Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Cell ; 181(7): 1502-1517.e23, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32559462

ABSTRACT

RNA viruses are a major human health threat. The life cycles of many highly pathogenic RNA viruses like influenza A virus (IAV) and Lassa virus depends on host mRNA, because viral polymerases cleave 5'-m7G-capped host transcripts to prime viral mRNA synthesis ("cap-snatching"). We hypothesized that start codons within cap-snatched host transcripts could generate chimeric human-viral mRNAs with coding potential. We report the existence of this mechanism of gene origination, which we named "start-snatching." Depending on the reading frame, start-snatching allows the translation of host and viral "untranslated regions" (UTRs) to create N-terminally extended viral proteins or entirely novel polypeptides by genetic overprinting. We show that both types of chimeric proteins are made in IAV-infected cells, generate T cell responses, and contribute to virulence. Our results indicate that during infection with IAV, and likely a multitude of other human, animal and plant viruses, a host-dependent mechanism allows the genesis of hybrid genes.


Subject(s)
RNA Caps/genetics , RNA Virus Infections/genetics , Recombinant Fusion Proteins/genetics , 5' Untranslated Regions/genetics , Animals , Cattle , Cell Line , Cricetinae , Dogs , Humans , Influenza A virus/metabolism , Mice , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Open Reading Frames/genetics , RNA Caps/metabolism , RNA Virus Infections/metabolism , RNA Viruses/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Fusion Proteins/metabolism , Transcription, Genetic/genetics , Viral Proteins/metabolism , Virus Replication/genetics
2.
Mol Cell ; 74(3): 481-493.e6, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30904393

ABSTRACT

The use of alternative translation initiation sites enables production of more than one protein from a single gene, thereby expanding the cellular proteome. Although several such examples have been serendipitously found in bacteria, genome-wide mapping of alternative translation start sites has been unattainable. We found that the antibiotic retapamulin specifically arrests initiating ribosomes at start codons of the genes. Retapamulin-enhanced Ribo-seq analysis (Ribo-RET) not only allowed mapping of conventional initiation sites at the beginning of the genes, but strikingly, it also revealed putative internal start sites in a number of Escherichia coli genes. Experiments demonstrated that the internal start codons can be recognized by the ribosomes and direct translation initiation in vitro and in vivo. Proteins, whose synthesis is initiated at internal in-frame and out-of-frame start sites, can be functionally important and contribute to the "alternative" bacterial proteome. The internal start sites may also play regulatory roles in gene expression.


Subject(s)
Genome, Bacterial/genetics , Peptide Chain Initiation, Translational , Proteome/genetics , Proteomics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Codon, Initiator/genetics , Diterpenes/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/drug effects , Genome, Bacterial/drug effects , RNA, Messenger/genetics , Ribosomes/drug effects , Ribosomes/genetics
3.
PLoS Pathog ; 20(5): e1012034, 2024 May.
Article in English | MEDLINE | ID: mdl-38814986

ABSTRACT

Ilarviruses are a relatively understudied but important group of plant RNA viruses that includes a number of crop pathogens. Their genomes comprise three RNA segments encoding two replicase subunits, movement protein, coat protein (CP), and (in some ilarvirus subgroups) a protein that suppresses RNA silencing. Here we report that, in many ilarviruses, RNA3 encodes an additional protein (termed CP-RT) as a result of ribosomal readthrough of the CP stop codon into a short downstream readthrough (RT) ORF. Using asparagus virus 2 as a model, we find that CP-RT is expressed in planta where it functions as a weak suppressor of RNA silencing. CP-RT expression is essential for persistent systemic infection in leaves and shoot apical meristem. CP-RT function is dependent on a putative zinc-finger motif within RT. Replacing the asparagus virus 2 RT with the RT of an ilarvirus from a different subgroup restored the ability to establish persistent infection. These findings open up a new avenue for research on ilarvirus silencing suppression, persistent meristem invasion and vertical transmission.


Subject(s)
Ilarvirus , Plant Diseases , RNA Interference , Capsid Proteins/genetics , Capsid Proteins/metabolism , Codon, Terminator/genetics , Ilarvirus/genetics , Nicotiana/virology , Nicotiana/genetics , Nicotiana/metabolism , Plant Diseases/virology , Plant Diseases/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
4.
EMBO Rep ; 24(12): e57224, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37818801

ABSTRACT

The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Coronaviridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrate that SARS-CoV-2 infection causes tetherin downregulation and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigate the potential viral proteins involved in abrogating tetherin function and find that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles where Coronaviruses bud, and increases tetherin localisation to late endocytic organelles via reduced retrograde recycling. We also find that expression of Spike protein causes a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.


Subject(s)
Bone Marrow Stromal Antigen 2 , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Release , Humans , Bone Marrow Stromal Antigen 2/antagonists & inhibitors , Bone Marrow Stromal Antigen 2/metabolism , COVID-19/virology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/genetics
5.
Nucleic Acids Res ; 51(2): 891-907, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36629253

ABSTRACT

The synthesis of mitochondrial OXPHOS complexes is central to cellular metabolism, yet many molecular details of mitochondrial translation remain elusive. It has been commonly held view that translation initiation in human mitochondria proceeded in a manner similar to bacterial systems, with the mitoribosomal small subunit bound to the initiation factors, mtIF2 and mtIF3, along with initiator tRNA and an mRNA. However, unlike in bacteria, most human mitochondrial mRNAs lack 5' leader sequences that can mediate small subunit binding, raising the question of how leaderless mRNAs are recognized by mitoribosomes. By using novel in vitro mitochondrial translation initiation assays, alongside biochemical and genetic characterization of cellular knockouts of mitochondrial translation factors, we describe unique features of translation initiation in human mitochondria. We show that in vitro, leaderless mRNA transcripts can be loaded directly onto assembled 55S mitoribosomes, but not onto the mitoribosomal small subunit (28S), in a manner that requires initiator fMet-tRNAMet binding. In addition, we demonstrate that in human cells and in vitro, mtIF3 activity is not required for translation of leaderless mitochondrial transcripts but is essential for translation of ATP6 in the case of the bicistronic ATP8/ATP6 transcript. Furthermore, we show that mtIF2 is indispensable for mitochondrial protein synthesis. Our results demonstrate an important evolutionary divergence of the mitochondrial translation system and further our fundamental understanding of a process central to eukaryotic metabolism.


Subject(s)
Mitochondria , Peptide Chain Initiation, Translational , Animals , Humans , Bacteria/genetics , Mammals/genetics , Mitochondria/physiology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Peptide Initiation Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35217614

ABSTRACT

Translation start site selection in eukaryotes is influenced by context nucleotides flanking the AUG codon and by levels of the eukaryotic translation initiation factors eIF1 and eIF5. In a search of mammalian genes, we identified five homeobox (Hox) gene paralogs initiated by AUG codons in conserved suboptimal context as well as 13 Hox genes that contain evolutionarily conserved upstream open reading frames (uORFs) that initiate at AUG codons in poor sequence context. An analysis of published cap analysis of gene expression sequencing (CAGE-seq) data and generated CAGE-seq data for messenger RNAs (mRNAs) from mouse somites revealed that the 5' leaders of Hox mRNAs of interest contain conserved uORFs, are generally much shorter than reported, and lack previously proposed internal ribosome entry site elements. We show that the conserved uORFs inhibit Hox reporter expression and that altering the stringency of start codon selection by overexpressing eIF1 or eIF5 modulates the expression of Hox reporters. We also show that modifying ribosome homeostasis by depleting a large ribosomal subunit protein or treating cells with sublethal concentrations of puromycin leads to lower stringency of start codon selection. Thus, altering global translation can confer gene-specific effects through altered start codon selection stringency.


Subject(s)
Codon, Initiator , Evolution, Molecular , Genes, Homeobox , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Mice , Open Reading Frames
7.
Mol Biol Evol ; 40(4)2023 04 04.
Article in English | MEDLINE | ID: mdl-37014783

ABSTRACT

RNA viruses are abundant and highly diverse and infect all or most eukaryotic organisms. However, only a tiny fraction of the number and diversity of RNA virus species have been catalogued. To cost-effectively expand the diversity of known RNA virus sequences, we mined publicly available transcriptomic data sets. We developed 77 family-level Hidden Markov Model profiles for the viral RNA-dependent RNA polymerase (RdRp)-the only universal "hallmark" gene of RNA viruses. By using these to search the National Center for Biotechnology Information Transcriptome Shotgun Assembly database, we identified 5,867 contigs encoding RNA virus RdRps or fragments thereof and analyzed their diversity, taxonomic classification, phylogeny, and host associations. Our study expands the known diversity of RNA viruses, and the 77 curated RdRp Profile Hidden Markov Models provide a useful resource for the virus discovery community.


Subject(s)
RNA Viruses , Transcriptome , RNA Viruses/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Phylogeny , RNA, Viral , Genome, Viral
9.
PLoS Pathog ; 17(3): e1009403, 2021 03.
Article in English | MEDLINE | ID: mdl-33735221

ABSTRACT

Arteriviruses are enveloped positive-strand RNA viruses that assemble and egress using the host cell's exocytic pathway. In previous studies, we demonstrated that most arteriviruses use a unique -2 ribosomal frameshifting mechanism to produce a C-terminally modified variant of their nonstructural protein 2 (nsp2). Like full-length nsp2, the N-terminal domain of this frameshift product, nsp2TF, contains a papain-like protease (PLP2) that has deubiquitinating (DUB) activity, in addition to its role in proteolytic processing of replicase polyproteins. In cells infected with porcine reproductive and respiratory syndrome virus (PRRSV), nsp2TF localizes to compartments of the exocytic pathway, specifically endoplasmic reticulum-Golgi intermediate compartment (ERGIC) and Golgi complex. Here, we show that nsp2TF interacts with the two major viral envelope proteins, the GP5 glycoprotein and membrane (M) protein, which drive the key process of arterivirus assembly and budding. The PRRSV GP5 and M proteins were found to be poly-ubiquitinated, both in an expression system and in cells infected with an nsp2TF-deficient mutant virus. In contrast, ubiquitinated GP5 and M proteins did not accumulate in cells infected with the wild-type, nsp2TF-expressing virus. Further analysis implicated the DUB activity of the nsp2TF PLP2 domain in deconjugation of ubiquitin from GP5/M proteins, thus antagonizing proteasomal degradation of these key viral structural proteins. Our findings suggest that nsp2TF is targeted to the exocytic pathway to reduce proteasome-driven turnover of GP5/M proteins, thus promoting the formation of GP5-M dimers that are critical for arterivirus assembly.


Subject(s)
Deubiquitinating Enzymes/metabolism , Gene Expression Regulation, Viral/physiology , Porcine respiratory and reproductive syndrome virus/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Humans , Porcine Reproductive and Respiratory Syndrome/virology , Swine , Virus Assembly/physiology , Virus Replication/physiology
10.
PLoS Pathog ; 17(6): e1009644, 2021 06.
Article in English | MEDLINE | ID: mdl-34138976

ABSTRACT

Coronavirus infection induces the unfolded protein response (UPR), a cellular signalling pathway composed of three branches, triggered by unfolded proteins in the endoplasmic reticulum (ER) due to high ER load. We have used RNA sequencing and ribosome profiling to investigate holistically the transcriptional and translational response to cellular infection by murine hepatitis virus (MHV), often used as a model for the Betacoronavirus genus to which the recently emerged SARS-CoV-2 also belongs. We found the UPR to be amongst the most significantly up-regulated pathways in response to MHV infection. To confirm and extend these observations, we show experimentally the induction of all three branches of the UPR in both MHV- and SARS-CoV-2-infected cells. Over-expression of the SARS-CoV-2 ORF8 or S proteins alone is itself sufficient to induce the UPR. Remarkably, pharmacological inhibition of the UPR greatly reduced the replication of both MHV and SARS-CoV-2, revealing the importance of this pathway for successful coronavirus replication. This was particularly striking when both IRE1α and ATF6 branches of the UPR were inhibited, reducing SARS-CoV-2 virion release (~1,000-fold). Together, these data highlight the UPR as a promising antiviral target to combat coronavirus infection.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Murine hepatitis virus/drug effects , Unfolded Protein Response/drug effects , Activating Transcription Factor 6/metabolism , Animals , Antiviral Agents/therapeutic use , Cell Line , Chlorocebus aethiops , Drug Delivery Systems , Endoribonucleases/metabolism , HEK293 Cells , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , RNA-Seq , Vero Cells , Viral Proteins/metabolism , Virus Replication/drug effects
11.
PLoS Pathog ; 17(8): e1009824, 2021 08.
Article in English | MEDLINE | ID: mdl-34398933

ABSTRACT

The herpes simplex virus (HSV)-1 protein pUL21 is essential for efficient virus replication and dissemination. While pUL21 has been shown to promote multiple steps of virus assembly and spread, the molecular basis of its function remained unclear. Here we identify that pUL21 is a virus-encoded adaptor of protein phosphatase 1 (PP1). pUL21 directs the dephosphorylation of cellular and virus proteins, including components of the viral nuclear egress complex, and we define a conserved non-canonical linear motif in pUL21 that is essential for PP1 recruitment. In vitro evolution experiments reveal that pUL21 antagonises the activity of the virus-encoded kinase pUS3, with growth and spread of pUL21 PP1-binding mutant viruses being restored in adapted strains where pUS3 activity is disrupted. This study shows that virus-directed phosphatase activity is essential for efficient herpesvirus assembly and spread, highlighting the fine balance between kinase and phosphatase activity required for optimal virus replication.


Subject(s)
Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Phosphoric Monoester Hydrolases/metabolism , Viral Proteins/metabolism , Virus Assembly , Virus Replication , Animals , Chlorocebus aethiops , HEK293 Cells , Herpesvirus 1, Human/enzymology , Humans , Phosphoric Monoester Hydrolases/genetics , Vero Cells , Viral Proteins/genetics , Virus Release
13.
Nucleic Acids Res ; 49(20): 11938-11958, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34751406

ABSTRACT

The 2A protein of Theiler's murine encephalomyelitis virus (TMEV) acts as a switch to stimulate programmed -1 ribosomal frameshifting (PRF) during infection. Here, we present the X-ray crystal structure of TMEV 2A and define how it recognises the stimulatory RNA element. We demonstrate a critical role for bases upstream of the originally predicted stem-loop, providing evidence for a pseudoknot-like conformation and suggesting that the recognition of this pseudoknot by beta-shell proteins is a conserved feature in cardioviruses. Through examination of PRF in TMEV-infected cells by ribosome profiling, we identify a series of ribosomal pauses around the site of PRF induced by the 2A-pseudoknot complex. Careful normalisation of ribosomal profiling data with a 2A knockout virus facilitated the identification, through disome analysis, of ribosome stacking at the TMEV frameshifting signal. These experiments provide unparalleled detail of the molecular mechanisms underpinning Theilovirus protein-stimulated frameshifting.


Subject(s)
Frameshifting, Ribosomal , Viral Proteins/metabolism , Ribosomes/metabolism , Theilovirus/genetics , Theilovirus/metabolism , Viral Proteins/chemistry
14.
Mol Plant Microbe Interact ; 35(9): 835-844, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35671468

ABSTRACT

Potyviruses comprise the largest and most important group of plant positive-strand RNA viruses. The potyviral cell-to-cell movement protein P3N-PIPO is expressed via transcriptional slippage at a conserved GAAAAAA sequence, leading to insertion of an extra 'A' in a proportion of viral transcripts. Transcriptional slippage is determined by the potyviral replicase, the conserved slippery site, and its flanking nucleotides. Here, we investigate the dynamics of transcriptional slippage at different slip-site sequences, infection stages, and environmental conditions. We detect a modest increase in the level of transcripts with insertion towards later timepoints. In addition, we investigate the fate of transcripts with insertion by separately looking at different RNA subpopulations: (+)RNA, (-)RNA, translated RNA, and virion RNA. We find differences in insertional slippage between (+)RNA and (-)RNA but not other subpopulations. Our results suggest that there can be selection against the use of (-)RNAs with insertions as templates for transcription or replication and demonstrate that insertional slippage can occur at high frequency also during (-)RNA synthesis. Since transcripts with insertions are potential targets for degradation, we investigate the connection to nonsense-mediated decay (NMD). We find that these transcripts are targeted to NMD, but we only observe an impact on the level of transcripts with insertion when the insertional slippage rate is high. Together, these results further our understanding of the mechanism and elucidate the dynamics of potyviral transcriptional slippage. [Formula: see text] Copyright © 2022 The Author(s). This is an open access article distributed under the CC BY 4.0 International license.


Subject(s)
Potyvirus , Viral Proteins , Nucleotides/metabolism , Potyvirus/genetics , Potyvirus/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Nicotiana/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
15.
J Virol ; 95(14): e0066321, 2021 06 24.
Article in English | MEDLINE | ID: mdl-33963053

ABSTRACT

RNA structural elements occur in numerous single-stranded positive-sense RNA viruses. The stem-loop 2 motif (s2m) is one such element with an unusually high degree of sequence conservation, being found in the 3' untranslated region (UTR) in the genomes of many astroviruses, some picornaviruses and noroviruses, and a variety of coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. The evolutionary conservation and its occurrence in all viral subgenomic transcripts imply a key role for s2m in the viral infection cycle. Our findings indicate that the element, while stably folded, can nonetheless be invaded and remodeled spontaneously by antisense oligonucleotides (ASOs) that initiate pairing in exposed loops and trigger efficient sequence-specific RNA cleavage in reporter assays. ASOs also act to inhibit replication in an astrovirus replicon model system in a sequence-specific, dose-dependent manner and inhibit SARS-CoV-2 replication in cell culture. Our results thus permit us to suggest that the s2m element is readily targeted by ASOs, which show promise as antiviral agents. IMPORTANCE The highly conserved stem-loop 2 motif (s2m) is found in the genomes of many RNA viruses, including SARS-CoV-2. Our findings indicate that the s2m element can be targeted by antisense oligonucleotides. The antiviral potential of this element represents a promising start for further research into targeting conserved elements in RNA viruses.


Subject(s)
COVID-19 , Genome, Viral , Nucleotide Motifs , RNA Folding , RNA, Viral , SARS-CoV-2/physiology , Virus Replication , Animals , COVID-19/genetics , COVID-19/metabolism , Chlorocebus aethiops , HEK293 Cells , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , Vero Cells
16.
J Gen Virol ; 102(11)2021 11.
Article in English | MEDLINE | ID: mdl-34738886

ABSTRACT

Nyamiviridae is a family of viruses in the order Mononegavirales, with unsegmented (except for members of the genus Tapwovirus), negative-sense RNA genomes of 10-13 kb. Nyamviruses have a genome organisation and content similar to that of other mononegaviruses. Nyamiviridae includes several genera that form monophyletic clades on phylogenetic analysis of the RNA polymerase. Nyamiviruses have been found associated with diverse invertebrates as well as land- and seabirds. Members of the genera Nyavirus and Socyvirus produce enveloped, spherical virions. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Nyamiviridae, which is available at ictv.global/report/nyamiviridae.


Subject(s)
Mononegavirales/classification , Mononegavirales/isolation & purification , Animals , Genome, Viral , Invertebrates/virology , Mononegavirales/genetics , Phylogeny , RNA, Viral/genetics , Viral Proteins/genetics , Virion/classification , Virion/genetics , Virion/isolation & purification
17.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31748389

ABSTRACT

CpG dinucleotides are suppressed in the genomes of many vertebrate RNA viruses, including HIV-1. The cellular antiviral protein ZAP (zinc finger antiviral protein) binds CpGs and inhibits HIV-1 replication when CpGs are introduced into the viral genome. However, it is not known if ZAP-mediated restriction is the only mechanism driving CpG suppression. To determine how CpG dinucleotides affect HIV-1 replication, we increased their abundance in multiple regions of the viral genome and analyzed the effect on RNA expression, protein abundance, and infectious-virus production. We found that the antiviral effect of CpGs was not correlated with their abundance. Interestingly, CpGs inserted into some regions of the genome sensitize the virus to ZAP antiviral activity more efficiently than insertions into other regions, and this sensitivity can be modulated by interferon treatment or ZAP overexpression. Furthermore, the sensitivity of the virus to endogenous ZAP was correlated with its sensitivity to the ZAP cofactor KHNYN. Finally, we show that CpGs in some contexts can also inhibit HIV-1 replication by ZAP-independent mechanisms, and one of these is the activation of a cryptic splice site at the expense of a canonical splice site. Overall, we show that the location and sequence context of the CpG in the viral genome determines its antiviral activity.IMPORTANCE Some RNA virus genomes are suppressed in the nucleotide combination of a cytosine followed by a guanosine (CpG), indicating that they are detrimental to the virus. The antiviral protein ZAP binds viral RNA containing CpGs and prevents the virus from multiplying. However, it remains unknown how the number and position of CpGs in viral genomes affect restriction by ZAP and whether CpGs have other antiviral mechanisms. Importantly, manipulating the CpG content in viral genomes could help create new vaccines. HIV-1 shows marked CpG suppression, and by introducing CpGs into its genome, we show that ZAP efficiently targets a specific region of the viral genome, that the number of CpGs does not predict the magnitude of antiviral activity, and that CpGs can inhibit HIV-1 gene expression through a ZAP-independent mechanism. Overall, the position of CpGs in the HIV-1 genome determines the magnitude and mechanism through which they inhibit the virus.


Subject(s)
Dinucleoside Phosphates/metabolism , Gene Expression Regulation, Viral/physiology , HIV-1/physiology , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Virus Replication/physiology , Dinucleoside Phosphates/genetics , HEK293 Cells , Humans , Muramidase , Peptide Fragments , RNA, Viral/genetics , RNA-Binding Proteins/genetics
18.
Nucleic Acids Res ; 47(15): 8207-8223, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31180502

ABSTRACT

Many viruses utilize programmed -1 ribosomal frameshifting (-1 PRF) to express additional proteins or to produce frameshift and non-frameshift protein products at a fixed stoichiometric ratio. PRF is also utilized in the expression of a small number of cellular genes. Frameshifting is typically stimulated by signals contained within the mRNA: a 'slippery' sequence and a 3'-adjacent RNA structure. Recently, we showed that -1 PRF in encephalomyocarditis virus (EMCV) is trans-activated by the viral 2A protein, leading to a temporal change in PRF efficiency from 0% to 70% during virus infection. Here we analyzed PRF in the related Theiler's murine encephalomyelitis virus (TMEV). We show that 2A is also required for PRF in TMEV and can stimulate PRF to levels as high as 58% in rabbit reticulocyte cell-free translations and 81% during virus infection. We also show that TMEV 2A trans-activates PRF on the EMCV signal but not vice versa. We present an extensive mutational analysis of the frameshift stimulators (mRNA signals and 2A protein) analysing activity in in vitro translation, electrophoretic mobility shift and in vitro ribosome pausing assays. We also investigate the PRF mRNA signal with RNA structure probing. Our results substantially extend previous characterization of protein-stimulated PRF.


Subject(s)
Frameshifting, Ribosomal/genetics , Protein Biosynthesis/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Ribosomes/genetics , Theilovirus/genetics , Animals , Base Sequence , Mice , Nucleic Acid Conformation , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Viral/chemistry , RNA, Viral/metabolism , Ribosomes/metabolism , Theilovirus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
19.
Nucleic Acids Res ; 47(17): 9386-9399, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31396629

ABSTRACT

In all biological systems, RNAs are associated with RNA-binding proteins (RBPs), forming complexes that control gene regulatory mechanisms, from RNA synthesis to decay. In mammalian mitochondria, post-transcriptional regulation of gene expression is conducted by mitochondrial RBPs (mt-RBPs) at various stages of mt-RNA metabolism, including polycistronic transcript production, its processing into individual transcripts, mt-RNA modifications, stability, translation and degradation. To date, only a handful of mt-RBPs have been characterized. Here, we describe a putative human mitochondrial protein, C6orf203, that contains an S4-like domain-an evolutionarily conserved RNA-binding domain previously identified in proteins involved in translation. Our data show C6orf203 to bind highly structured RNA in vitro and associate with the mitoribosomal large subunit in HEK293T cells. Knockout of C6orf203 leads to a decrease in mitochondrial translation and consequent OXPHOS deficiency, without affecting mitochondrial RNA levels. Although mitoribosome stability is not affected in C6orf203-depleted cells, mitoribosome profiling analysis revealed a global disruption of the association of mt-mRNAs with the mitoribosome, suggesting that C6orf203 may be required for the proper maturation and functioning of the mitoribosome. We therefore propose C6orf203 to be a novel RNA-binding protein involved in mitochondrial translation, expanding the repertoire of factors engaged in this process.


Subject(s)
Mitochondria/genetics , Mitochondrial Proteins/biosynthesis , RNA, Mitochondrial/genetics , RNA-Binding Proteins/genetics , Animals , HEK293 Cells , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Mitochondrial Ribosomes/metabolism , RNA, Messenger/genetics , RNA, Ribosomal/genetics , RNA-Binding Proteins/physiology
20.
J Gen Virol ; 101(10): 1085-1089, 2020 10.
Article in English | MEDLINE | ID: mdl-32667280

ABSTRACT

Identification of the full complement of genes in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a crucial step towards gaining a fuller understanding of its molecular biology. However, short and/or overlapping genes can be difficult to detect using conventional computational approaches, whereas high-throughput experimental approaches - such as ribosome profiling - cannot distinguish translation of functional peptides from regulatory translation or translational noise. By studying regions showing enhanced conservation at synonymous sites in alignments of SARS-CoV-2 and related viruses (subgenus Sarbecovirus) and correlating the results with the conserved presence of an open reading frame (ORF) and a plausible translation mechanism, a putative new gene - ORF3c - was identified. ORF3c overlaps ORF3a in an alternative reading frame. A recently published ribosome profiling study confirmed that ORF3c is indeed translated during infection. ORF3c is conserved across the subgenus Sarbecovirus, and encodes a 40-41 amino acid predicted transmembrane protein.


Subject(s)
Betacoronavirus/genetics , Genes, Overlapping/genetics , Reading Frames/genetics , Amino Acid Sequence/genetics , COVID-19 , Coronavirus Infections/virology , Humans , Pandemics , Phylogeny , Pneumonia, Viral/virology , SARS-CoV-2 , Sequence Alignment , Viral Regulatory and Accessory Proteins/genetics , Viroporin Proteins
SELECTION OF CITATIONS
SEARCH DETAIL