Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Bioessays ; 46(7): e2400058, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38724251

RESUMEN

The genetic code is a set of instructions that determine how the information in our genetic material is translated into amino acids. In general, it is universal for all organisms, from viruses and bacteria to humans. However, in the last few decades, exceptions to this rule have been identified both in pro- and eukaryotes. In this review, we discuss the 16 described alternative eukaryotic nuclear genetic codes and observe theories of their appearance in evolution. We consider possible molecular mechanisms that allow codon reassignment. Most reassignments in nuclear genetic codes are observed for stop codons. Moreover, in several organisms, stop codons can simultaneously encode amino acids and serve as termination signals. In this case, the meaning of the codon is determined by the additional factors besides the triplets. A comprehensive review of various non-standard coding events in the nuclear genomes provides a new insight into the translation mechanism in eukaryotes.


Asunto(s)
Código Genético , Biosíntesis de Proteínas , ARN Mensajero , Código Genético/genética , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Biosíntesis de Proteínas/genética , Animales , Codón de Terminación/genética , Núcleo Celular/genética , Evolución Molecular , Codón/genética , Eucariontes/genética
2.
Nucleic Acids Res ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38874498

RESUMEN

The poly(A) tail plays an important role in maintaining mRNA stability and influences translation efficiency via binding with PABP. However, the impact of poly(A) tail length on mRNA translation remains incompletely understood. This study explores the effects of poly(A) tail length on human translation. We determined the translation rates in cell lysates using mRNAs with different poly(A) tails. Cap-dependent translation was stimulated by the poly(A) tail, however, it was largely independent of poly(A) tail length, with an exception observed in the case of the 75 nt poly(A) tail. Conversely, cap-independent translation displayed a positive correlation with poly(A) tail length. Examination of translation stages uncovered the dependence of initiation and termination on the presence of the poly(A) tail, but the efficiency of initiation remained unaffected by poly(A) tail extension. Further study unveiled that increased binding of eRFs to the ribosome with the poly(A) tail extension induced more efficient hydrolysis of peptidyl-tRNA. Building upon these findings, we propose a crucial role for the 75 nt poly(A) tail in orchestrating the formation of a double closed-loop mRNA structure within human cells which couples the initiation and termination phases of translation.

3.
Int J Mol Sci ; 25(3)2024 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-38338898

RESUMEN

The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 system is a revolutionary tool for precise genome editing across various cell types. Ribonucleoproteins (RNPs), encompassing the Cas9 protein and guide RNA (gRNA), have emerged as a promising technique due to their increased specificity and reduced off-target effects. This method eliminates the need for plasmid DNA introduction, thereby preventing potential integration of foreign DNA into the target cell genome. Given the requirement for large quantities of highly purified protein in various Cas9 studies, we present an efficient and simple method for the preparation of recombinant Streptococcus pyogenes Cas9 (SpCas9) protein. This method leverages the Small Ubiquitin Like Modifier(SUMO) tag system, which includes metal-affinity chromatography followed by anion-exchange chromatography purification. Furthermore, we compare two methods of CRISPR-Cas9 system delivery into cells: transfection with plasmid DNA encoding the CRISPR-Cas9 system and RNP transfection with the Cas9-gRNA complex. We estimate the efficiency of genomic editing and protein lifespan post-transfection. Intriguingly, we found that RNP treatment of cells, even in the absence of a transfection system, is a relatively efficient method for RNP delivery into cell culture. This discovery is particularly promising as it can significantly reduce cytotoxicity, which is crucial for certain cell cultures such as induced pluripotent stem cells (iPSCs).


Asunto(s)
Sistemas CRISPR-Cas , ARN Guía de Sistemas CRISPR-Cas , Sistemas CRISPR-Cas/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Edición Génica/métodos , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , ADN
4.
J Biol Chem ; 298(7): 102133, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35700825

RESUMEN

The nucleotide context surrounding stop codons significantly affects the efficiency of translation termination. In eukaryotes, various 3' contexts that are unfavorable for translation termination have been described; however, the exact molecular mechanism that mediates their effects remains unknown. In this study, we used a reconstituted mammalian translation system to examine the efficiency of stop codons in different contexts, including several previously described weak 3' stop codon contexts. We developed an approach to estimate the level of stop codon readthrough in the absence of eukaryotic release factors (eRFs). In this system, the stop codon is recognized by the suppressor or near-cognate tRNAs. We observed that in the absence of eRFs, readthrough occurs in a 3' nucleotide context-dependent manner, and the main factors determining readthrough efficiency were the type of stop codon and the sequence of the 3' nucleotides. Moreover, the efficiency of translation termination in weak 3' contexts was almost equal to that in the tested standard context. Therefore, the ability of eRFs to recognize stop codons and induce peptide release is not affected by mRNA context. We propose that ribosomes or other participants of the elongation cycle can independently recognize certain contexts and increase the readthrough of stop codons. Thus, the efficiency of translation termination is regulated by the 3' nucleotide context following the stop codon and depends on the concentrations of eRFs and suppressor/near-cognate tRNAs.


Asunto(s)
Nucleótidos , Biosíntesis de Proteínas , Animales , Codón de Terminación/genética , Codón de Terminación/metabolismo , Eucariontes/metabolismo , Humanos , Mamíferos/metabolismo , Nucleótidos/genética , Nucleótidos/metabolismo , Extensión de la Cadena Peptídica de Translación , Terminación de la Cadena Péptídica Traduccional/genética , Factores de Terminación de Péptidos/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Ribosomas/genética , Ribosomas/metabolismo
5.
Nucleic Acids Res ; 49(19): 11181-11196, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34591963

RESUMEN

eIF3j is one of the eukaryotic translation factors originally reported as the labile subunit of the eukaryotic translation initiation factor eIF3. The yeast homolog of this protein, Hcr1, has been implicated in stringent AUG recognition as well as in controlling translation termination and stop codon readthrough. Using a reconstituted mammalian in vitro translation system, we showed that the human protein eIF3j is also important for translation termination. We showed that eIF3j stimulates peptidyl-tRNA hydrolysis induced by a complex of eukaryotic release factors, eRF1-eRF3. Moreover, in combination with the initiation factor eIF3, which also stimulates peptide release, eIF3j activity in translation termination increases. We found that eIF3j interacts with the pre-termination ribosomal complex, and eRF3 destabilises this interaction. In the solution, these proteins bind to each other and to other participants of translation termination, eRF1 and PABP, in the presence of GTP. Using a toe-printing assay, we determined the stage at which eIF3j functions - binding of release factors to the A-site of the ribosome before GTP hydrolysis. Based on these data, we assumed that human eIF3j is involved in the regulation of translation termination by loading release factors into the ribosome.


Asunto(s)
Factor 3 de Iniciación Eucariótica/genética , Terminación de la Cadena Péptídica Traduccional , Factores de Terminación de Péptidos/genética , Proteína I de Unión a Poli(A)/genética , Ribosomas/genética , Animales , Sistema Libre de Células , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Factor 3 de Iniciación Eucariótica/química , Factor 3 de Iniciación Eucariótica/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Hidrólisis , Modelos Moleculares , Factores de Terminación de Péptidos/metabolismo , Proteína I de Unión a Poli(A)/metabolismo , Unión Proteica , Conformación Proteica , Isoformas de Proteínas , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reticulocitos/química , Reticulocitos/metabolismo , Ribosomas/metabolismo , Transducción de Señal
6.
J Biol Chem ; 297(5): 101269, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34606825

RESUMEN

Programmed cell death 4 protein (PDCD4) regulates many vital cell processes, although is classified as a tumor suppressor because it inhibits neoplastic transformation and tumor growth. For example, PCDC4 has been implicated in the regulation of transcription and mRNA translation. PDCD4 is known to inhibit translation initiation by binding to eukaryotic initiation factor 4A and elongation of oncogenic c- and A-myb mRNAs. Additionally, PDCD4 has been shown to interact with poly(A)-binding protein (PABP), which affects translation termination, although the significance of this interaction is not fully understood. Considering the interaction between PABP and PDCD4, we hypothesized that PDCD4 may also be involved in translation termination. Using in vitro translation systems, we revealed that PDCD4 directly activates translation termination. PDCD4 stimulates peptidyl-tRNA hydrolysis induced by a complex of eukaryotic release factors, eRF1-eRF3. Moreover, in combination with the PABP, which also stimulates peptide release, PDCD4 activity in translation termination increases. PDCD4 regulates translation termination by facilitating the binding of release factors to the ribosome, increasing the GTPase activity of eRF3, and dissociating eRF3 from the posttermination complex. Using a toe-printing assay, we determined the first stage at which PDCD4 functions-binding of release factors to the A-site of the ribosome. However, preventing binding of eRF3 with PABP, PDCD4 suppresses subsequent rounds of translation termination. Based on these data, we assumed that human PDCD4 controls protein synthesis during translation termination. The described mechanism of the activity of PDCD4 in translation termination provides a new insight into its functioning during suppression of protein biosynthesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Terminación de la Cadena Péptídica Traduccional , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Sistema Libre de Células/metabolismo , Humanos , Factores de Terminación de Péptidos/metabolismo , Proteínas de Unión a Poli(A)/metabolismo
7.
Mol Cell ; 53(4): 645-54, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24486019

RESUMEN

Efficient stop codon recognition and peptidyl-tRNA hydrolysis are essential in order to terminate translational elongation and maintain protein sequence fidelity. Eukaryotic translational termination is mediated by a release factor complex that includes eukaryotic release factor 1 (eRF1) and eRF3. The N terminus of eRF1 contains highly conserved sequence motifs that couple stop codon recognition at the ribosomal A site to peptidyl-tRNA hydrolysis. We reveal that Jumonji domain-containing 4 (Jmjd4), a 2-oxoglutarate- and Fe(II)-dependent oxygenase, catalyzes carbon 4 (C4) lysyl hydroxylation of eRF1. This posttranslational modification takes place at an invariant lysine within the eRF1 NIKS motif and is required for optimal translational termination efficiency. These findings further highlight the role of 2-oxoglutarate/Fe(II) oxygenases in fundamental cellular processes and provide additional evidence that ensuring fidelity of protein translation is a major role of hydroxylation.


Asunto(s)
Regulación de la Expresión Génica , Histona Demetilasas/metabolismo , Oxigenasas de Función Mixta/química , Terminación de la Cadena Péptídica Traduccional/genética , Factores de Terminación de Péptidos/química , Biosíntesis de Proteínas , Secuencia de Aminoácidos , Animales , Catálisis , Línea Celular Tumoral , Codón de Terminación , Células HeLa , Humanos , Hidrólisis , Hidroxilación , Histona Demetilasas con Dominio de Jumonji , Modelos Moleculares , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
8.
RNA Biol ; 18(sup2): 804-817, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34793288

RESUMEN

Nsp1 of SARS-CoV-2 regulates the translation of host and viral mRNAs in cells. Nsp1 inhibits host translation initiation by occluding the entry channel of the 40S ribosome subunit. The structural study of the Nsp1-ribosomal complexes reported post-termination 80S complex containing Nsp1, eRF1 and ABCE1. Considering the presence of Nsp1 in the post-termination 80S ribosomal complex, we hypothesized that Nsp1 may be involved in translation termination. Using a cell-free translation system and reconstituted in vitro translation system, we show that Nsp1 stimulates peptide release and formation of termination complexes. Detailed analysis of Nsp1 activity during translation termination stages reveals that Nsp1 facilitates stop codon recognition. We demonstrate that Nsp1 stimulation targets eRF1 and does not affect eRF3. Moreover, Nsp1 increases amount of the termination complexes at all three stop codons. The activity of Nsp1 in translation termination is provided by its N-terminal domain and the minimal required part of eRF1 is NM domain. We assume that the biological meaning of Nsp1 activity in translation termination is binding with the 80S ribosomes translating host mRNAs and remove them from the pool of the active ribosomes.


Asunto(s)
Biosíntesis de Proteínas , SARS-CoV-2 , Proteínas no Estructurales Virales/fisiología , Animales , Sistema Libre de Células , Codón de Terminación/metabolismo , GTP Fosfohidrolasas/metabolismo , Células HeLa , Humanos , Mutación , Terminación de la Cadena Péptídica Traduccional , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/metabolismo , Péptidos/química , Unión Proteica , Conformación Proteica , Dominios Proteicos , ARN Mensajero/metabolismo , Conejos , Ribosomas/metabolismo
9.
Int J Mol Sci ; 22(10)2021 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-34069057

RESUMEN

Pairs of unnatural nucleotides are used to expand the genetic code and create artificial DNA or RNA templates. In general, an approach is used to engineer orthogonal systems capable of reading codons comprising artificial nucleotides; however, DNA and RNA polymerases capable of recognizing unnatural nucleotides are required for amplification and transcription of templates. Under favorable conditions, in the presence of modified nucleotide triphosphates, DNA polymerases are able to synthesize unnatural DNA with high efficiency; however, the currently available RNA polymerases reveal high specificity to the natural nucleotides and may not easily recognize the unnatural nucleotides. Due to the absence of simple and rapid methods for testing the activity of mutant RNA polymerases, the development of RNA polymerase recognizing unnatural nucleotides is limited. To fill this gap, we developed a method for rapid analysis of mutant RNA polymerase activity on templates containing unnatural nucleotides. Herein, we optimized a coupled cell-free translation system and tested the ability of three unnatural nucleotides to be transcribed by different T7 RNA polymerase mutants, by demonstrating high sensitivity and simplicity of the developed method. This approach can be applied to various unnatural nucleotides and can be simultaneously scaled up to determine the activity of numerous polymerases on different templates. Due to the simplicity and small amounts of material required, the developed cell-free system provides a highly scalable and versatile tool to study RNA polymerase activity.


Asunto(s)
Bacteriófago T7/enzimología , ARN Polimerasas Dirigidas por ADN/metabolismo , Mutación , Nucleótidos/análisis , Moldes Genéticos , Proteínas Virales/metabolismo , Sistema Libre de Células , ARN Polimerasas Dirigidas por ADN/genética , Transcripción Genética , Proteínas Virales/genética
10.
J Biol Chem ; 294(21): 8630-8639, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30992367

RESUMEN

Polyadenylate-binding protein (PABP) stimulates translation termination via interaction of its C-terminal domain with eukaryotic polypeptide chain release factor, eRF3. Additionally, two other proteins, poly(A)-binding protein-interacting proteins 1 and 2 (PAIP1 and PAIP2), bind the same domain of PABP and regulate its translation-related activity. To study the biochemistry of eRF3 and PAIP1/2 competition for PABP binding, we quantified the effects of PAIPs on translation termination in the presence or absence of PABP. Our results demonstrated that both PAIP1 and PAIP2 prevented translation termination at the premature termination codon, by controlling PABP activity. Moreover, PAIP1 and PAIP2 inhibited the activity of free PABP on translation termination in vitro However, after binding the poly(A) tail, PABP became insensitive to suppression by PAIPs and efficiently activated translation termination in the presence of eRF3a. Additionally, we revealed that PAIP1 binds eRF3 in solution, which stabilizes the post-termination complex. These results indicated that PAIP1 and PAIP2 participate in translation termination and are important regulators of readthrough at the premature termination codon.


Asunto(s)
Terminación de la Cadena Péptídica Traduccional , Factores de Iniciación de Péptidos/metabolismo , Factores de Terminación de Péptidos/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Humanos , Factores de Iniciación de Péptidos/química , Factores de Terminación de Péptidos/química , Poli A/química , Poli A/metabolismo , ARN Mensajero/química , Proteínas de Unión al ARN/química , Proteínas Represoras/química
11.
Methods ; 162-163: 54-59, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31201933

RESUMEN

Classical toeprinting is generally used to determine the position of ribosomes on mRNA; however, it has several disadvantages. We describe a fluorescent toeprinting assay that enables easier identification of ribosomal complexes bound to mRNA in vitro. The procedure involves the use of stable and safe fluorescently labeled oligonucleotides for reverse transcription reactions as primers, followed by the analysis of cDNA products using an automatic sequencer. This procedure allows the multiplexing and simultaneous analysis of a large number of samples. Over the past ten years, fluorescent toeprinting was applied to determine the activities of eukaryotic release factors and additional proteins involved in translation termination, to study the dynamics of translation initiation and elongation complexes, and to quantitatively evaluate the observed ribosomal complexes. Because of the simplicity and small amounts of material required, fluorescent toeprinting provides a highly scalable and versatile tool to study ribosomal complexes.


Asunto(s)
Bioensayo/métodos , Técnicas Genéticas , Ribosomas/metabolismo , Fluorescencia , Células HeLa , Humanos , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Biosíntesis de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Transcripción Reversa , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/aislamiento & purificación , Proteínas Ribosómicas/metabolismo
12.
Nucleic Acids Res ; 46(5): 2678-2689, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29401259

RESUMEN

Eukaryotic translation initiation is tightly regulated, requiring a set of conserved initiation factors (eIFs). Translation of a capped mRNA depends on the trimeric eIF4F complex and eIF4B to load the mRNA onto the 43S pre-initiation complex comprising 40S and initiation factors 1, 1A, 2, 3 and 5 as well as initiator-tRNA. Binding of the mRNA is followed by mRNA scanning in the 48S pre-initiation complex, until a start codon is recognised. Here, we use a reconstituted system to prepare human 48S complexes assembled on capped mRNA in the presence of eIF4B and eIF4F. The highly purified h-48S complexes are used for cross-linking/mass spectrometry, revealing the protein interaction network in this complex. We report the electron cryo-microscopy structure of the h-48S complex at 6.3 Å resolution. While the majority of eIF4B and eIF4F appear to be flexible with respect to the ribosome, additional density is detected at the entrance of the 40S mRNA channel which we attribute to the RNA-recognition motif of eIF4B. The eight core subunits of eIF3 are bound at the 40S solvent-exposed side, as well as the subunits eIF3d, eIF3b and eIF3i. elF2 and initiator-tRNA bound to the start codon are present at the 40S intersubunit side. This cryo-EM structure represents a molecular snap-shot revealing the h-48S complex following start codon recognition.


Asunto(s)
Factores Eucarióticos de Iniciación/química , Modelos Moleculares , Subunidades Ribosómicas Pequeñas de Eucariotas/química , Microscopía por Crioelectrón , Factor 3 de Iniciación Eucariótica/química , Humanos , Espectrometría de Masas , Iniciación de la Cadena Peptídica Traduccional , Caperuzas de ARN/química , ARN Mensajero/química , ARN Ribosómico 18S/química , Proteínas Ribosómicas/química
13.
J Biol Chem ; 293(14): 5220-5229, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29453282

RESUMEN

During protein synthesis, a ribosome moves along the mRNA template and, using aminoacyl-tRNAs, decodes the template nucleotide triplets to assemble a protein amino acid sequence. This movement is accompanied by shifting of mRNA-tRNA complexes within the ribosome in a process called translocation. In living cells, this process proceeds in a unidirectional manner, bringing the ribosome to the 3' end of mRNA, and is catalyzed by the GTPase translation elongation factor 2 (EF-G in prokaryotes and eEF2 in eukaryotes). Interestingly, the possibility of spontaneous backward translocation has been shown in vitro for bacterial ribosomes, suggesting a potential reversibility of this reaction. However, this possibility has not yet been tested for eukaryotic ribosomes. Here, using a reconstituted mammalian translation system, we show that the eukaryotic elongation factor eEF2 catalyzes ribosomal reverse translocation at one mRNA triplet. We found that this process requires a cognate tRNA in the ribosomal E-site and cannot occur spontaneously without eEF2. The efficiency of this reaction depended on the concentrations of eEF2 and cognate tRNAs and increased in the presence of nonhydrolyzable GTP analogues. Of note, ADP-ribosylation of eEF2 domain IV blocked reverse translocation, suggesting a crucial role of interactions of this domain with the ribosome for the catalysis of the reaction. In summary, our findings indicate that eEF2 is able to induce ribosomal translocation in forward and backward directions, highlighting the universal mechanism of tRNA-mRNA movements within the ribosome.


Asunto(s)
Extensión de la Cadena Peptídica de Translación/fisiología , Factor 2 de Elongación Peptídica/metabolismo , Ribosomas/metabolismo , Animales , Escherichia coli/metabolismo , Eucariontes/metabolismo , Células Eucariotas/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Modelos Moleculares , Factor 2 de Elongación Peptídica/fisiología , Factor G de Elongación Peptídica/metabolismo , Factores de Elongación de Péptidos/metabolismo , Unión Proteica , Biosíntesis de Proteínas/fisiología , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Aminoacil-ARN de Transferencia/metabolismo , Conejos , Proteínas Recombinantes
14.
Bioessays ; 39(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28009453

RESUMEN

The genetic code determines how amino acids are encoded within mRNA. It is universal among the vast majority of organisms, although several exceptions are known. Variant genetic codes are found in ciliates, mitochondria, and numerous other organisms. All revealed genetic codes (standard and variant) have at least one codon encoding a translation stop signal. However, recently two new genetic codes with a reassignment of all three stop codons were revealed in studies examining the protozoa transcriptomes. Here, we discuss this finding and the recent studies of variant genetic codes in eukaryotes. We consider the possible molecular mechanisms allowing the use of certain codons as sense and stop signals simultaneously. The results obtained by studying these amazing organisms represent a new and exciting insight into the mechanism of stop codon decoding in eukaryotes. Also see the video abstract here.


Asunto(s)
Codón de Terminación , Terminación de la Cadena Péptídica Traduccional , Regiones no Traducidas 3' , Animales , Eucariontes/genética , Evolución Molecular , Humanos , Factores de Terminación de Péptidos/fisiología
15.
Nucleic Acids Res ; 45(3): 1307-1318, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28180304

RESUMEN

The human DEAD-box RNA-helicase DDX19 functions in mRNA export through the nuclear pore complex. The yeast homolog of this protein, Dbp5, has been reported to participate in translation termination. Using a reconstituted mammalian in vitro translation system, we show that the human protein DDX19 is also important for translation termination. It is associated with the fraction of translating ribosomes. We show that DDX19 interacts with pre-termination complexes (preTCs) in a nucleotide-dependent manner. Furthermore, DDX19 increases the efficiency of termination complex (TC) formation and the peptide release in the presence of eukaryotic release factors. Using the eRF1(AGQ) mutant protein or a non-hydrolysable analog of GTP to inhibit subsequent peptidyl-tRNA hydrolysis, we reveal that the activation of translation termination by DDX19 occurs during the stop codon recognition. This activation is a result of DDX19 binding to preTC and a concomitant stabilization of terminating ribosomes. Moreover, we show that DDX19 stabilizes ribosome complexes with translation elongation factors eEF1 and eEF2. Taken together, our findings reveal that the human RNA helicase DDX19 actively participates in protein biosynthesis.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Extensión de la Cadena Peptídica de Translación/fisiología , Terminación de la Cadena Péptídica Traduccional/fisiología , Ribosomas/metabolismo , Codón de Terminación , ARN Helicasas DEAD-box/genética , Células HEK293 , Humanos , Mutación , Proteínas de Transporte Nucleocitoplasmático/genética , Factor 1 de Elongación Peptídica/metabolismo , Factor 2 de Elongación Peptídica/metabolismo , Polirribosomas/metabolismo , Aminoacil-ARN de Transferencia/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
16.
Nucleic Acids Res ; 44(16): 7766-76, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27418677

RESUMEN

Poly(A)-binding protein (PABP) is a major component of the messenger RNA-protein complex. PABP is able to bind the poly(A) tail of mRNA, as well as translation initiation factor 4G and eukaryotic release factor 3a (eRF3a). PABP has been found to stimulate translation initiation and to inhibit nonsense-mediated mRNA decay. Using a reconstituted mammalian in vitro translation system, we show that PABP directly stimulates translation termination. PABP increases the efficiency of translation termination by recruitment of eRF3a and eRF1 to the ribosome. PABP's function in translation termination depends on its C-terminal domain and its interaction with the N-terminus of eRF3a. Interestingly, we discover that full-length eRF3a exerts a different mode of function compared to its truncated form eRF3c, which lacks the N-terminal domain. Pre-association of eRF3a, but not of eRF3c, with pre-termination complexes (preTCs) significantly increases the efficiency of peptidyl-tRNA hydrolysis by eRF1. This implicates new, additional interactions of full-length eRF3a with the ribosomal preTC. Based on our findings, we suggest that PABP enhances the productive binding of the eRF1-eRF3 complex to the ribosome, via interactions with the N-terminal domain of eRF3a which itself has an active role in translation termination.


Asunto(s)
Codón de Terminación/metabolismo , Terminación de la Cadena Péptídica Traduccional/genética , Factores de Terminación de Péptidos/metabolismo , Proteínas de Unión a Poli(A)/metabolismo , Humanos , Hidrólisis , Modelos Biológicos , Unión Proteica , Aminoacil-ARN de Transferencia
17.
Nucleic Acids Res ; 43(5): 2874-87, 2015 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-25697503

RESUMEN

The human genome contains about 1.5 million Alu elements, which are transcribed into Alu RNAs by RNA polymerase III. Their expression is upregulated following stress and viral infection, and they associate with the SRP9/14 protein dimer in the cytoplasm forming Alu RNPs. Using cell-free translation, we have previously shown that Alu RNPs inhibit polysome formation. Here, we describe the mechanism of Alu RNP-mediated inhibition of translation initiation and demonstrate its effect on translation of cellular and viral RNAs. Both cap-dependent and IRES-mediated initiation is inhibited. Inhibition involves direct binding of SRP9/14 to 40S ribosomal subunits and requires Alu RNA as an assembly factor but its continuous association with 40S subunits is not required for inhibition. Binding of SRP9/14 to 40S prevents 48S complex formation by interfering with the recruitment of mRNA to 40S subunits. In cells, overexpression of Alu RNA decreases translation of reporter mRNAs and this effect is alleviated with a mutation that reduces its affinity for SRP9/14. Alu RNPs also inhibit the translation of cellular mRNAs resuming translation after stress and of viral mRNAs suggesting a role of Alu RNPs in adapting the translational output in response to stress and viral infection.


Asunto(s)
Elementos Alu/genética , ARN/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Ribosomas/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Electroforesis en Gel de Poliacrilamida , Células HEK293 , Humanos , Datos de Secuencia Molecular , Unión Proteica , ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/genética , Ribosomas/genética , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Partícula de Reconocimiento de Señal/genética , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/metabolismo
18.
Nucleic Acids Res ; 43(6): 3332-43, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25753665

RESUMEN

Stabilization of the ribosomal complexes plays an important role in translational control. Mechanisms of ribosome stabilization have been studied in detail for initiation and elongation of eukaryotic translation, but almost nothing is known about stabilization of eukaryotic termination ribosomal complexes. Here, we present one of the mechanisms of fine-tuning of the translation termination process in eukaryotes. We show that certain deacylated tRNAs, remaining in the E site of the ribosome at the end of the elongation cycle, increase the stability of the termination and posttermination complexes. Moreover, only the part of eRF1 recognizing the stop codon is stabilized in the A site of the ribosome, and the stabilization is not dependent on the hydrolysis of peptidyl-tRNA. The determinants, defining this property of the tRNA, reside in the acceptor stem. It was demonstrated by site-directed mutagenesis of tRNA(Val) and construction of a mini-helix structure identical to the acceptor stem of tRNA. The mechanism of this stabilization is different from the fixation of the unrotated state of the ribosome by CCA end of tRNA or by cycloheximide in the E site. Our data allow to reveal the possible functions of the isodecoder tRNAs in eukaryotes.


Asunto(s)
Terminación de la Cadena Péptídica Traduccional , ARN de Transferencia/metabolismo , Ribosomas/metabolismo , Acilación , Animales , Codón de Terminación , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/genética , Factores de Terminación de Péptidos/metabolismo , Estabilidad Proteica , Estabilidad del ARN , ARN de Transferencia/química , ARN de Transferencia/genética , ARN de Transferencia de Valina/química , ARN de Transferencia de Valina/genética , ARN de Transferencia de Valina/metabolismo , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Nucleic Acids Res ; 41(8): 4573-86, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23435318

RESUMEN

Release factor eRF1 plays a key role in the termination of protein synthesis in eukaryotes. The eRF1 consists of three domains (N, M and C) that perform unique roles in termination. Previous studies of eRF1 point mutants and standard/variant code eRF1 chimeras unequivocally demonstrated a direct involvement of the highly conserved N-domain motifs (NIKS, YxCxxxF and GTx) in stop codon recognition. In the current study, we extend this work by investigating the role of the 41 invariant and conserved N-domain residues in stop codon decoding by human eRF1. Using a combination of the conservative and non-conservative amino acid substitutions, we measured the functional activity of >80 mutant eRF1s in an in vitro reconstituted eukaryotic translation system and selected 15 amino acid residues essential for recognition of different stop codon nucleotides. Furthermore, toe-print analyses provide evidence of a conformational rearrangement of ribosomal complexes that occurs during binding of eRF1 to messenger RNA and reflects stop codon decoding activity of eRF1. Based on our experimental data and molecular modelling of the N-domain at the ribosomal A site, we propose a two-step model of stop codon decoding in the eukaryotic ribosome.


Asunto(s)
Codón de Terminación , Modelos Moleculares , Terminación de la Cadena Péptídica Traduccional , Factores de Terminación de Péptidos/química , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Mutación , Factores de Terminación de Péptidos/genética , Factores de Terminación de Péptidos/metabolismo , Péptidos/metabolismo , Estructura Terciaria de Proteína , Ribosomas/metabolismo , Alineación de Secuencia
20.
bioRxiv ; 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38260612

RESUMEN

Nonsense variants underlie many genetic diseases. The phenotypic impact of nonsense variants is determined by Nonsense-mediated mRNA decay (NMD), which degrades transcripts with premature termination codons (PTCs). NMD activity varies across transcripts and cellular contexts via poorly understood mechanisms. Here, by leveraging human genetic datasets, we uncover that the amino acid preceding the PTC dramatically affects NMD activity in human cells. We find that glycine codons in particular support high levels of NMD and are enriched before PTCs but depleted before normal termination codons (NTCs). Gly-PTC enrichment is most pronounced in human genes that tolerate loss-of-function variants. This suggests a strong biological impact for Gly-PTC in ensuring robust elimination of potentially toxic truncated proteins from non-essential genes. Biochemical assays revealed that the peptide release rate during translation termination is highly dependent on the identity of the amino acid preceding the stop codon. This release rate is the most critical feature determining NMD activity across our massively parallel reporter assays. Together, we conclude that NMD activity is significantly modulated by the "window of opportunity" offered by translation termination kinetics. Integrating the window of opportunity model with the existing framework of NMD would enable more accurate nonsense variant interpretation in the clinic.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA