex with its cognate ωRNA and target DNA. In the structure, the ωRNA adopts an unexpected architecture and forms a pseudoknot, which is conserved among all guide RNAs of Cas12 enzymes. Furthermore, the structure, along with our functional analysis, reveals how the compact TnpB recognizes the ωRNA and cleaves target DNA complementary to the guide. A structural comparison of TnpB with Cas12 enzymes suggests that CRISPR-Cas12 effectors acquired an ability to recognize the protospacer-adjacent motif-distal end of the guide RNA-target DNA heteroduplex, by either asymmetric dimer formation or diverse REC2 insertions, enabling engagement in CRISPR-Cas adaptive immunity. Collectively, our findings provide mechanistic insights into TnpB function and advance our understanding of the evolution from transposon-encoded TnpB proteins to CRISPR-Cas12 effectors.


Asunto(s)
Proteínas Bacterianas , Microscopía por Crioelectrón , Elementos Transponibles de ADN , Deinococcus , Endodesoxirribonucleasas , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Proteínas Asociadas a CRISPR/química , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , ADN/química , ADN/genética , ADN/metabolismo , ADN/ultraestructura , Elementos Transponibles de ADN/genética , ARN Guía de Sistemas CRISPR-Cas/química , ARN Guía de Sistemas CRISPR-Cas/genética , ARN Guía de Sistemas CRISPR-Cas/metabolismo , ARN Guía de Sistemas CRISPR-Cas/ultraestructura , Endodesoxirribonucleasas/química , Endodesoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/ultraestructura , Deinococcus/enzimología , Deinococcus/genética , Especificidad por Sustrato
4.
Mol Cell ; 81(15): 3160-3170.e9, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34174184

RESUMEN

RelA-SpoT Homolog (RSH) enzymes control bacterial physiology through synthesis and degradation of the nucleotide alarmone (p)ppGpp. We recently discovered multiple families of small alarmone synthetase (SAS) RSH acting as toxins of toxin-antitoxin (TA) modules, with the FaRel subfamily of toxSAS abrogating bacterial growth by producing an analog of (p)ppGpp, (pp)pApp. Here we probe the mechanism of growth arrest used by four experimentally unexplored subfamilies of toxSAS: FaRel2, PhRel, PhRel2, and CapRel. Surprisingly, all these toxins specifically inhibit protein synthesis. To do so, they transfer a pyrophosphate moiety from ATP to the tRNA 3' CCA. The modification inhibits both tRNA aminoacylation and the sensing of cellular amino acid starvation by the ribosome-associated RSH RelA. Conversely, we show that some small alarmone hydrolase (SAH) RSH enzymes can reverse the pyrophosphorylation of tRNA to counter the growth inhibition by toxSAS. Collectively, we establish RSHs as RNA-modifying enzymes.


Asunto(s)
Toxinas Bacterianas/metabolismo , Guanosina Pentafosfato/metabolismo , Ligasas/metabolismo , ARN de Transferencia/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/farmacología , Bacilos Grampositivos Asporogénicos/química , Bacilos Grampositivos Asporogénicos/metabolismo , Guanosina Pentafosfato/química , Ligasas/química , Ligasas/genética , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/fisiología , Inhibidores de la Síntesis de la Proteína/farmacología , Pirofosfatasas , Ribosomas/metabolismo
5.
Nature ; 605(7909): 372-379, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35477761

RESUMEN

Post-transcriptional modifications have critical roles in tRNA stability and function1-4. In thermophiles, tRNAs are heavily modified to maintain their thermal stability under extreme growth temperatures5,6. Here we identified 2'-phosphouridine (Up) at position 47 of tRNAs from thermophilic archaea. Up47 confers thermal stability and nuclease resistance to tRNAs. Atomic structures of native archaeal tRNA showed a unique metastable core structure stabilized by Up47. The 2'-phosphate of Up47 protrudes from the tRNA core and prevents backbone rotation during thermal denaturation. In addition, we identified the arkI gene, which encodes an archaeal RNA kinase responsible for Up47 formation. Structural studies showed that ArkI has a non-canonical kinase motif surrounded by a positively charged patch for tRNA binding. A knockout strain of arkI grew slowly at high temperatures and exhibited a synthetic growth defect when a second tRNA-modifying enzyme was depleted. We also identified an archaeal homologue of KptA as an eraser that efficiently dephosphorylates Up47 in vitro and in vivo. Taken together, our findings show that Up47 is a reversible RNA modification mediated by ArkI and KptA that fine-tunes the structural rigidity of tRNAs under extreme environmental conditions.


Asunto(s)
Archaea , ARN de Transferencia , Termotolerancia , Archaea/genética , Ambientes Extremos , Fosforilación , Procesamiento Postranscripcional del ARN , ARN de Archaea/química , ARN de Archaea/metabolismo , ARN de Transferencia/química , ARN de Transferencia/metabolismo , Uridina
6.
Nucleic Acids Res ; 52(10): 5987-6001, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38485701

RESUMEN

Mycobacterium tuberculosis transfer RNA (tRNA) terminal nucleotidyltransferase toxin, MenT3, incorporates nucleotides at the 3'-CCA end of tRNAs, blocking their aminoacylation and inhibiting protein synthesis. Here, we show that MenT3 most effectively adds CMPs to the 3'-CCA end of tRNA. The crystal structure of MenT3 in complex with CTP reveals a CTP-specific nucleotide-binding pocket. The 4-NH2 and the N3 and O2 atoms of cytosine in CTP form hydrogen bonds with the main-chain carbonyl oxygen of P120 and the side chain of R238, respectively. MenT3 expression in Escherichia coli selectively reduces the levels of seryl-tRNASers, indicating specific inactivation of tRNASers by MenT3. Consistently, MenT3 incorporates CMPs into tRNASer most efficiently, among the tested E. coli tRNA species. The longer variable loop unique to class II tRNASers is crucial for efficient CMP incorporation into tRNASer by MenT3. Replacing the variable loop of E. coli tRNAAla with the longer variable loop of M. tuberculosis tRNASer enables MenT3 to incorporate CMPs into the chimeric tRNAAla. The N-terminal positively charged region of MenT3 is required for CMP incorporation into tRNASer. A docking model of tRNA onto MenT3 suggests that an interaction between the N-terminal region and the longer variable loop of tRNASer facilitates tRNA substrate selection.


Asunto(s)
Mycobacterium tuberculosis , ARN de Transferencia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/enzimología , Especificidad por Sustrato , ARN de Transferencia/metabolismo , ARN de Transferencia/química , Escherichia coli/genética , Escherichia coli/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Modelos Moleculares , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Citidina/química , Citidina/metabolismo , Sitios de Unión , Cristalografía por Rayos X , ARN Nucleotidiltransferasas/metabolismo , ARN Nucleotidiltransferasas/química , ARN Nucleotidiltransferasas/genética
7.
Nucleic Acids Res ; 52(7): 3938-3949, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38477328

RESUMEN

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.


Asunto(s)
Anticodón , Biosíntesis de Proteínas , ARN Catalítico , Aminoacil-ARN de Transferencia , ARN Catalítico/metabolismo , ARN Catalítico/genética , Anticodón/genética , Aminoacil-ARN de Transferencia/metabolismo , Aminoacil-ARN de Transferencia/genética , Fenilalanina/metabolismo , Fenilalanina/análogos & derivados , Aminoacil-ARNt Sintetasas/metabolismo , Aminoacil-ARNt Sintetasas/genética , Aminoacilación de ARN de Transferencia , Aminoacilación , Extensión de la Cadena Peptídica de Translación
8.
EMBO J ; 40(15): e107976, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34184765

RESUMEN

Nuclear stress bodies (nSBs) are nuclear membraneless organelles formed around stress-inducible HSATIII architectural long noncoding RNAs (lncRNAs). nSBs repress splicing of hundreds of introns during thermal stress recovery, which are partly regulated by CLK1 kinase phosphorylation of temperature-dependent Ser/Arg-rich splicing factors (SRSFs). Here, we report a distinct mechanism for this splicing repression through protein sequestration by nSBs. Comprehensive identification of RNA-binding proteins revealed HSATIII association with proteins related to N6 -methyladenosine (m6 A) RNA modification. 11% of the first adenosine in the repetitive HSATIII sequence were m6 A-modified. nSBs sequester the m6 A writer complex to methylate HSATIII, leading to subsequent sequestration of the nuclear m6 A reader, YTHDC1. Sequestration of these factors from the nucleoplasm represses m6 A modification of pre-mRNAs, leading to repression of m6 A-dependent splicing during stress recovery phase. Thus, nSBs serve as a common platform for regulation of temperature-dependent splicing through dual mechanisms employing two distinct ribonucleoprotein modules with partially m6 A-modified architectural lncRNAs.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Factores de Empalme de ARN/genética , Empalme del ARN , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Núcleo Celular/genética , Células HeLa , Humanos , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Factores de Empalme de ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Temperatura
9.
Nucleic Acids Res ; 50(8): 4713-4731, 2022 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-35411396

RESUMEN

Contact-dependent growth inhibition is a mechanism of interbacterial competition mediated by delivery of the C-terminal toxin domain of CdiA protein (CdiA-CT) into neighboring bacteria. The CdiA-CT of enterohemorrhagic Escherichia coli EC869 (CdiA-CTEC869) cleaves the 3'-acceptor regions of specific tRNAs in a reaction that requires the translation factors Tu/Ts and GTP. Here, we show that CdiA-CTEC869 has an intrinsic ability to recognize a specific sequence in substrate tRNAs, and Tu:Ts complex promotes tRNA cleavage by CdiA-CTEC869. Uncharged and aminoacylated tRNAs (aa-tRNAs) were cleaved by CdiA-CTEC869 to the same extent in the presence of Tu/Ts, and the CdiA-CTEC869:Tu:Ts:tRNA(aa-tRNA) complex formed in the presence of GTP. CdiA-CTEC869 interacts with domain II of Tu, thereby preventing the 3'-moiety of tRNA to bind to Tu as in canonical Tu:GTP:aa-tRNA complexes. Superimposition of the Tu:GTP:aa-tRNA structure onto the CdiA-CTEC869:Tu structure suggests that the 3'-portion of tRNA relocates into the CdiA-CTEC869 active site, located on the opposite side to the CdiA-CTEC869 :Tu interface, for tRNA cleavage. Thus, CdiA-CTEC869 is recruited to Tu:GTP:Ts, and CdiA-CT:Tu:GTP:Ts recognizes substrate tRNAs and cleaves them. Tu:GTP:Ts serves as a reaction scaffold that increases the affinity of CdiA-CTEC869 for substrate tRNAs and induces a structural change of tRNAs for efficient cleavage by CdiA-CTEC869.


Asunto(s)
Escherichia coli Enterohemorrágica , Proteínas de Escherichia coli , Escherichia coli Enterohemorrágica/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Inhibidores de Crecimiento , Guanosina Trifosfato/metabolismo , Proteínas de la Membrana/metabolismo , Factor Tu de Elongación Peptídica/metabolismo , ARN de Transferencia/metabolismo , Aminoacil-ARN de Transferencia
10.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34642250

RESUMEN

The tRNA pool determines the efficiency, throughput, and accuracy of translation. Previous studies have identified dynamic changes in the tRNA (transfer RNA) supply and mRNA (messenger RNA) demand during cancerous proliferation. Yet dynamic changes may also occur during physiologically normal proliferation, and these are less well characterized. We examined the tRNA and mRNA pools of T cells during their vigorous proliferation and differentiation upon triggering their antigen receptor. We observed a global signature of switch in demand for codons at the early proliferation phase of the response, accompanied by corresponding changes in tRNA expression levels. In the later phase, upon differentiation, the response of the tRNA pool relaxed back to the basal level, potentially restraining excessive proliferation. Sequencing of tRNAs allowed us to evaluate their diverse base-modifications. We found that two types of tRNA modifications, wybutosine and ms2t6A, are reduced dramatically during T cell activation. These modifications occur in the anticodon loops of two tRNAs that decode "slippery codons," which are prone to ribosomal frameshifting. Attenuation of these frameshift-protective modifications is expected to increase the potential for proteome-wide frameshifting during T cell proliferation. Indeed, human cell lines deleted of a wybutosine writer showed increased ribosomal frameshifting, as detected with an HIV gag-pol frameshifting site reporter. These results may explain HIV's specific tropism toward proliferating T cells since it requires ribosomal frameshift exactly on the corresponding codon for infection. The changes in tRNA expression and modifications uncover a layer of translation regulation during T cell proliferation and expose a potential tradeoff between cellular growth and translation fidelity.


Asunto(s)
Activación de Linfocitos , ARN de Transferencia/metabolismo , Linfocitos T/inmunología , Proliferación Celular/genética , Codón , Mutación del Sistema de Lectura , Humanos , Procesamiento Postranscripcional del ARN , Linfocitos T/citología
11.
Proc Natl Acad Sci U S A ; 117(34): 20785-20793, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32778592

RESUMEN

Transfer RNA (tRNA) activity is tightly regulated to provide a physiological protein translation, and tRNA chemical modifications control its function in a complex with ribosomes and messenger RNAs (mRNAs). In this regard, the correct hypermodification of position G37 of phenylalanine-tRNA, adjacent to the anticodon, is critical to prevent ribosome frameshifting events. Here we report that the tRNA-yW Synthesizing Protein 2 (TYW2) undergoes promoter hypermethylation-associated transcriptional silencing in human cancer, particularly in colorectal tumors. The epigenetic loss of TYW2 induces guanosine hypomodification in phenylalanine-tRNA, an increase in -1 ribosome frameshift events, and down-regulation of transcripts by mRNA decay, such as of the key cancer gene ROBO1. Importantly, TYW2 epigenetic inactivation is linked to poor overall survival in patients with early-stage colorectal cancer, a finding that could be related to the observed acquisition of enhanced migration properties and epithelial-to-mesenchymal features in the colon cancer cells that harbor TYW2 DNA methylation-associated loss. These findings provide an illustrative example of how epigenetic changes can modify the epitranscriptome and further support a role for tRNA modifications in cancer biology.


Asunto(s)
Neoplasias del Colon/genética , Sistema de Lectura Ribosómico , ARN de Transferencia/genética , Ribosomas/genética , ARNt Metiltransferasas/deficiencia , Adulto , Anciano , Anticodón/genética , Anticodón/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Islas de CpG , Epigénesis Genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Conformación de Ácido Nucleico , Fenilalanina/genética , Fenilalanina/metabolismo , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Ribosomas/metabolismo , ARNt Metiltransferasas/genética , ARNt Metiltransferasas/metabolismo
12.
Nucleic Acids Res ; 48(13): 7532-7544, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32501503

RESUMEN

Escherichia coli ItaT toxin reportedly acetylates the α-amino group of the aminoacyl-moiety of Ile-tRNAIle specifically, using acetyl-CoA as an acetyl donor, thereby inhibiting protein synthesis. The mechanism of the substrate specificity of ItaT had remained elusive. Here, we present functional and structural analyses of E. coli ItaT, which revealed the mechanism of ItaT recognition of specific aminoacyl-tRNAs for acetylation. In addition to Ile-tRNAIle, aminoacyl-tRNAs charged with hydrophobic residues, such as Val-tRNAVal and Met-tRNAMet, were acetylated by ItaT in vivo. Ile-tRNAIle, Val-tRNAVal and Met-tRNAMet were acetylated by ItaT in vitro, while aminoacyl-tRNAs charged with other hydrophobic residues, such as Ala-tRNAAla, Leu-tRNALeu and Phe-tRNAPhe, were less efficiently acetylated. A comparison of the structures of E. coli ItaT and the protein N-terminal acetyltransferase identified the hydrophobic residues in ItaT that possibly interact with the aminoacyl moiety of aminoacyl-tRNAs. Mutations of the hydrophobic residues of ItaT reduced the acetylation activity of ItaT toward Ile-tRNAIlein vitro, as well as the ItaT toxicity in vivo. Altogether, the size and shape of the hydrophobic pocket of ItaT are suitable for the accommodation of the specific aminoacyl-moieties of aminoacyl-tRNAs, and ItaT has broader specificity toward aminoacyl-tRNAs charged with certain hydrophobic amino acids.


Asunto(s)
Acetiltransferasas/química , Toxinas Bacterianas/química , Proteínas de Escherichia coli/química , Aminoacilación de ARN de Transferencia , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Secuencias de Aminoácidos , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Escherichia coli , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Mutación , Aminoacil-ARN de Transferencia/química , Aminoacil-ARN de Transferencia/metabolismo , Especificidad por Sustrato
13.
Nature ; 521(7553): 533-6, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25822791

RESUMEN

Small RNAs such as small interfering RNAs (siRNAs) and microRNAs (miRNAs) silence the expression of their complementary target messenger RNAs via the formation of effector RNA-induced silencing complexes (RISCs), which contain Argonaute (Ago) family proteins at their core. Although loading of siRNA duplexes into Drosophila Ago2 requires the Dicer-2-R2D2 heterodimer and the Hsc70/Hsp90 (Hsp90 also known as Hsp83) chaperone machinery, the details of RISC assembly remain unclear. Here we reconstitute RISC assembly using only Ago2, Dicer-2, R2D2, Hsc70, Hsp90, Hop, Droj2 (an Hsp40 homologue) and p23. By following the assembly of single RISC molecules, we find that, in the absence of the chaperone machinery, an siRNA bound to Dicer-2-R2D2 associates with Ago2 only transiently. The chaperone machinery extends the dwell time of the Dicer-2-R2D2-siRNA complex on Ago2, in a manner dependent on recognition of the 5'-phosphate on the siRNA guide strand. We propose that the chaperone machinery supports a productive state of Ago2, allowing it to load siRNA duplexes from Dicer-2-R2D2 and thereby assemble RISC.


Asunto(s)
Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Interferencia de ARN , Complejo Silenciador Inducido por ARN/química , Complejo Silenciador Inducido por ARN/metabolismo , Animales , Proteínas Argonautas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Choque Térmico/metabolismo , Técnicas In Vitro , Quinasas Janus/metabolismo , Unión Proteica , ARN Helicasas/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/metabolismo , Factores de Transcripción/metabolismo
14.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-34768885

RESUMEN

Transfer RNA[Ser]Sec carries multiple post-transcriptional modifications. The A37G mutation in tRNA[Ser]Sec abrogates isopentenylation of base 37 and has a profound effect on selenoprotein expression in mice. Patients with a homozygous pathogenic p.R323Q variant in tRNA-isopentenyl-transferase (TRIT1) show a severe neurological disorder, and hence we wondered whether selenoprotein expression was impaired. Patient fibroblasts with the homozygous p.R323Q variant did not show a general decrease in selenoprotein expression. However, recombinant human TRIT1R323Q had significantly diminished activities towards several tRNA substrates in vitro. We thus engineered mice conditionally deficient in Trit1 in hepatocytes and neurons. Mass-spectrometry revealed that hypermodification of U34 to mcm5Um occurs independently of isopentenylation of A37 in tRNA[Ser]Sec. Western blotting and 75Se metabolic labeling showed only moderate effects on selenoprotein levels and 75Se incorporation. A detailed analysis of Trit1-deficient liver using ribosomal profiling demonstrated that UGA/Sec re-coding was moderately affected in Selenop, Txnrd1, and Sephs2, but not in Gpx1. 2'O-methylation of U34 in tRNA[Ser]Sec depends on FTSJ1, but does not affect UGA/Sec re-coding in selenoprotein translation. Taken together, our results show that a lack of isopentenylation of tRNA[Ser]Sec affects UGA/Sec read-through but differs from a A37G mutation.


Asunto(s)
Transferasas Alquil y Aril/genética , ARN de Transferencia/metabolismo , Selenoproteínas/metabolismo , Transferasas Alquil y Aril/metabolismo , Animales , Línea Celular , Cisteína/metabolismo , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Ratones , Neuronas/metabolismo , Fosfotransferasas/genética , Fosfotransferasas/metabolismo , Biosíntesis de Proteínas/genética , ARN de Transferencia/genética , Ribosomas/metabolismo , Selenio/metabolismo , Selenocisteína/genética , Selenoproteína P/genética , Selenoproteínas/genética
15.
Nat Chem Biol ; 14(11): 1010-1020, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150682

RESUMEN

Modification of tRNA anticodons plays a critical role in ensuring accurate translation. N4-acetylcytidine (ac4C) is present at the anticodon first position (position 34) of bacterial elongator tRNAMet. Herein, we identified Bacillus subtilis ylbM (renamed tmcAL) as a novel gene responsible for ac4C34 formation. Unlike general acetyltransferases that use acetyl-CoA, TmcAL activates an acetate ion to form acetyladenylate and then catalyzes ac4C34 formation through a mechanism similar to tRNA aminoacylation. The crystal structure of TmcAL with an ATP analog reveals the molecular basis of ac4C34 formation. The ΔtmcAL strain displayed a cold-sensitive phenotype and a strong genetic interaction with tilS that encodes the enzyme responsible for synthesizing lysidine (L) at position 34 of tRNAIle to facilitate AUA decoding. Mistranslation of the AUA codon as Met in the ΔtmcAL strain upon tilS repression suggests that ac4C34 modification of tRNAMet and L34 modification of tRNAIle act cooperatively to prevent misdecoding of the AUA codon.


Asunto(s)
Acetatos/química , Bacillus subtilis/genética , ARN de Transferencia/química , Adenosina Trifosfato/química , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Catálisis , Dominio Catalítico , Codón de Terminación , Cristalografía por Rayos X , Análisis Mutacional de ADN , Escherichia coli/metabolismo , Mutación , Mycoplasma/metabolismo , Conformación de Ácido Nucleico , Fenotipo , Mutación Puntual , Biosíntesis de Proteínas , Dominios Proteicos , Proteínas Recombinantes/metabolismo
16.
Nucleic Acids Res ; 46(4): 1565-1583, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29390138

RESUMEN

Modified uridine containing taurine, 5-taurinomethyluridine (τm5U), is found at the anticodon first position of mitochondrial (mt-)transfer RNAs (tRNAs). Previously, we reported that τm5U is absent in mt-tRNAs with pathogenic mutations associated with mitochondrial diseases. However, biogenesis and physiological role of τm5U remained elusive. Here, we elucidated τm5U biogenesis by confirming that 5,10-methylene-tetrahydrofolate and taurine are metabolic substrates for τm5U formation catalyzed by MTO1 and GTPBP3. GTPBP3-knockout cells exhibited respiratory defects and reduced mitochondrial translation. Very little τm5U34 was detected in patient's cells with the GTPBP3 mutation, demonstrating that lack of τm5U results in pathological consequences. Taurine starvation resulted in downregulation of τm5U frequency in cultured cells and animal tissues (cat liver and flatfish). Strikingly, 5-carboxymethylaminomethyluridine (cmnm5U), in which the taurine moiety of τm5U is replaced with glycine, was detected in mt-tRNAs from taurine-depleted cells. These results indicate that tRNA modifications are dynamically regulated via sensing of intracellular metabolites under physiological condition.


Asunto(s)
ARN de Transferencia/metabolismo , Taurina/deficiencia , Uridina/análogos & derivados , Animales , Proteínas Portadoras/fisiología , Gatos , Preescolar , Femenino , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/fisiología , Células HEK293 , Células HeLa , Humanos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , ARN de Transferencia/química , Proteínas de Unión al ARN , Uridina/biosíntesis
17.
Proc Natl Acad Sci U S A ; 114(19): 4954-4959, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28439027

RESUMEN

Two-thiouridine (s2U) at position 54 of transfer RNA (tRNA) is a posttranscriptional modification that enables thermophilic bacteria to survive in high-temperature environments. s2U is produced by the combined action of two proteins, 2-thiouridine synthetase TtuA and 2-thiouridine synthesis sulfur carrier protein TtuB, which act as a sulfur (S) transfer enzyme and a ubiquitin-like S donor, respectively. Despite the accumulation of biochemical data in vivo, the enzymatic activity by TtuA/TtuB has rarely been observed in vitro, which has hindered examination of the molecular mechanism of S transfer. Here we demonstrate by spectroscopic, biochemical, and crystal structure analyses that TtuA requires oxygen-labile [4Fe-4S]-type iron (Fe)-S clusters for its enzymatic activity, which explains the previously observed inactivation of this enzyme in vitro. The [4Fe-4S] cluster was coordinated by three highly conserved cysteine residues, and one of the Fe atoms was exposed to the active site. Furthermore, the crystal structure of the TtuA-TtuB complex was determined at a resolution of 2.5 Å, which clearly shows the S transfer of TtuB to tRNA using its C-terminal thiocarboxylate group. The active site of TtuA is connected to the outside by two channels, one occupied by TtuB and the other used for tRNA binding. Based on these observations, we propose a molecular mechanism of S transfer by TtuA using the ubiquitin-like S donor and the [4Fe-4S] cluster.


Asunto(s)
Proteínas Bacterianas , Proteínas Hierro-Azufre , Ligasas , Thermus thermophilus , Tiouridina/análogos & derivados , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Catálisis , Cristalografía por Rayos X , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Ligasas/química , Ligasas/metabolismo , ARN Bacteriano/química , ARN Bacteriano/metabolismo , ARN de Transferencia/química , ARN de Transferencia/metabolismo , Thermus thermophilus/química , Thermus thermophilus/metabolismo , Tiouridina/química , Tiouridina/metabolismo
18.
Nucleic Acids Res ; 45(9): 5423-5436, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28119416

RESUMEN

Human RNA methyltransferase BCDIN3D is overexpressed in breast cancer cells, and is related to the tumorigenic phenotype and poor prognosis of breast cancer. Here, we show that cytoplasmic tRNAHis is the primary target of BCDIN3D in human cells. Recombinant human BCDIN3D, expressed in Escherichia coli, monomethylates the 5΄-monophosphate of cytoplasmic tRNAHis efficiently in vitro. In BCDN3D-knockout cells, established by CRISPR/Cas9 editing, the methyl moiety at the 5΄-monophosphate of cytoplasmic tRNAHis is lost, and the exogenous expression of BCDIN3D in the knockout cells restores the modification in cytoplasmic tRNAHis. BCIDN3D recognizes the 5΄-guanosine nucleoside at position -1 (G-1) and the eight-nucleotide acceptor helix with the G-1-A73 mis-pair at the top of the acceptor stem of cytoplasmic tRNAHis, which are exceptional structural features among cytoplasmic tRNA species. While the monomethylation of the 5΄-monophosphate of cytoplasmic tRNAHis affects neither the overall aminoacylation process in vitro nor the steady-state level of cytoplasmic tRNAHisin vivo, it protects the cytoplasmic tRNAHis transcript from degradation in vitro. Thus, BCDIN3D acts as a cytoplasmic tRNAHis-specific 5΄-methylphosphate capping enzyme. The present results also suggest the possible involvement of the monomethylation of the 5΄-monophosphate of cytoplasmic tRNAHis and/or cytoplasmic tRNAHis itself in the tumorigenesis of breast cancer cells.


Asunto(s)
Metiltransferasas/metabolismo , ARN de Transferencia de Histidina/metabolismo , Aminoacilación , Secuencia de Bases , Citoplasma/metabolismo , Células HEK293 , Humanos , Metilación , Conformación de Ácido Nucleico , Estabilidad del ARN , ARN de Transferencia de Histidina/química , ARN de Transferencia de Histidina/genética
19.
Nucleic Acids Res ; 45(4): 2124-2136, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-27913733

RESUMEN

Transfer RNA modifications play pivotal roles in protein synthesis. N6-threonylcarbamoyladenosine (t6A) and its derivatives are modifications found at position 37, 3΄-adjacent to the anticodon, in tRNAs responsible for ANN codons. These modifications are universally conserved in all domains of life. t6A and its derivatives have pleiotropic functions in protein synthesis including aminoacylation, decoding and translocation. We previously discovered a cyclic form of t6A (ct6A) as a chemically labile derivative of t6A in tRNAs from bacteria, fungi, plants and protists. Here, we report 2-methylthio cyclic t6A (ms2ct6A), a novel derivative of ct6A found in tRNAs from Bacillus subtilis, plants and Trypanosoma brucei. In B. subtilis and T. brucei, ms2ct6A disappeared and remained to be ms2t6A and ct6A by depletion of tcdA and mtaB homologs, respectively, demonstrating that TcdA and MtaB are responsible for biogenesis of ms2ct6A.


Asunto(s)
Adenosina/análogos & derivados , ARN de Transferencia/química , ARN de Transferencia/metabolismo , Treonina/análogos & derivados , Adenosina/química , Adenosina/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Secuencia de Bases , Conformación de Ácido Nucleico , Fenotipo , ARN de Planta/química , ARN de Planta/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Treonina/química , Treonina/metabolismo
20.
Mol Cell ; 39(2): 292-9, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20605501

RESUMEN

Small silencing RNAs--small interfering RNAs (siRNAs) or microRNAs (miRNAs)--direct posttranscriptional gene silencing of their mRNA targets as guides for the RNA-induced silencing complex (RISC). Both siRNAs and miRNAs are born double stranded. Surprisingly, loading these small RNA duplexes into Argonaute proteins, the core components of RISC, requires ATP, whereas separating the two small RNA strands within Argonaute does not. Here we show that the Hsc70/Hsp90 chaperone machinery is required to load small RNA duplexes into Argonaute proteins, but not for subsequent strand separation or target cleavage. We envision that the chaperone machinery uses ATP and mediates a conformational opening of Ago proteins so that they can receive bulky small RNA duplexes. Our data suggest that the chaperone machinery may serve as the driving force for the RISC assembly pathway.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , MicroARNs/metabolismo , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas del Choque Térmico HSC70/genética , Proteínas HSP90 de Choque Térmico/genética , MicroARNs/genética , ARN Bicatenario/genética , ARN Interferente Pequeño/genética , Complejo Silenciador Inducido por ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA