Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Methods ; 229: 94-107, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38834165

ABSTRACT

In this report, non-isomerisable analogs of arginine tRNA (Arg-triazole-tRNA) have been synthesized as tools to study tRNA-dependent aminoacyl-transferases. The synthesis involves the incorporation of 1,4 substituted-1,2,3 triazole ring to mimic the ester bond that connects the amino acid to the terminal adenosine in the natural substrate. The synthetic procedure includes (i) a coupling between 2'- or 3'-azido-adenosine derivatives and a cytidine phosphoramidite to access dinucleotide molecules, (ii) Cu-catalyzed cycloaddition reactions between 2'- or 3'-azido dinucleotide in the presence of an alkyne molecule mimicking the arginine, providing the corresponding Arg-triazole-dinucleotides, (iii) enzymatic phosphorylation of the 5'-end extremity of the Arg-triazole-dinucleotides with a polynucleotide kinase, and (iv) enzymatic ligation of the 5'-phosphorylated dinucleotides with a 23-nt RNA micro helix that mimics the acceptor arm of arg-tRNA or with a full tRNAarg. Characterization of nucleoside and nucleotide compounds involved MS spectrometry, 1H, 13C and 31P NMR analysis. This strategy allows to obtain the pair of the two stable regioisomers of arg-tRNA analogs (2' and 3') which are instrumental to explore the regiospecificity of arginyl transferases enzyme. In our study, a first binding assay of the arg-tRNA micro helix with the Arginyl-tRNA-protein transferase 1 (ATE1) was performed by gel shift assays.


Subject(s)
Copper , Cycloaddition Reaction , Catalysis , Copper/chemistry , Cycloaddition Reaction/methods , Arginine/chemistry , Arginine/analogs & derivatives , RNA, Transfer, Arg/chemistry , RNA, Transfer, Arg/genetics , RNA, Transfer, Arg/metabolism , Phosphorylation , Triazoles/chemistry , Triazoles/chemical synthesis , Stereoisomerism , Adenosine/analogs & derivatives , Adenosine/chemistry , Aminoacyltransferases/metabolism , Aminoacyltransferases/chemistry , Aminoacyltransferases/genetics
2.
Nucleic Acids Res ; 50(15): 8529-8546, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35904807

ABSTRACT

Staphylococcus aureus, a human opportunist pathogen, adjusts its metabolism to cope with iron deprivation within the host. We investigated the potential role of small non-coding RNAs (sRNAs) in dictating this process. A single sRNA, named here IsrR, emerged from a competition assay with tagged-mutant libraries as being required during iron starvation. IsrR is iron-repressed and predicted to target mRNAs expressing iron-containing enzymes. Among them, we demonstrated that IsrR down-regulates the translation of mRNAs of enzymes that catalyze anaerobic nitrate respiration. The IsrR sequence reveals three single-stranded C-rich regions (CRRs). Mutational and structural analysis indicated a differential contribution of these CRRs according to targets. We also report that IsrR is required for full lethality of S. aureus in a mouse septicemia model, underscoring its role as a major contributor to the iron-sparing response for bacterial survival during infection. IsrR is conserved among staphylococci, but it is not ortholog to the proteobacterial sRNA RyhB, nor to other characterized sRNAs down-regulating mRNAs of iron-containing enzymes. Remarkably, these distinct sRNAs regulate common targets, illustrating that RNA-based regulation provides optimal evolutionary solutions to improve bacterial fitness when iron is scarce.


Subject(s)
RNA, Bacterial , RNA, Small Untranslated , Animals , Bacteria/genetics , Gene Expression Regulation, Bacterial , Humans , Iron/metabolism , Mice , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Staphylococcus/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
3.
Nucleic Acids Res ; 48(15): 8276-8289, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32735675

ABSTRACT

The manual production of reliable RNA structure models from chemical probing experiments benefits from the integration of information derived from multiple protocols and reagents. However, the interpretation of multiple probing profiles remains a complex task, hindering the quality and reproducibility of modeling efforts. We introduce IPANEMAP, the first automated method for the modeling of RNA structure from multiple probing reactivity profiles. Input profiles can result from experiments based on diverse protocols, reagents, or collection of variants, and are jointly analyzed to predict the dominant conformations of an RNA. IPANEMAP combines sampling, clustering and multi-optimization, to produce secondary structure models that are both stable and well-supported by experimental evidences. The analysis of multiple reactivity profiles, both publicly available and produced in our study, demonstrates the good performances of IPANEMAP, even in a mono probing setting. It confirms the potential of integrating multiple sources of probing data, informing the design of informative probing assays.


Subject(s)
Nucleic Acid Conformation , RNA/chemistry , Software , Amoebozoa/genetics , Benchmarking , Datasets as Topic , Mutation , RNA/genetics
4.
EMBO J ; 36(8): 981-994, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28167697

ABSTRACT

Long non-coding RNAs (lncRNAs) play diverse roles in physiological and pathological processes. Several lncRNAs have been suggested to modulate gene expression by guiding chromatin-modifying complexes to specific sites in the genome. However, besides the example of Xist, clear-cut evidence demonstrating this novel mode of regulation remains sparse. Here, we focus on HOTAIR, a lncRNA that is overexpressed in several tumor types and previously proposed to play a key role in gene silencing through direct recruitment of Polycomb Repressive Complex 2 (PRC2) to defined genomic loci. Using genetic tools and a novel RNA-tethering system, we investigated the interplay between HOTAIR and PRC2 in gene silencing. Surprisingly, we observed that forced overexpression of HOTAIR in breast cancer cells leads to subtle transcriptomic changes that appear to be independent of PRC2. Mechanistically, we found that artificial tethering of HOTAIR to chromatin causes transcriptional repression, but that this effect does not require PRC2. Instead, PRC2 recruitment appears to be a consequence of gene silencing. We propose that PRC2 binding to RNA might serve functions other than chromatin targeting.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Neoplasm Proteins/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , Transcription, Genetic , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Neoplasm Proteins/genetics , Polycomb Repressive Complex 2/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics
5.
Methods ; 162-163: 108-127, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31145972

ABSTRACT

Determination of the tridimensional structure of ribonucleic acid molecules is fundamental for understanding their function in the cell. A common method to investigate RNA structures of large molecules is the use of chemical probes such as SHAPE (2'-hydroxyl acylation analyzed by primer extension) reagents, DMS (dimethyl sulfate) and CMCT (1-cyclohexyl-3-(2-morpholinoethyl) carbodiimide metho-p-toluene sulfate), the reaction of which is dependent on the local structural properties of each nucleotide. In order to understand the interplay between local flexibility, sugar pucker, canonical pairing and chemical reactivity of the probes, we performed all-atom molecular dynamics simulations on a set of RNA molecules for which both tridimensional structure and chemical probing data are available and we analyzed the correlations between geometrical parameters and the chemical reactivity. Our study confirms that SHAPE reactivity is guided by the local flexibility of the different chemical moieties but suggests that a combination of multiple parameters is needed to better understand the implications of the reactivity at the molecular level. This is also the case for DMS and CMCT for which the reactivity appears to be more complex than commonly accepted.


Subject(s)
Molecular Dynamics Simulation , Nucleic Acid Conformation , Nucleotides/chemistry , RNA/chemistry , Acylation , CME-Carbodiimide/analogs & derivatives , CME-Carbodiimide/chemistry , Hydrogen Bonding , Hydroxyl Radical/chemistry , Indicators and Reagents/chemistry , RNA/genetics , RNA/metabolism , Sulfuric Acid Esters/chemistry
6.
Nucleic Acids Res ; 46(20): 11030-11047, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30215750

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is the etiological agent of adult T-cell leukemia (ATL). The HTLV-1 basic leucine zipper protein (HBZ) is expressed in all cases of ATL and is directly associated with virus pathogenicity. The two isoforms of the HBZ protein are synthesized from antisense messenger RNAs (mRNAs) that are either spliced (sHBZ) or unspliced (usHBZ) versions of the HBZ transcript. The sHBZ and usHBZ mRNAs have entirely different 5'untranslated regions (5'UTR) and are differentially expressed in cells, with the sHBZ protein being more abundant. Here, we show that differential expression of the HBZ isoforms is regulated at the translational level. Translation initiation of the usHBZ mRNA relies on a cap-dependent mechanism, while the sHBZ mRNA uses internal initiation. Based on the structural data for the sHBZ 5'UTR generated by SHAPE in combination with 5' and 3' deletion mutants, the minimal region harboring IRES activity was mapped to the 5'end of the sHBZ mRNA. In addition, the sHBZ IRES recruited the 40S ribosomal subunit upstream of the initiation codon, and IRES activity was found to be dependent on the ribosomal protein eS25 and eIF5A.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Human T-lymphotropic virus 1/genetics , Peptide Chain Initiation, Translational , RNA, Messenger/genetics , RNA, Viral/genetics , Retroviridae Proteins/genetics , 5' Untranslated Regions/genetics , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , COS Cells , Chlorocebus aethiops , Gene Expression Regulation, Viral , HEK293 Cells , HeLa Cells , Human T-lymphotropic virus 1/metabolism , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA, Viral/metabolism , Retroviridae Proteins/metabolism
7.
Nucleic Acids Res ; 45(12): 7382-7400, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28449096

ABSTRACT

In the late phase of the HIV virus cycle, the unspliced genomic RNA is exported to the cytoplasm for the necessary translation of the Gag and Gag-pol polyproteins. Three distinct translation initiation mechanisms ensuring Gag production have been described with little rationale for their multiplicity. The Gag-IRES has the singularity to be located within Gag ORF and to directly interact with ribosomal 40S. Aiming at elucidating the specificity and the relevance of this interaction, we probed HIV-1 Gag-IRES structure and developed an innovative integrative modelling strategy to take into account all the gathered information. We propose a novel Gag-IRES secondary structure strongly supported by all experimental data. We further demonstrate the presence of two regions within Gag-IRES that independently and directly interact with the ribosome. Importantly, these binding sites are functionally relevant to Gag translation both in vitro and ex vivo. This work provides insight into the Gag-IRES molecular mechanism and gives compelling evidence for its physiological importance. It allows us to propose original hypotheses about the IRES physiological role and conservation among primate lentiviruses.


Subject(s)
HIV-1/genetics , Internal Ribosome Entry Sites , Peptide Chain Initiation, Translational , Ribosome Subunits, Small, Eukaryotic/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , Genes, Reporter , HIV-1/metabolism , Humans , Jurkat Cells , Kinetics , Luciferases/genetics , Luciferases/metabolism , Models, Molecular , Nucleic Acid Conformation , Open Reading Frames , Ribosome Subunits, Small, Eukaryotic/ultrastructure , gag Gene Products, Human Immunodeficiency Virus/metabolism
8.
Nucleic Acids Res ; 45(22): 13016-13028, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29069411

ABSTRACT

Viral internal ribosomes entry site (IRES) elements coordinate the recruitment of the host translation machinery to direct the initiation of viral protein synthesis. Within hepatitis C virus (HCV)-like IRES elements, the sub-domain IIId(1) is crucial for recruiting the 40S ribosomal subunit. However, some HCV-like IRES elements possess an additional sub-domain, termed IIId2, whose function remains unclear. Herein, we show that IIId2 sub-domains from divergent viruses have different functions. The IIId2 sub-domain present in Seneca valley virus (SVV), a picornavirus, is dispensable for IRES activity, while the IIId2 sub-domains of two pestiviruses, classical swine fever virus (CSFV) and border disease virus (BDV), are required for 80S ribosomes assembly and IRES activity. Unlike in SVV, the deletion of IIId2 from the CSFV and BDV IRES elements impairs initiation of translation by inhibiting the assembly of 80S ribosomes. Consequently, this negatively affects the replication of CSFV and BDV. Finally, we show that the SVV IIId2 sub-domain is required for efficient viral RNA synthesis and growth of SVV, but not for IRES function. This study sheds light on the molecular evolution of viruses by clearly demonstrating that conserved RNA structures, within distantly related RNA viruses, have acquired different roles in the virus life cycles.


Subject(s)
Internal Ribosome Entry Sites/genetics , Pestivirus/genetics , Picornaviridae/genetics , RNA, Viral/genetics , Animals , Base Sequence , Binding Sites/genetics , Border disease virus/genetics , Border disease virus/physiology , Cell Line , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/physiology , HEK293 Cells , Host-Pathogen Interactions , Humans , Nucleic Acid Conformation , Pestivirus/physiology , Picornaviridae/physiology , RNA, Viral/chemistry , RNA, Viral/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Swine
9.
Nucleic Acids Res ; 44(3): 1309-25, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26626152

ABSTRACT

As obligatory intracellular parasites, viruses rely on cellular machines to complete their life cycle, and most importantly they recruit the host ribosomes to translate their mRNA. The Hepatitis C viral mRNA initiates translation by directly binding the 40S ribosomal subunit in such a way that the initiation codon is correctly positioned in the P site of the ribosome. Such a property is likely to be central for many viruses, therefore the description of host-pathogen interaction at the molecular level is instrumental to provide new therapeutic targets. In this study, we monitored the 40S ribosomal subunit and the viral RNA structural rearrangement induced upon the formation of the binary complex. We further took advantage of an IRES viral mutant mRNA deficient for translation to identify the interactions necessary to promote translation. Using a combination of structure probing in solution and molecular modeling we establish a whole atom model which appears to be very similar to the one obtained recently by cryoEM. Our model brings new information on the complex, and most importantly reveals some structural rearrangement within the ribosome. This study suggests that the formation of a 'kissing complex' between the viral RNA and the 18S ribosomal RNA locks the 40S ribosomal subunit in a conformation proficient for translation.


Subject(s)
Hepacivirus/genetics , Internal Ribosome Entry Sites/genetics , RNA, Viral/genetics , Ribosome Subunits, Small, Eukaryotic/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell-Free System , Codon, Initiator/genetics , Cryoelectron Microscopy , HeLa Cells , Hepacivirus/metabolism , Hepacivirus/physiology , Host-Pathogen Interactions , Humans , Macromolecular Substances/metabolism , Macromolecular Substances/ultrastructure , Models, Molecular , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Peptide Chain Initiation, Translational/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 18S/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism , Rabbits , Ribosome Subunits, Small, Eukaryotic/metabolism
10.
RNA ; 20(11): 1803-14, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25246653

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus, the etiological agent of Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL). One of the key viral proteins that contributes to tumorigenesis is vFLIP, a viral homolog of the FLICE inhibitory protein. This KSHV protein interacts with the NFκB pathway to trigger the expression of antiapoptotic and proinflammatory genes and ultimately leads to tumor formation. The expression of vFLIP is regulated at the translational level by an internal ribosomal entry site (IRES) element. However, the precise mechanism by which ribosomes are recruited internally and the exact location of the IRES has remained elusive. Here we show that a 252-nt fragment directly upstream of vFLIP, within a coding region, directs translation. We have established its RNA structure and demonstrate that IRES activity requires the presence of eIF4A and an intact eIF4G. Furthermore, and unusually for an IRES, eIF4E is part of the complex assembled onto the vFLIP IRES to direct translation. These molecular interactions define a new paradigm for IRES-mediated translation.


Subject(s)
Herpesvirus 8, Human/genetics , RNA, Viral/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Binding Sites , Cell Line, Tumor , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Models, Molecular , Nucleic Acid Conformation , RNA, Viral/genetics , Ribosomes/metabolism , Transcription, Genetic
11.
Nucleic Acids Res ; 42(16): 10373-84, 2014.
Article in English | MEDLINE | ID: mdl-25159618

ABSTRACT

Initiation of translation on Type II IRESs, such as those of EMCV and FMDV viruses, has been well documented in the recent years. For EMCV, the current model argues for a mechanism in which the key interaction necessary for the pre-initiation complex recruitment is eIF4G binding to the central J-K domains of EMCV-IRES. Here we demonstrate that, in contrast with the current model, the molecular mechanism of EMCV-IRES involves direct recruitment of the 40S subunit. Importantly, we identified a specific structural element that prevents the correct positioning of the initiation codon in the close vicinity of the ribosomal P site. This work clarifies how this interaction could not be anticipated by earlier studies and allows us to propose a new model for initiation complex assembly on EMCV-IRES. The role attributed to eIF4G/4A can thus be refined as stabilizing/promoting the conformational changes that are necessary for IRES function, thus resembling the role conventionally assigned to ITAFs. This raises the interesting possibility that IRESs are primarily ribosome binders, some of which having partly lost the ability to fold into the active structure without the help of proteins.


Subject(s)
5' Untranslated Regions , Encephalomyocarditis virus/genetics , Models, Genetic , Peptide Chain Initiation, Translational , Ribosome Subunits, Small, Eukaryotic/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Nucleic Acid Conformation , Open Reading Frames , RNA, Viral/chemistry , RNA, Viral/metabolism
13.
Proc Natl Acad Sci U S A ; 110(15): 5903-8, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23530232

ABSTRACT

The multiprotein exon junction complex (EJC), deposited by the splicing machinery, is an important constituent of messenger ribonucleoprotein particles because it participates to numerous steps of the mRNA lifecycle from splicing to surveillance via nonsense-mediated mRNA decay pathway. By an unknown mechanism, the EJC also stimulates translation efficiency of newly synthesized mRNAs. Here, we show that among the four EJC core components, the RNA-binding protein metastatic lymph node 51 (MLN51) is a translation enhancer. Overexpression of MLN51 preferentially increased the translation of intron-containing reporters via the EJC, whereas silencing MLN51 decreased translation. In addition, modulation of the MLN51 level in cell-free translational extracts confirmed its direct role in protein synthesis. Immunoprecipitations indicated that MLN51 associates with translation-initiating factors and ribosomal subunits, and in vitro binding assays revealed that MLN51, alone or as part of the EJC, interacts directly with the pivotal eukaryotic translation initiation factor eIF3. Taken together, our data define MLN51 as a translation activator linking the EJC and the translation machinery.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Biosynthesis , Biological Transport , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Introns , Protein Structure, Tertiary , RNA Splicing , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism
15.
Nucleic Acids Res ; 39(6): 2367-77, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21071421

ABSTRACT

Translation initiation on HIV genomic RNA relies on both cap and Internal Ribosome Entry Site (IRES) dependant mechanisms that are regulated throughout the cell cycle. During a unique phenomenon, the virus recruits initiation complexes through RNA structures located within Gag coding sequence, downstream of the initiation codon. We analyzed initiation complexes paused on the HIV-2 gag IRES and revealed that they contain all the canonical initiation factors except eIF4E and eIF1. We report that eIF3 and the small ribosomal subunit bind HIV RNA within gag open reading frame. We thus propose a novel two step model whereby the initial event is the formation of a ternary eIF3/40S/IRES complex. In a second step, dependent on most of the canonical initiation factors, the complex is rearranged to transfer the ribosome on the initiation codons. The absolute requirement of this large structure for HIV translation defines a new function for a coding region. Moreover, the level of information compaction within this viral genome reveals an additional level of evolutionary constraint on the coding sequence. The conservation of this IRES and its properties in rapidly evolving viruses suggest an important role in the virus life cycle and highlight an attractive new therapeutic target.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , HIV-2/genetics , Peptide Chain Initiation, Translational , RNA, Viral/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , 5' Untranslated Regions , Eukaryotic Initiation Factor-4F/metabolism , Nucleic Acid Conformation , Open Reading Frames , Protein Subunits/metabolism , RNA, Viral/metabolism
16.
Nucleic Acids Res ; 39(14): 6186-200, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21482538

ABSTRACT

The 5' leader of the human immunodeficiency virus type 1 (HIV-1) genomic RNA harbors an internal ribosome entry site (IRES) that is functional during the G2/M phase of the cell cycle. Here we show that translation initiation mediated by the HIV-1 IRES requires the participation of trans-acting cellular factors other than the canonical translational machinery. We used 'standard' chemical and enzymatic probes and an 'RNA SHAPE' analysis to model the structure of the HIV-1 5' leader and we show, by means of a footprinting assay, that G2/M extracts provide protections to regions previously identified as crucial for HIV-1 IRES activity. We also assessed the impact of mutations on IRES function. Strikingly, mutations did not significantly affect IRES activity suggesting that the requirement for pre-formed stable secondary or tertiary structure within the HIV-1 IRES may not be as strict as has been described for other viral IRESes. Finally, we used a proteomic approach to identify cellular proteins within the G2/M extracts that interact with the HIV-1 5' leader. Together, data show that HIV-1 IRES-mediated translation initiation is modulated by cellular proteins.


Subject(s)
5' Untranslated Regions , HIV-1/genetics , Peptide Chain Initiation, Translational , RNA, Viral/chemistry , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid , Base Sequence , Cell Cycle/genetics , Cytoplasm/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Point Mutation , RNA, Viral/metabolism
17.
Int J Mol Sci ; 14(3): 4400-18, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-23439549

ABSTRACT

Coxsackievirus B3 (CVB3) is an enterovirus of the family of Picornaviridae. The Group B coxsackieviruses include six serotypes (B1 to B6) that cause a variety of human diseases, including myocarditis, meningitis, and diabetes. Among the group B, the B3 strain is mostly studied for its cardiovirulence and its ability to cause acute and persistent infections. Translation initiation of CVB3 RNA has been shown to be mediated by a highly ordered structure of the 5'-untranslated region (5'UTR), which harbors an internal ribosome entry site (IRES). Translation initiation is a complex process in which initiator tRNA, 40S and 60S ribosomal subunits are assembled by eukaryotic initiation factors (eIFs) into an 80S ribosome at the initiation codon of the mRNA. We have previously addressed the question of whether the attenuating mutations of domain V of the poliovirus IRES were specific for a given genomic context or whether they could be transposed and extrapolated to a genomic related virus, i.e., CVB3 wild-type strain. In this context, we have described that Sabin3-like mutation (U473→C) introduced in CVB3 genome led to a defective mutant with a serious reduction in translation efficiency. In this study, we analyzed the efficiency of formation of ribosomal initiation complexes 48S and 80S through 10%-30% and 10%-50% sucrose gradients using rabbit reticulocyte lysates (RRLs) and stage-specific translation inhibitors: 5'-Guanylyl-imidodiphosphate (GMP-PNP) and Cycloheximide (CHX), respectively. We demonstrated that the interaction of 48S and 80S ribosomal complexes within the mutant CVB3 RNA was abolished compared with the wild-type RNA by ribosome assembly analysis. Taken together, it is possible that the mutant RNA was unable to interact with some trans-acting factors critical for enhanced IRES function.

18.
ACS Omega ; 8(4): 3850-3860, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36743074

ABSTRACT

Aminoacyl- and peptidyl-tRNA are specific biomolecules involved in many biological processes, from ribosomal protein synthesis to the synthesis of peptidoglycan precursors. Here, we report a post-synthetic approach based on traceless Staudinger ligation for the synthesis of a stable amide bond to access aminoacyl- or peptidyl-di-nucleotide. A series of amino-acid and peptide ester phenyl phosphines were synthetized, and their reactivity was studied on a 2'-N3 di-nucleotide. The corresponding 2'-amide di-nucleotides were obtained and characterized by LC-HRMS, and mechanistic interpretations of the influence of the amino acid phenyl ester phosphine were proposed. We also demonstrated that enzymatic 5'-OH phosphorylation is compatible with the acylated di-nucleotide, allowing the possibility to access stable aminoacylated-tRNA.

19.
Nucleic Acids Res ; 38(4): 1367-81, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19969542

ABSTRACT

Genomic RNA of primate lentiviruses serves both as an mRNA that encodes Gag and Gag-Pol polyproteins and as a propagated genome. Translation of this RNA is initiated by standard cap dependant mechanism or by internal entry of the ribosome. Two regions of the genomic RNA are able to attract initiation complexes, the 5' untranslated region and the gag coding region itself. Relying on probing data and a phylogenetic study, we have modelled the secondary structure of HIV-1, HIV-2 and SIV(Mac) coding region. This approach brings to light conserved secondary-structure elements that were shown by mutations to be required for internal entry of the ribosome. No structural homologies with other described viral or cellular IRES can be identified and lentiviral IRESes show many peculiar properties. Most notably, the IRES present in HIV-2 gag coding region is endowed with the unique ability to recruit up to three initiation complexes on a single RNA molecule. The structural and functional properties of gag coding sequence define a new type of IRES. Although its precise role is unknown, the conservation of the IRES among fast evolving lentiviruses suggests an important physiological role.


Subject(s)
HIV-2/genetics , Peptide Chain Initiation, Translational , RNA, Viral/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics , Ecthyma, Contagious , Eukaryotic Initiation Factors/metabolism , Genome, Viral , HIV-1/genetics , Mutagenesis, Site-Directed , Nucleic Acid Conformation , RNA, Viral/metabolism , Simian Immunodeficiency Virus/genetics
20.
Viruses ; 15(1)2022 12 20.
Article in English | MEDLINE | ID: mdl-36680049

ABSTRACT

Translation initiation of the hepatitis C virus (HCV) mRNA depends on an internal ribosome entry site (IRES) that encompasses most of the 5'UTR and includes nucleotides of the core coding region. This study shows that the polypyrimidine-tract-binding protein (PTB), an RNA-binding protein with four RNA recognition motifs (RRMs), binds to the HCV 5'UTR, stimulating its IRES activity. There are three isoforms of PTB: PTB1, PTB2, and PTB4. Our results show that PTB1 and PTB4, but not PTB2, stimulate HCV IRES activity in HuH-7 and HEK293T cells. In HuH-7 cells, PTB1 promotes HCV IRES-mediated initiation more strongly than PTB4. Mutations in PTB1, PTB4, RRM1/RRM2, or RRM3/RRM4, which disrupt the RRM's ability to bind RNA, abrogated the protein's capacity to stimulate HCV IRES activity in HuH-7 cells. In HEK293T cells, PTB1 and PTB4 stimulate HCV IRES activity to similar levels. In HEK293T cells, mutations in RRM1/RRM2 did not impact PTB1's ability to promote HCV IRES activity; and mutations in PTB1 RRM3/RRM4 domains reduced, but did not abolish, the protein's capacity to stimulate HCV IRES activity. In HEK293T cells, mutations in PTB4 RRM1/RRM2 abrogated the protein's ability to promote HCV IRES activity, and mutations in RRM3/RRM4 have no impact on PTB4 ability to enhance HCV IRES activity. Therefore, PTB1 and PTB4 differentially stimulate the IRES activity in a cell type-specific manner. We conclude that PTB1 and PTB4, but not PTB2, act as IRES transacting factors of the HCV IRES.


Subject(s)
Hepatitis C , Polypyrimidine Tract-Binding Protein , Humans , 5' Untranslated Regions , HEK293 Cells , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/genetics , Internal Ribosome Entry Sites , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/chemistry , Polypyrimidine Tract-Binding Protein/metabolism , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL