727 results on '"Internal Ribosome Entry Sites"'
Search Results
2. DeepCIP: A multimodal deep learning method for the prediction of internal ribosome entry sites of circRNAs
- Author
-
Zhou, Yuxuan, Wu, Jingcheng, Yao, Shihao, Xu, Yulian, Zhao, Wenbin, Tong, Yunguang, and Zhou, Zhan
- Published
- 2023
- Full Text
- View/download PDF
3. Interplay between subthalamic nucleus and spinal cord controls parkinsonian nociceptive disorders.
- Author
-
Charles, Keri-Ann, Sierra, Elba Molpeceres, Bouali-Benazzouz, Rabia, Tibar, Houyam, Oudaha, Khalid, Naudet, Frédéric, Duveau, Alexia, Fossat, Pascal, and Benazzouz, Abdelhamid
- Subjects
- *
DEEP brain stimulation , *PARKINSON'S disease , *PARKINSONIAN disorders , *DESIGNER drugs , *SPINAL cord , *SUBTHALAMIC nucleus - Abstract
Pain is a non-motor symptom that impairs quality of life in patients with Parkinson's disease. Pathological nociceptive hypersensitivity in patients could be due to changes in the processing of somatosensory information at the level of the basal ganglia, including the subthalamic nucleus (STN), but the underlying mechanisms are not yet defined. Here, we investigated the interaction between the STN and the dorsal horn of the spinal cord (DHSC), by first examining the nature of STN neurons that respond to peripheral nociceptive stimulation and the nature of their responses under normal and pathological conditions. Next, we studied the consequences of deep brain stimulation (DBS) of the STN on the electrical activity of DHSC neurons. Then, we investigated whether the therapeutic effect of STN-DBS would be mediated by the brainstem descending pathway involving the rostral ventromedial medulla. Finally, to better understand how the STN modulates allodynia, we used Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) expressed in the STN. The study was carried out on the 6-OHDA rodent model of Parkinson's disease, obtained by stereotactic injection of the neurotoxin into the medial forebrain bundle of rats and mice. In these animals, we used motor and nociceptive behavioural tests, in vivo electrophysiology of STN and wide dynamic range (WDR) DHSC neurons in response to peripheral stimulation, deep brain stimulation of the STN and the selective DREADD approach. Vglut2-ires-cre mice were used to specifically target and inhibit STN glutamatergic neurons. STN neurons are able to detect nociceptive stimuli, encode their intensity and generate windup-like plasticity, like WDR neurons in the DHSC. These phenomena are impaired in dopamine-depleted animals, as the intensity response is altered in both spinal and subthalamic neurons. Furthermore, as with L- DOPA, STN-DBS in rats ameliorated 6-OHDA-induced allodynia, and this effect is mediated by descending brainstem projections leading to normalization of nociceptive integration in DHSC neurons. Furthermore, this therapeutic effect was reproduced by selective inhibition of STN glutamatergic neurons in Vglut2-ires-cre mice. Our study highlights the centrality of the STN in nociceptive circuits, its interaction with the DHSC and its key involvement in pain sensation in Parkinson's disease. Furthermore, our results provide for the first-time evidence that subthalamic DBS produces analgesia by normalizing the responses of spinal WDR neurons via descending brainstem pathways. These effects are due to direct inhibition, rather than activation of glutamatergic neurons in the STN or passage fibres, as shown in the DREADDs experiment. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
4. m6A-modification of cyclin D1 and c-myc IRESs in glioblastoma controls ITAF activity and resistance to mTOR inhibition
- Author
-
Benavides-Serrato, Angelica, Saunders, Jacquelyn T, Kumar, Sunil, Holmes, Brent, Benavides, Kennedy E, Bashir, Muhammad T, Nishimura, Robert N, and Gera, Joseph
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Rare Diseases ,Brain Cancer ,Cancer ,Brain Disorders ,Genetics ,Neurosciences ,5.1 Pharmaceuticals ,Humans ,Cyclin D1 ,Glioblastoma ,Heterogeneous Nuclear Ribonucleoprotein A1 ,Internal Ribosome Entry Sites ,Methyltransferases ,Protein Biosynthesis ,TOR Serine-Threonine Kinases ,Genes ,myc ,Drug resistance ,mTOR inhibitors ,N6-methyladenosine modification ,IRES ,ITAF ,N(6)-methyladenosine modification ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
A major mechanism conferring resistance to mTOR inhibitors is activation of a salvage pathway stimulating internal ribosome entry site (IRES)-mediated mRNA translation, driving the synthesis of proteins promoting resistance of glioblastoma (GBM). Previously, we found this pathway is stimulated by the requisite IRES-trans-acting factor (ITAF) hnRNP A1, which itself is subject to phosphorylation and methylation events regulating cyclin D1 and c-myc IRES activity. Here we describe the requirement for m6A-modification of IRES RNAs for efficient translation and resistance to mTOR inhibition. DRACH-motifs within these IRES RNAs upon m6A modification resulted in enhanced IRES activity via increased hnRNP A1-binding following mTOR inhibitor exposure. Inhibitor exposure stimulated the expression of m6A-methylosome components resulting in increased activity in GBM. Silencing of METTL3-14 complexes reduced IRES activity upon inhibitor exposure and sensitized resistant GBM lines. YTHDF3 associates with m6A-modified cyclin D1 or c-myc IRESs, regulating IRES activity, and mTOR inhibitor sensitivity in vitro and in xenograft experiments. YTHDF3 interacted directly with hnRNP A1 and together stimulated hnRNP A1-dependent nucleic acid strand annealing activity. These data demonstrate that m6A-methylation of IRES RNAs regulate GBM responses to this class of inhibitors.
- Published
- 2023
5. Host-like RNA Elements Regulate Virus Translation.
- Author
-
Khan, Debjit and Fox, Paul L.
- Subjects
- *
TRANSFER RNA , *RNA , *GENE expression , *GAIT in humans , *VIRAL proteins , *CARRIER proteins - Abstract
Viruses are obligate, intracellular parasites that co-opt host cell machineries for propagation. Critical among these machineries are those that translate RNA into protein and their mechanisms of control. Most regulatory mechanisms effectuate their activity by targeting sequence or structural features at the RNA termini, i.e., at the 5′ or 3′ ends, including the untranslated regions (UTRs). Translation of most eukaryotic mRNAs is initiated by 5′ cap-dependent scanning. In contrast, many viruses initiate translation at internal RNA regions at internal ribosome entry sites (IRESs). Eukaryotic mRNAs often contain upstream open reading frames (uORFs) that permit condition-dependent control of downstream major ORFs. To offset genome compression and increase coding capacity, some viruses take advantage of out-of-frame overlapping uORFs (oORFs). Lacking the essential machinery of protein synthesis, for example, ribosomes and other translation factors, all viruses utilize the host apparatus to generate virus protein. In addition, some viruses exhibit RNA elements that bind host regulatory factors that are not essential components of the translation machinery. SARS-CoV-2 is a paradigm example of a virus taking advantage of multiple features of eukaryotic host translation control: the virus mimics the established human GAIT regulatory element and co-opts four host aminoacyl tRNA synthetases to form a stimulatory binding complex. Utilizing discontinuous transcription, the elements are present and identical in all SARS-CoV-2 subgenomic RNAs (and the genomic RNA). Thus, the virus exhibits a post-transcriptional regulon that improves upon analogous eukaryotic regulons, in which a family of functionally related mRNA targets contain elements that are structurally similar but lacking sequence identity. This "thrifty" virus strategy can be exploited against the virus since targeting the element can suppress the expression of all subgenomic RNAs as well as the genomic RNA. Other 3′ end viral elements include 3′-cap-independent translation elements (3′-CITEs) and 3′-tRNA-like structures. Elucidation of virus translation control elements, their binding proteins, and their mechanisms can lead to novel therapeutic approaches to reduce virus replication and pathogenicity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Cotranslational prolyl hydroxylation is essential for flavivirus biogenesis
- Author
-
Aviner, Ranen, Li, Kathy H, Frydman, Judith, and Andino, Raul
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Biodefense ,Vector-Borne Diseases ,Rare Diseases ,Emerging Infectious Diseases ,Biotechnology ,Neurosciences ,Genetics ,Infectious Diseases ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Infection ,Good Health and Well Being ,Cell Line ,Collagen ,Dengue Virus ,Flavivirus ,Gene Expression Regulation ,Viral ,Genomics ,Host-Derived Cellular Factors ,Host-Pathogen Interactions ,Humans ,Hydroxylation ,Internal Ribosome Entry Sites ,Molecular Chaperones ,Peptide Chain Initiation ,Translational ,Poliovirus ,Polyribosomes ,Procollagen-Proline Dioxygenase ,Proline ,Protein Aggregates ,Protein Biosynthesis ,Protein Folding ,Protein Interaction Maps ,Proteolysis ,Proteomics ,Zika Virus ,General Science & Technology - Abstract
Viral pathogens are an ongoing threat to public health worldwide. Analysing their dependence on host biosynthetic pathways could lead to effective antiviral therapies1. Here we integrate proteomic analyses of polysomes with functional genomics and pharmacological interventions to define how enteroviruses and flaviviruses remodel host polysomes to synthesize viral proteins and disable host protein production. We find that infection with polio, dengue or Zika virus markedly modifies polysome composition, without major changes to core ribosome stoichiometry. These viruses use different strategies to evict a common set of translation initiation and RNA surveillance factors from polysomes while recruiting host machineries that are specifically required for viral biogenesis. Targeting these specialized viral polysomes could provide a new approach for antiviral interventions. For example, we find that both Zika and dengue use the collagen proline hydroxylation machinery to mediate cotranslational modification of conserved proline residues in the viral polyprotein. Genetic or pharmacological inhibition of proline hydroxylation impairs nascent viral polyprotein folding and induces its aggregation and degradation. Notably, such interventions prevent viral polysome remodelling and lower virus production. Our findings delineate the modular nature of polysome specialization at the virus-host interface and establish a powerful strategy to identify targets for selective antiviral interventions.
- Published
- 2021
7. Repurposing Potential of Riluzole as an ITAF Inhibitor in mTOR Therapy Resistant Glioblastoma
- Author
-
Benavides-Serrato, Angelica, Saunders, Jacquelyn T, Holmes, Brent, Nishimura, Robert N, Lichtenstein, Alan, and Gera, Joseph
- Subjects
Medicinal and Biomolecular Chemistry ,Chemical Sciences ,Rare Diseases ,Brain Disorders ,Orphan Drug ,Cancer ,Brain Cancer ,Neurosciences ,5.1 Pharmaceuticals ,Animals ,Antineoplastic Agents ,Apoptosis ,Cell Line ,Tumor ,Cell Movement ,Cell Proliferation ,Drug Repositioning ,Drug Resistance ,Neoplasm ,Drug Synergism ,Female ,Glioblastoma ,Heterogeneous Nuclear Ribonucleoprotein A1 ,Humans ,Internal Ribosome Entry Sites ,Mice ,Mice ,SCID ,Molecular Docking Simulation ,Protein Biosynthesis ,Riluzole ,TOR Serine-Threonine Kinases ,riluzole ,hnRNP A1 ,ITAF ,mTOR ,drug resistance ,glioblastoma ,Other Chemical Sciences ,Genetics ,Other Biological Sciences ,Chemical Physics ,Biochemistry and cell biology ,Microbiology ,Medicinal and biomolecular chemistry - Abstract
Internal ribosome entry site (IRES)-mediated protein synthesis has been demonstrated to play an important role in resistance to mechanistic target of rapamycin (mTOR) targeted therapies. Previously, we have demonstrated that the IRES trans-acting factor (ITAF), hnRNP A1 is required to promote IRES activity and small molecule inhibitors which bind specifically to this ITAF and curtail IRES activity, leading to mTOR inhibitor sensitivity. Here we report the identification of riluzole (Rilutek®), an FDA-approved drug for amyotrophic lateral sclerosis (ALS), via an in silico docking analysis of FDA-approved compounds, as an inhibitor of hnRNP A1. In a riluzole-bead coupled binding assay and in surface plasmon resonance imaging analyses, riluzole was found to directly bind to hnRNP A1 and inhibited IRES activity via effects on ITAF/RNA-binding. Riluzole also demonstrated synergistic anti-glioblastoma (GBM) affects with mTOR inhibitors in vitro and in GBM xenografts in mice. These data suggest that repurposing riluzole, used in conjunction with mTOR inhibitors, may serve as an effective therapeutic option in glioblastoma.
- Published
- 2020
8. The protein arginine methyltransferase PRMT5 confers therapeutic resistance to mTOR inhibition in glioblastoma
- Author
-
Holmes, Brent, Benavides-Serrato, Angelica, Saunders, Jacquelyn T, Landon, Kenna A, Schreck, Adam J, Nishimura, Robert N, and Gera, Joseph
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Oncology and Carcinogenesis ,Rare Diseases ,Brain Disorders ,Orphan Drug ,Genetics ,Cancer ,Brain Cancer ,5.1 Pharmaceuticals ,Animals ,Apoptosis ,Cell Proliferation ,Cyclin D1 ,DNA Methylation ,Drug Resistance ,Neoplasm ,Gene Expression Regulation ,Neoplastic ,Glioblastoma ,Heterogeneous Nuclear Ribonucleoprotein A1 ,Humans ,Indoles ,Internal Ribosome Entry Sites ,Isoquinolines ,Mice ,Protein-Arginine N-Methyltransferases ,Proto-Oncogene Proteins c-myc ,Purines ,Pyrimidines ,Signal Transduction ,TOR Serine-Threonine Kinases ,Tumor Cells ,Cultured ,Xenograft Model Antitumor Assays ,PRMT5 ,mTOR ,PP242 ,Rapamycin ,EPZ015666 ,Drug resistance ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
IntroductionClinical trials directed at mechanistic target of rapamycin (mTOR) inhibition have yielded disappointing results in glioblastoma (GBM). A major mechanism of resistance involves the activation of a salvage pathway stimulating internal ribosome entry site (IRES)-mediated protein synthesis. PRMT5 activity has been implicated in the enhancement of IRES activity.MethodsWe analyzed the expression and activity of PRMT5 in response to mTOR inhibition in GBM cell lines and short-term patient cultures. To determine whether PRMT5 conferred resistance we used genetic and pharmacological approaches to ablate PRMT5 activity and assessed the effects on in vitro and in vivo sensitivity. Mutational analyses of the requisite IRES-trans-acting factor (ITAF), hnRNP A1 determined whether PRMT5-mediated methylation was necessary for ITAF RNA binding and IRES activity.ResultsPRMT5 activity is stimulated in response to mTOR inhibitors. Knockdown or treatment with a PRMT5 inhibitor blocked IRES activity and sensitizes GBM cells. Ectopic expression of non-methylatable hnRNP A1 mutants demonstrated that methylation of either arginine residues 218 or 225 was sufficient to maintain IRES binding and hnRNP A1-dependent cyclin D1 or c-MYC IRES activity, however a double R218K/R225K mutant was unable to do so. The PRMT5 inhibitor EPZ015666 displayed synergistic anti-GBM effects in vitro and in a xenograft mouse model in combination with PP242.ConclusionsThese results demonstrate that PRMT5 activity is stimulated upon mTOR inhibition in GBM. Our data further support a signaling cascade in which PRMT5-mediated methylation of hnRNP A1 promotes IRES RNA binding and activation of IRES-mediated protein synthesis and resultant mTOR inhibitor resistance.
- Published
- 2019
9. Direct and Indirect Effects on Viral Translation and RNA Replication Are Required for AUF1 Restriction of Enterovirus Infections in Human Cells
- Author
-
Ullmer, Wendy and Semler, Bert L
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Biodefense ,Infectious Diseases ,Genetics ,Emerging Infectious Diseases ,Digestive Diseases ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Infection ,Good Health and Well Being ,Enterovirus B ,Human ,Gene Knockdown Techniques ,HEK293 Cells ,HeLa Cells ,Heterogeneous Nuclear Ribonucleoprotein D0 ,Heterogeneous-Nuclear Ribonucleoprotein D ,Host Microbial Interactions ,Host-Pathogen Interactions ,Humans ,Internal Ribosome Entry Sites ,Poliovirus ,Protein Binding ,Protein Biosynthesis ,Protein Transport ,Real-Time Polymerase Chain Reaction ,Virus Replication ,AUF1 ,IRES-mediated translation ,RNA replication ,coxsackievirus ,enterovirus ,host restriction ,picornavirus ,poliovirus ,Hela Cells ,Microbiology ,Biochemistry and cell biology ,Medical microbiology - Abstract
The cellular mRNA decay protein AUF1 acts as a restriction factor during infection by picornaviruses, including poliovirus, coxsackievirus, and human rhinovirus. AUF1 relocalizes from the nucleus to the cytoplasm during infection by these viruses due to the disruption of nucleocytoplasmic trafficking by viral proteinases. Previous studies have demonstrated that AUF1 binds to poliovirus and coxsackievirus B3 (CVB3) RNA during infection, with binding shown to occur within the internal ribosome entry site (IRES) of the 5' noncoding region (NCR) or the 3' NCR, respectively. Binding to different sites within the viral RNA suggests that AUF1 may negatively regulate infection by these viruses using different mechanisms. The work presented here addresses the mechanism of AUF1 inhibition of the replication of poliovirus and CVB3. We demonstrate that AUF1 knockdown in human cells results in increased viral translation, RNA synthesis, and virus production. AUF1 is shown to negatively regulate translation of a poliovirus and CVB3 IRES reporter RNA during infection but not in uninfected cells. We found that this inhibitory activity is not mediated through destabilization of viral genomic RNA; however, it does require virus-induced relocalization of AUF1 from the nucleus to the cytoplasm during the early phases of infection. Our findings suggest that AUF1 restriction of poliovirus and CVB3 replication uses a common mechanism through the viral IRES, which is distinct from the canonical role that AUF1 plays in regulated mRNA decay in uninfected host cells.IMPORTANCE Picornaviruses primarily infect the gastrointestinal or upper respiratory tracts of humans and animals and may disseminate to tissues of the central nervous system, heart, skin, liver, or pancreas. Many common human pathogens belong to the Picornaviridae family, which includes viruses known to cause paralytic poliomyelitis (poliovirus); myocarditis (coxsackievirus B3 [CVB3]); the common cold (human rhinovirus [HRV]); and hand, foot, and mouth disease (enterovirus 71 [EV71]), among other illnesses. There are no specific treatments for infection, and vaccines exist for only two picornaviruses: poliovirus and hepatitis A virus. Given the worldwide distribution and prevalence of picornaviruses, it is important to gain insight into the host mechanisms used to restrict infection. Other than proteins involved in the innate immune response, few host factors have been identified that restrict picornavirus replication. The work presented here seeks to define the mechanism of action for the host restriction factor AUF1 during infection by poliovirus and CVB3.
- Published
- 2018
10. Molecular mechanism of poliovirus Sabin vaccine strain attenuation
- Author
-
Avanzino, Brian C, Jue, Helen, Miller, Clare M, Cheung, Emily, Fuchs, Gabriele, and Fraser, Christopher S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Emerging Infectious Diseases ,Genetics ,Biodefense ,Infectious Diseases ,Good Health and Well Being ,Animals ,Baculoviridae ,Base Sequence ,Cloning ,Molecular ,Escherichia coli ,Eukaryotic Initiation Factor-4A ,Eukaryotic Initiation Factor-4G ,Gene Expression ,Genes ,Reporter ,Genetic Vectors ,HeLa Cells ,Humans ,Internal Ribosome Entry Sites ,Luciferases ,Mutation ,Nucleic Acid Conformation ,Poliovirus ,Poliovirus Vaccine ,Oral ,Polypyrimidine Tract-Binding Protein ,Protein Biosynthesis ,Recombinant Proteins ,Sequence Alignment ,Sf9 Cells ,Spodoptera ,Vaccines ,Attenuated ,poliovirus ,eukaryotic translation initiation factor 4G ,eukaryotic initiation factor 4A ,RNA-binding protein ,eukaryotic translation initiation ,plus-stranded RNA virus ,internal ribosome entry site ,vaccine ,virulence ,Hela Cells ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Recruitment of poliovirus (PV) RNA to the human ribosome requires the coordinated interaction of the viral internal ribosome entry site (IRES) and several host cellular initiation factors and IRES trans-acting factors (ITAFs). Attenuated PV Sabin strains contain point mutations in the PV IRES domain V (dV) that inhibit viral translation. Remarkably, attenuation is most apparent in cells of the central nervous system, but the molecular basis to explain this is poorly understood. The dV contains binding sites for eukaryotic initiation factor 4G (eIF4G) and polypyrimidine tract-binding protein (PTB). Impaired binding of these proteins to the mutant IRESs has been observed, but these effects have not been quantitated. We used a fluorescence anisotropy assay to reveal that the Sabin mutants reduce the equilibrium dissociation constants of eIF4G and PTB to the PV IRES by up to 6-fold. Using the most inhibitory Sabin 3 mutant, we used a real-time fluorescence helicase assay to show that the apparent affinity of an active eIF4G/4A/4B helicase complex for the IRES is reduced by 2.5-fold. The Sabin 3 mutant did not alter the maximum rate of eIF4A-dependent helicase activity, suggesting that this mutant primarily reduces the affinity, rather than activity, of the unwinding complex. To confirm this affinity model of attenuation, we show that eIF4G overexpression in HeLa cells overcomes the attenuation of a Sabin 3 mutant PV-luciferase replicon. Our study provides a quantitative framework for understanding the mechanism of PV Sabin attenuation and provides an explanation for the previously observed cell type-specific translational attenuation.
- Published
- 2018
11. Translation of circular RNAs.
- Author
-
Margvelani G, Maquera KAA, Welden JR, Rodgers DW, and Stamm S
- Subjects
- Humans, Animals, RNA Editing, RNA, Messenger genetics, RNA, Messenger metabolism, Adenosine analogs & derivatives, Adenosine metabolism, Adenosine genetics, Internal Ribosome Entry Sites, Eukaryotic Initiation Factor-3 metabolism, Eukaryotic Initiation Factor-3 genetics, MicroRNAs metabolism, MicroRNAs genetics, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics, Neoplasms genetics, Neoplasms metabolism, Eukaryotic Initiation Factor-4A, DEAD-box RNA Helicases, RNA, Circular metabolism, RNA, Circular genetics, Protein Biosynthesis, RNA genetics, RNA metabolism
- Abstract
Circular RNAs (circRNAs) are covalently closed RNAs that are present in all eukaryotes tested. Recent RNA sequencing (RNA-seq) analyses indicate that although generally less abundant than messenger RNAs (mRNAs), over 1.8 million circRNA isoforms exist in humans, much more than the number of currently known mRNA isoforms. Most circRNAs are generated through backsplicing that depends on pre-mRNA structures, which are influenced by intronic elements, for example, primate-specific Alu elements, leading to species-specific circRNAs. CircRNAs are mostly cytosolic, stable and some were shown to influence cells by sequestering miRNAs and RNA-binding proteins. We review the increasing evidence that circRNAs are translated into proteins using several cap-independent translational mechanisms, that include internal ribosomal entry sites, N6-methyladenosine RNA modification, adenosine to inosine RNA editing and interaction with the eIF4A3 component of the exon junction complex. CircRNAs are translated under conditions that favor cap-independent translation, notably in cancer and generate proteins that are shorter than mRNA-encoded proteins, which can acquire new functions relevant in diseases., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2025
- Full Text
- View/download PDF
12. Stimulation of IRES-Dependent Translation by Rocaglamide A Increases the Replication and Virulence of Cricket Paralysis Virus in Lepidopteran Insect Cells.
- Author
-
Santos D, Christopoulou VM, Taning CNT, Avgeris S, Papadopoulou A, Kletsas D, Voutsinas GE, Labropoulou V, and Swevers L
- Subjects
- Animals, Virulence, Cell Line, Benzofurans pharmacology, Protein Biosynthesis drug effects, Dicistroviridae pathogenicity, Virus Replication drug effects, Internal Ribosome Entry Sites
- Abstract
The discovery that infections of viruses are pervasive among insects has considerable potential for future applications, such as new strategies for pest control through the manipulation of virus-host interactions. However, few studies can be found that aim to minimize (for beneficial insects) or maximize (for pests) virus impact or virulence. Viruses generally employ molecular mechanisms that deviate from the cells' to increase their replication efficiency and to avoid the immune response. In this research, a screening system is presented for the detection of molecules that interfere with the internal ribosomal entry site (IRES) of Cricket paralysis virus (Dicistroviridae) which has been well characterized in previous research. Over-expression and RNAi experiments identified the importance of eIF4A, a component of the cap-dependent translation initiation complex, to modify the activity of IRES-mediated translation. Application of Rocaglamide A (RocA), a natural product from Aglaia plants and inhibitor of eIF4A, resulted in strong stimulation of IRES-mediated translation in reporter assays as well as increased CrPV genome replication and virion production in lepidopteran Hi5 cells. At 100 nM of RocA, dsRNA molecules accumulated in infected cells, corresponding to full-length genome (9.5 kb) and a smaller fragment (0.8 kb) with unknown function. Treatment of silkworm larvae with RocA by injection or topically was highly toxic while no strong stimulation of CrPV infection could be observed. The prospect of the use of rocaglamates as insecticides and enhancers of CrPV infection is discussed together with its potential impact on mammalian cells., (© 2025 Wiley Periodicals LLC.)
- Published
- 2025
- Full Text
- View/download PDF
13. Heterogeneous nuclear ribonucleoprotein A3 binds to the internal ribosomal entry site of enterovirus A71 and affects virus replication in neural cells.
- Author
-
Lin JY, Lin JY, Kuo RL, and Huang HI
- Subjects
- Humans, Neurons metabolism, Neurons virology, Protein Binding, Enterovirus Infections virology, Enterovirus Infections metabolism, Enterovirus Infections genetics, Virus Replication, Internal Ribosome Entry Sites, Enterovirus A, Human genetics, Enterovirus A, Human physiology, Enterovirus A, Human metabolism, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Heterogeneous-Nuclear Ribonucleoprotein Group A-B genetics, 5' Untranslated Regions
- Abstract
Enterovirus A71 (EV-A71) belongs to the genus Enterovirus of the Picornaviridae family and often causes outbreaks in Asia. EV-A71 infection usually causes hand, foot, and mouth disease and can even affect the central nervous system, causing neurological complications or death. The 5'-untranslated region (5'-UTR) of EV-A71 contains an internal ribosome entry site (IRES) that is responsible for the translation of viral proteins. IRES-transacting factors can interact with the EV-A71 5'-UTR to regulate IRES activity. Heterogeneous nuclear ribonucleoprotein (hnRNP) A3 is a member of the hnRNP A/B protein family of RNA-binding proteins and is involved in RNA transport and modification. We found that hnRNP A3 knockdown promoted the replication of EV-A71 in neural calls. Conversely, increasing the expression of hnRNP A3 within cells inhibits the growth of EV-A71. HnRNP A3 can bind to the EV-A71 5'-UTR, and knockdown of hnRNP A3 enhances the luciferase activity of the EV-A71 5'-UTR IRES. The localization of hnRNP A3 shifts from the nucleus to the cytoplasm of infected cells during viral infection. Additionally, EV-A71 infection can increase the protein expression of hnRNP A3, and the protein level is correlated with efficient viral growth. Based on these findings, we concluded that hnRNP A3 plays a negative regulatory role in EV-A71 replication within neural cells., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
14. Altered Baseline and Nicotine-Mediated Behavioral and Cholinergic Profiles in ChAT-Cre Mouse Lines
- Author
-
Chen, Edison, Lallai, Valeria, Sherafat, Yasmine, Grimes, Nickolas P, Pushkin, Anna N, Fowler, JP, and Fowler, Christie D
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Neurosciences ,Substance Misuse ,Brain Disorders ,Behavioral and Social Science ,Mental Illness ,Biotechnology ,Genetics ,Mental Health ,Basic Behavioral and Social Science ,Drug Abuse (NIDA only) ,Mental health ,Animals ,Behavior ,Animal ,Choline O-Acetyltransferase ,Chromosomes ,Artificial ,Bacterial ,Female ,Genetic Techniques ,Hippocampus ,Integrases ,Internal Ribosome Entry Sites ,Locomotion ,Male ,Mice ,Mice ,Transgenic ,Models ,Animal ,Nicotine ,Nicotinic Agonists ,Vesicular Acetylcholine Transport Proteins ,acetylcholine ,choline acetyltransferase ,cre recombinase ,intravenous nicotine self-administration ,mice ,transgenic ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
The recent development of transgenic rodent lines expressing cre recombinase in a cell-specific manner, along with advances in engineered viral vectors, has permitted in-depth investigations into circuit function. However, emerging evidence has begun to suggest that genetic modifications may introduce unexpected caveats. In the current studies, we sought to extensively characterize male and female mice from both the ChAT(BAC)-Cre mouse line, created with the bacterial artificial chromosome (BAC) method, and ChAT(IRES)-Cre mouse line, generated with the internal ribosome entry site (IRES) method. ChAT(BAC)-Cre transgenic and wild-type mice did not differ in general locomotor behavior, anxiety measures, drug-induced cataplexy, nicotine-mediated hypolocomotion, or operant food training. However, ChAT(BAC)-Cre transgenic mice did exhibit significant deficits in intravenous nicotine self-administration, which paralleled an increase in vesicular acetylcholine transporter and choline acetyltransferase (ChAT) hippocampal expression. For the ChAT(IRES)-Cre line, transgenic mice exhibited deficits in baseline locomotor, nicotine-mediated hypolocomotion, and operant food training compared with wild-type and hemizygous littermates. No differences among ChAT(IRES)-Cre wild-type, hemizygous, and transgenic littermates were found in anxiety measures, drug-induced cataplexy, and nicotine self-administration. Given that increased cre expression was present in the ChAT(IRES)-Cre transgenic mice, as well as a decrease in ChAT expression in the hippocampus, altered neuronal function may underlie behavioral phenotypes. In contrast, ChAT(IRES)-Cre hemizygous mice were more similar to wild-type mice in both protein expression and the majority of behavioral assessments. As such, interpretation of data derived from ChAT-Cre rodents must consider potential limitations dependent on the line and/or genotype used in research investigations.SIGNIFICANCE STATEMENT Altered baseline and/or nicotine-mediated behavioral profiles were discovered in transgenic mice from the ChAT(BAC)-Cre and ChAT(IRES)-Cre lines. Given that these cre-expressing mice have become increasingly used by the scientific community, either independently with chemicogenetic and optogenetic viral vectors or crossed with other transgenic lines, the current studies highlight important considerations for the interpretation of data from previous and future experimental investigations. Moreover, the current findings detail the behavioral effects of either increased or decreased baseline cholinergic signaling mechanisms on locomotor, anxiety, learning/memory, and intravenous nicotine self-administration behaviors.
- Published
- 2018
15. Cellular cap-binding protein, eIF4E, promotes picornavirus genome restructuring and translation
- Author
-
Avanzino, Brian C, Fuchs, Gabriele, and Fraser, Christopher S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Biodefense ,Infectious Diseases ,Emerging Infectious Diseases ,Generic health relevance ,Good Health and Well Being ,Animals ,Cell-Free System ,Encephalomyocarditis virus ,Eukaryotic Initiation Factor-4A ,Eukaryotic Initiation Factor-4E ,Eukaryotic Initiation Factor-4F ,Eukaryotic Initiation Factor-4G ,Genome ,Viral ,Hepatitis A virus ,Humans ,In Vitro Techniques ,Internal Ribosome Entry Sites ,Models ,Biological ,Picornaviridae ,Poliovirus ,Protein Biosynthesis ,Rabbits ,Replicon ,translation initiation ,IRES ,eIF4E ,eIF4A ,picornavirus - Abstract
Picornaviruses use internal ribosome entry sites (IRESs) to translate their genomes into protein. A typical feature of these IRESs is their ability to bind directly to the eukaryotic initiation factor (eIF) 4G component of the eIF4F cap-binding complex. Remarkably, the hepatitis A virus (HAV) IRES requires eIF4E for its translation, but no mechanism has been proposed to explain this. Here we demonstrate that eIF4E regulates HAV IRES-mediated translation by two distinct mechanisms. First, eIF4E binding to eIF4G generates a high-affinity binding conformation of the eIF4F complex for the IRES. Second, eIF4E binding to eIF4G strongly stimulates the rate of duplex unwinding by eIF4A on the IRES. Our data also reveal that eIF4E promotes eIF4F binding and increases the rate of restructuring of the poliovirus (PV) IRES. This provides a mechanism to explain why PV IRES-mediated translation is stimulated by eIF4E availability in nuclease-treated cell-free extracts. Using a PV replicon and purified virion RNA, we also show that eIF4E promotes the rate of eIF4G cleavage by the 2A protease. Finally, we show that cleavage of eIF4G by the poliovirus 2A protease generates a high-affinity IRES binding truncation of eIF4G that stimulates eIF4A duplex unwinding independently of eIF4E. Therefore, our data reveal how picornavirus IRESs use eIF4E-dependent and -independent mechanisms to promote their translation.
- Published
- 2017
16. Internal Ribosome Entry Site (IRES)-Mediated Translation and Its Potential for Novel mRNA-Based Therapy Development.
- Author
-
Marques, Rita, Lacerda, Rafaela, and Romão, Luísa
- Subjects
MUSCULAR atrophy ,PROTEIN synthesis ,THERAPEUTICS ,NEUROLOGICAL disorders ,DRUG target - Abstract
Many conditions can benefit from RNA-based therapies, namely, those targeting internal ribosome entry sites (IRESs) and their regulatory proteins, the IRES trans-acting factors (ITAFs). IRES-mediated translation is an alternative mechanism of translation initiation, known for maintaining protein synthesis when canonical translation is impaired. During a stress response, it contributes to cell reprogramming and adaptation to the new environment. The relationship between IRESs and ITAFs with tumorigenesis and resistance to therapy has been studied in recent years, proposing new therapeutic targets and treatments. In addition, IRES-dependent translation initiation dysregulation is also related to neurological and cardiovascular diseases, muscular atrophies, or other syndromes. The participation of these structures in the development of such pathologies has been studied, yet to a far lesser extent than in cancer. Strategies involving the disruption of IRES–ITAF interactions or the modification of ITAF expression levels may be used with great impact in the development of new therapeutics. In this review, we aim to comprehend the current data on groups of human pathologies associated with IRES and/or ITAF dysregulation and their application in the designing of new therapeutic approaches using them as targets or tools. Thus, we wish to summarise the evidence in the field hoping to open new promising lines of investigation toward personalised treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
17. Mammalian Polycistronic mRNAs and Disease
- Author
-
Karginov, Timofey A, Pastor, Daniel Parviz Hejazi, Semler, Bert L, and Gomez, Christopher M
- Subjects
Biological Sciences ,Genetics ,Orphan Drug ,Rare Diseases ,Animals ,Genetic Diseases ,Inborn ,Humans ,Internal Ribosome Entry Sites ,Mammals ,Molecular Targeted Therapy ,Protein Biosynthesis ,RNA ,Messenger ,IRES ,ITAF ,bicistronic ,cap-independent ,eIF ,polycistronic ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Our understanding of gene expression has come far since the 'one-gene one-polypeptide' hypothesis proposed by Beadle and Tatum. In this review, we address the gradual recognition that a growing number of polycistronic genes, originally discovered in viruses, are being identified within the mammalian genome, and that these may provide new insights into disease mechanisms and treatment. We carried out a systematic literature review identifying 13 mammalian genes for which there is evidence for polycistronic expression via translation through an internal ribosome entry site (IRES). Although the canonical mechanism of translation initiation has been studied extensively, here we highlight a process of noncanonical translation, IRES-mediated translation, that is a growing source for understanding complex inheritance, the elucidation of disease mechanisms, and the discovery of novel therapeutic targets. Identification of additional polycistronic genes may provide new insights into disease therapy and allow for new discoveries of both translational and disease mechanisms.
- Published
- 2017
18. Extensive Replication of a Retroviral Replicating Vector Can Expand the A Bulge in the Encephalomyocarditis Virus Internal Ribosome Entry Site and Change Translation Efficiency of the Downstream Transgene
- Author
-
Lin, Amy H, Liu, Yanzheng, Burrascano, Cynthia, Cunanan, Kathrina, Logg, Christopher R, Robbins, Joan M, Kasahara, Noriyuki, Gruber, Harry, Ibañez, Carlos, and Jolly, Douglas J
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Rare Diseases ,Gene Therapy ,Genetics ,Biotechnology ,Infectious Diseases ,Infection ,Animals ,Cell Line ,Tumor ,Cytosine Deaminase ,Encephalomyocarditis virus ,Female ,Genetic Vectors ,Green Fluorescent Proteins ,HEK293 Cells ,Humans ,Internal Ribosome Entry Sites ,Mice ,Inbred BALB C ,Moloney murine leukemia virus ,Protein Biosynthesis ,RNA ,Viral ,Transgenes - Abstract
We have developed retroviral replicating vectors (RRV) derived from Moloney murine gammaretrovirus with an amphotropic envelope and an encephalomyocarditis virus (EMCV) internal ribosome entry site (IRES)-transgene cassette downstream of the env gene. During long-term (180 days) replication of the vector in animals, a bulge of 7 adenosine residues (A's) in the J-K bifurcation domain sometimes serially added A's. Therefore, vectors with 4-12 A's in the A bulge in the J-K bifurcation domain were generated, and the impact of the variants on transgene protein expression, vector stability, and IRES sequence upon multiple infection cycles was assessed in RRV encoding yeast-derived cytosine deaminase and green fluorescent protein in vitro. For transgene protein expression, after multiple infection cycles, RRV-IRES with 5-7 A's gave roughly comparable levels, 4 and 8 A's were within about 4-5-fold of the 6 A's, whereas 10 and 12 A's were marked lower. In terms of stability, after 10 infection cycles, expansion of A's appeared to be a more frequent event affecting transgene protein expression than viral genome deletions or rearrangement: 4 and 5 A's appeared completely stable; 6, 7, and particularly 8 A's showed some level of expansion in the A bulge; 10 and 12 A's underwent both expansion and transgene deletion. The strong relative translational activity of the 5 A's in the EMCV IRES has not been reported previously. The 5A RRV-IRES may have utility for preclinical and clinical applications where extended replication is required.
- Published
- 2016
19. The Polypyrimidine Tract-Binding Protein Is a Transacting Factor for the Dengue Virus Internal Ribosome Entry Site.
- Author
-
Fernández-García L, Angulo J, and López-Lastra M
- Subjects
- Animals, Cell Line, Humans, Virus Replication, Dengue virology, Cricetinae, Protein Biosynthesis, RNA, Messenger genetics, RNA, Messenger metabolism, Internal Ribosome Entry Sites, Polypyrimidine Tract-Binding Protein metabolism, Polypyrimidine Tract-Binding Protein genetics, Dengue Virus genetics, Dengue Virus physiology, RNA, Viral genetics, RNA, Viral metabolism
- Abstract
Dengue virus (DENV) is an enveloped, positive sense, single-stranded RNA virus belonging to the Flaviviridae . Translation initiation of the DENV mRNA (vRNA) can occur following a cap-dependent, 5'-3'end-dependent internal ribosome entry site (IRES)-independent or IRES-dependent mechanism. This study evaluated the activity of DENV IRES in BHK-21 cells and the role of the polypyrimidine-tract binding protein (PTB) isoforms PTB1, PTB2, and PTB4 as IRES-transacting factors (ITAFs) for the DENV IRES. The results show that DENV-IRES activity is stimulated in DENV-replicating BHK-21 cells and cells expressing the Foot-and-mouth disease virus leader or Human rhinovirus 2A proteases. Protease activity was necessary, although a complete shutdown of cap-dependent translation initiation was not a requirement to stimulate DENV IRES activity. Regarding PTB, the results show that PTB1 > PTB2 > PTB4 stimulates DENV-IRES activity in BHK-21 cells. Mutations in the PTB RNA recognition motifs (RRMs), RRM1/RRM2 or RRM3/RRM4, differentially impact PTB1, PTB2, and PTB4's ability to promote DENV IRES-mediated translation initiation in BHK-21 cells. PTB1-induced DENV-IRES stimulation is rescinded when RRM1/RRM2 or RRM3/RRM4 are disrupted. Mutations in RRM1/RRM2 or RRM3/RRM4 do not affect the ITAF activity of PTB2. Mutating RRM3/RRM4, but not RRM1/RRM2, abolishes the ability of PTB4 to stimulate the DENV IRES. Thus, PTB1, PTB2, and PTB4 are ITAFs for the DENV IRES.
- Published
- 2024
- Full Text
- View/download PDF
20. VP3 protein of Senecavirus A promotes viral IRES-driven translation and attenuates innate immunity by specifically relocalizing hnRNPA2B1.
- Author
-
Li L, Li X, Zhong H, Li M, Wan B, He W, Zhang Y, Du Y, Chen D, Zhang W, Ji P, Jiang D, and Han S
- Subjects
- Humans, Host-Pathogen Interactions immunology, HEK293 Cells, Virus Replication, Immune Evasion, Picornaviridae Infections immunology, Picornaviridae Infections virology, Picornaviridae Infections metabolism, Cell Line, Immunity, Innate, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Internal Ribosome Entry Sites, Protein Biosynthesis, Picornaviridae immunology
- Abstract
Viruses deploy sophisticated strategies to hijack the host's translation machinery to favor viral protein synthesis and counteract innate cellular defenses. However, little is known about the mechanisms by which Senecavirus A (SVA) controls the host's translation. Using a series of sophisticated molecular cell manipulation techniques, heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) was identified as an essential host factor involved in translation control in SVA-infected cells. It was also determined that the SVA structural protein, VP3, binds to and relocalizes hnRNPA2B1, which interferes with the host's protein synthesis machinery to establish a cellular environment that facilitates viral propagation via a two-pronged strategy: first, hnRNPA2B1 serves as a potent internal ribosome entry site (IRES) trans -acting factor, which is selectively co-opted to promote viral IRES-driven translation by supporting the assembly of translation initiation complexes. Second, a strong repression of host cell translation occurs in the context of the VP3-hnRNPA2B1 interaction, resulting in attenuation of the interferons response. This is the first study to demonstrate the interaction between SVA VP3 and hnRNPA2B1, and to characterize their key roles in manipulating translation. This novel dual mechanism, which regulates selective mRNA translation and immune evasion of virus-infected cells, highlights the VP3-hnRNPA2B1 complex as a potential target for the development of modified antiviral or oncolytic reagents., Importance: Viral reproduction is contingent on viral protein synthesis, which relies entirely on the host's translation machinery. As such, viruses often need to control the cellular translational apparatus to favor viral protein production and avoid host innate defenses. Senecavirus A (SVA) is an important virus, both as an emerging pathogen in the pork industry and as a potential oncolytic virus for neuroendocrine cancers. Here, heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) was identified as a critical regulator of the translational landscape during SVA infection. This study supports a model whereby the VP3 protein of SVA efficiently subverts the host's protein synthesis machinery through its ability to bind to and relocalize hnRNPA2B1, not only selectively promoting viral internal ribosome entry site-driven translation but also resulting in global translation shutdown and immune evasion. Together, these data provide new insights into how the complex interactions between translation machinery, SVA, and innate immunity contribute to the pathogenicity of the SVA., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
21. Diversity of Reporter Expression Patterns in Transgenic Mouse Lines Targeting Corticotropin-Releasing Hormone-Expressing Neurons
- Author
-
Chen, Yuncai, Molet, Jenny, Gunn, Benjamin G, Ressler, Kerry, and Baram, Tallie Z
- Subjects
Biotechnology ,Mental Health ,Neurosciences ,Underpinning research ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Aetiology ,Amygdala ,Animals ,Brain ,Corticotropin-Releasing Hormone ,Genes ,Reporter ,Green Fluorescent Proteins ,Hippocampus ,Hypothalamus ,Internal Ribosome Entry Sites ,Male ,Mice ,Mice ,Transgenic ,Neurons ,Paraventricular Hypothalamic Nucleus ,RNA ,Messenger ,Septal Nuclei ,Transcriptome ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Endocrinology & Metabolism - Abstract
Transgenic mice, including lines targeting corticotropin-releasing factor (CRF or CRH), have been extensively employed to study stress neurobiology. These powerful tools are poised to revolutionize our understanding of the localization and connectivity of CRH-expressing neurons, and the crucial roles of CRH in normal and pathological conditions. Accurate interpretation of studies using cell type-specific transgenic mice vitally depends on congruence between expression of the endogenous peptide and reporter. If reporter expression does not faithfully reproduce native gene expression, then effects of manipulating unintentionally targeted cells may be misattributed. Here, we studied CRH and reporter expression patterns in 3 adult transgenic mice: Crh-IRES-Cre;Ai14 (tdTomato mouse), Crfp3.0CreGFP, and Crh-GFP BAC. We employed the CRH antiserum generated by Vale after validating its specificity using CRH-null mice. We focused the analyses on stress-salient regions, including hypothalamus, amygdala, bed nucleus of the stria terminalis, and hippocampus. Expression patterns of endogenous CRH were consistent among wild-type and transgenic mice. In tdTomato mice, most CRH-expressing neurons coexpressed the reporter, yet the reporter identified a few non-CRH-expressing pyramidal-like cells in hippocampal CA1 and CA3. In Crfp3.0CreGFP mice, coexpression of CRH and the reporter was found in central amygdala and, less commonly, in other evaluated regions. In Crh-GFP BAC mice, the large majority of neurons expressed either CRH or reporter, with little overlap. These data highlight significant diversity in concordant expression of reporter and endogenous CRH among 3 available transgenic mice. These findings should be instrumental in interpreting important scientific findings emerging from the use of these potent neurobiological tools.
- Published
- 2015
22. The 5′-untranslated region of p16 INK4 a melanoma tumor suppressor acts as a cellular IRES, controlling mRNA translation under hypoxia through YBX1 binding
- Author
-
Bisio, Alessandra, Latorre, Elisa, Andreotti, Virginia, Paillerets, Brigitte Bressac-de, Harland, Mark, Scarra, Giovanna Bianchi, Ghiorzo, Paola, Spitale, Robert C, Provenzani, Alessandro, and Inga, Alberto
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Genetics ,Cancer ,5' Untranslated Regions ,Blotting ,Western ,Cell Hypoxia ,Cell Line ,Tumor ,Cyclin-Dependent Kinase Inhibitor p16 ,Gene Expression Regulation ,Neoplastic ,HCT116 Cells ,Humans ,Internal Ribosome Entry Sites ,MCF-7 Cells ,Melanoma ,Mutation ,Naphthyridines ,Protein Binding ,Protein Biosynthesis ,RNA Interference ,RNA ,Messenger ,Sirolimus ,TOR Serine-Threonine Kinases ,Y-Box-Binding Protein 1 ,IRES ,YBX1 ,hypoxia ,melanoma ,p16INK4a ,Oncology and carcinogenesis - Abstract
CDKN2A/p16INK4a is an essential tumor suppressor gene that controls cell cycle progression and replicative senescence. It is also the main melanoma susceptibility gene. Here we report that p16INK4a 5'UTR mRNA acts as a cellular Internal Ribosome Entry Site (IRES). The potential for p16INK4a 5'UTR to drive cap-independent translation was evaluated by dual-luciferase assays using bicistronic and monocistronic vectors. Results of reporters' relative activities coupled to control analyses for actual bicistronic mRNA transcription, indicated that the wild type p16INK4a 5'UTR could stimulate cap-independent translation. Notably, hypoxic stress and the treatment with mTOR inhibitors enhanced the translation-stimulating property of p16INK4a 5'UTR. RNA immunoprecipitation performed in melanoma-derived SK-Mel-28 and in a patient-derived lymphoblastoid cell line indicated that YBX1 can bind the wild type p16INK4a mRNA increasing its translation efficiency, particularly during hypoxic stress. Modulation of YBX1 expression further supported its involvement in cap-independent translation of the wild type p16INK4a but not a c.-42T>A variant. RNA SHAPE assays revealed local flexibility changes for the c.-42T>A variant at the predicted YBX1 binding site region. Our results indicate that p16INK4a 5'UTR contains a cellular IRES that can enhance mRNA translation efficiency, in part through YBX1.
- Published
- 2015
23. The 5'-untranslated region of p16INK4a melanoma tumor suppressor acts as a cellular IRES, controlling mRNA translation under hypoxia through YBX1 binding.
- Author
-
Bisio, Alessandra, Latorre, Elisa, Andreotti, Virginia, Bressac-de Paillerets, Brigitte, Harland, Mark, Scarra, Giovanna Bianchi, Ghiorzo, Paola, Spitale, Robert C, Provenzani, Alessandro, and Inga, Alberto
- Subjects
Cell Line ,Tumor ,HCT116 Cells ,Humans ,Melanoma ,Sirolimus ,Naphthyridines ,RNA ,Messenger ,5' Untranslated Regions ,Blotting ,Western ,Cell Hypoxia ,Protein Biosynthesis ,Gene Expression Regulation ,Neoplastic ,RNA Interference ,Protein Binding ,Mutation ,Cyclin-Dependent Kinase Inhibitor p16 ,Y-Box-Binding Protein 1 ,TOR Serine-Threonine Kinases ,MCF-7 Cells ,Internal Ribosome Entry Sites ,IRES ,YBX1 ,hypoxia ,melanoma ,p16INK4a ,Oncology and Carcinogenesis - Abstract
CDKN2A/p16INK4a is an essential tumor suppressor gene that controls cell cycle progression and replicative senescence. It is also the main melanoma susceptibility gene. Here we report that p16INK4a 5'UTR mRNA acts as a cellular Internal Ribosome Entry Site (IRES). The potential for p16INK4a 5'UTR to drive cap-independent translation was evaluated by dual-luciferase assays using bicistronic and monocistronic vectors. Results of reporters' relative activities coupled to control analyses for actual bicistronic mRNA transcription, indicated that the wild type p16INK4a 5'UTR could stimulate cap-independent translation. Notably, hypoxic stress and the treatment with mTOR inhibitors enhanced the translation-stimulating property of p16INK4a 5'UTR. RNA immunoprecipitation performed in melanoma-derived SK-Mel-28 and in a patient-derived lymphoblastoid cell line indicated that YBX1 can bind the wild type p16INK4a mRNA increasing its translation efficiency, particularly during hypoxic stress. Modulation of YBX1 expression further supported its involvement in cap-independent translation of the wild type p16INK4a but not a c.-42T>A variant. RNA SHAPE assays revealed local flexibility changes for the c.-42T>A variant at the predicted YBX1 binding site region. Our results indicate that p16INK4a 5'UTR contains a cellular IRES that can enhance mRNA translation efficiency, in part through YBX1.
- Published
- 2015
24. Internal Ribosome Entry Site (IRES)-Mediated Translation and Its Potential for Novel mRNA-Based Therapy Development
- Author
-
Rita Marques, Rafaela Lacerda, and Luísa Romão
- Subjects
RNA-based therapies ,internal ribosome entry sites ,IRES trans-acting factors ,antisense oligonucleotides ,IRES-based multicistronic vectors ,Biology (General) ,QH301-705.5 - Abstract
Many conditions can benefit from RNA-based therapies, namely, those targeting internal ribosome entry sites (IRESs) and their regulatory proteins, the IRES trans-acting factors (ITAFs). IRES-mediated translation is an alternative mechanism of translation initiation, known for maintaining protein synthesis when canonical translation is impaired. During a stress response, it contributes to cell reprogramming and adaptation to the new environment. The relationship between IRESs and ITAFs with tumorigenesis and resistance to therapy has been studied in recent years, proposing new therapeutic targets and treatments. In addition, IRES-dependent translation initiation dysregulation is also related to neurological and cardiovascular diseases, muscular atrophies, or other syndromes. The participation of these structures in the development of such pathologies has been studied, yet to a far lesser extent than in cancer. Strategies involving the disruption of IRES–ITAF interactions or the modification of ITAF expression levels may be used with great impact in the development of new therapeutics. In this review, we aim to comprehend the current data on groups of human pathologies associated with IRES and/or ITAF dysregulation and their application in the designing of new therapeutic approaches using them as targets or tools. Thus, we wish to summarise the evidence in the field hoping to open new promising lines of investigation toward personalised treatments.
- Published
- 2022
- Full Text
- View/download PDF
25. A cis-acting ligase ribozyme generates circular RNA in vitro for ectopic protein functioning.
- Author
-
Su CI, Chuang ZS, Shie CT, Wang HI, Kao YT, and Yu CY
- Subjects
- Humans, Internal Ribosome Entry Sites, Protein Biosynthesis, RNA metabolism, RNA genetics, HEK293 Cells, Exoribonucleases, RNA, Circular metabolism, RNA, Circular genetics, RNA, Catalytic metabolism, RNA, Catalytic genetics
- Abstract
Delivering synthetic protein-coding RNA bypassing the DNA stage for ectopic protein functioning is a novel therapeutic strategy. Joining the linear RNA head-to-tail covalently could be a state-of-the-art strategy for functioning longer. Here we enroll a cis-acting ligase ribozyme (RzL) to generate circular RNA (circRNA) in vitro for ectopic protein expression. The RNA circularization is confirmed by masking the 5' phosphate group, resisting exonuclease RNase R digestion, failing for further tailing, and sequencing the RT-PCR products of the joined region. Interestingly, one internal ribosome entry site (IRES) renders circRNA translation competent, but two IRES in cis, not trans, hamper the translation. The circRNA with highly potent in translation is conferred for antiviral functioning. Accompanying specific guided RNA, a circRNA expressing ribonuclease Cas13 shows excellent potential against the corresponding RNA virus, further extending circRNA functioning in its growing list of applications., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
26. DeepIRES: a hybrid deep learning model for accurate identification of internal ribosome entry sites in cellular and viral mRNAs.
- Author
-
Zhao J, Chen Z, Zhang M, Zou L, He S, Liu J, Wang Q, Song X, and Wu J
- Subjects
- Computational Biology methods, RNA, Viral genetics, RNA, Viral metabolism, Humans, Neural Networks, Computer, Algorithms, Deep Learning, Internal Ribosome Entry Sites, RNA, Messenger genetics, RNA, Messenger metabolism
- Abstract
The internal ribosome entry site (IRES) is a cis-regulatory element that can initiate translation in a cap-independent manner. It is often related to cellular processes and many diseases. Thus, identifying the IRES is important for understanding its mechanism and finding potential therapeutic strategies for relevant diseases since identifying IRES elements by experimental method is time-consuming and laborious. Many bioinformatics tools have been developed to predict IRES, but all these tools are based on structure similarity or machine learning algorithms. Here, we introduced a deep learning model named DeepIRES for precisely identifying IRES elements in messenger RNA (mRNA) sequences. DeepIRES is a hybrid model incorporating dilated 1D convolutional neural network blocks, bidirectional gated recurrent units, and self-attention module. Tenfold cross-validation results suggest that DeepIRES can capture deeper relationships between sequence features and prediction results than other baseline models. Further comparison on independent test sets illustrates that DeepIRES has superior and robust prediction capability than other existing methods. Moreover, DeepIRES achieves high accuracy in predicting experimental validated IRESs that are collected in recent studies. With the application of a deep learning interpretable analysis, we discover some potential consensus motifs that are related to IRES activities. In summary, DeepIRES is a reliable tool for IRES prediction and gives insights into the mechanism of IRES elements., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
27. Molecular mechanisms of circular RNA translation.
- Author
-
Hwang HJ and Kim YK
- Subjects
- Humans, Animals, RNA genetics, RNA metabolism, Internal Ribosome Entry Sites, RNA, Messenger genetics, RNA, Messenger metabolism, RNA Stability, RNA Splicing, RNA, Circular genetics, Protein Biosynthesis
- Abstract
Circular RNAs (circRNAs) are covalently closed single-stranded RNAs without a 5' cap structure and a 3' poly(A) tail typically present in linear mRNAs of eukaryotic cells. CircRNAs are predominantly generated through a back-splicing process within the nucleus. CircRNAs have long been considered non-coding RNAs seemingly devoid of protein-coding potential. However, many recent studies have challenged this idea and have provided substantial evidence that a subset of circRNAs can associate with polysomes and indeed be translated. Therefore, in this review, we primarily highlight the 5' cap-independent internal initiation of translation that occurs on circular RNAs. Several molecular features of circRNAs, including the internal ribosome entry site, N
6 -methyladenosine modification, and the exon junction complex deposited around the back-splicing junction after back-splicing event, play pivotal roles in their efficient internal translation. We also propose a possible relationship between the translatability of circRNAs and their stability, with a focus on nonsense-mediated mRNA decay and nonstop decay, both of which are well-characterized mRNA surveillance mechanisms. An in-depth understanding of circRNA translation will reshape and expand our current knowledge of proteomics., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
28. Internal Ribosome Entry Sites Mediate Cap-Independent Translation of Bmi1 in Nasopharyngeal Carcinoma
- Author
-
Hongbo Wang, Yunjia Zhu, Lijuan Hu, Yangyang Li, Guihong Liu, Tianliang Xia, Dan Xiong, Yiling Luo, Binliu Liu, Yu An, Manzhi Li, Yuehua Huang, Qian Zhong, and Musheng Zeng
- Subjects
internal ribosome entry sites ,Bmi1 ,nasopharyngeal carcinoma ,5′-untranslated region ,translational activity ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Bmi1 is overexpressed in multiple human cancers. We previously reported the oncogenic function and the transcription regulation mechanisms of Bmi1 in nasopharyngeal carcinoma (NPC). In this study, we observed that the mRNA and the protein levels of Bmi1 were strictly inconsistent in NPC cell lines and cancer tissues. The inhibitors of proteasome and lysosome could not enhance the protein level of Bmi1, indicating that Bmi1 may be post-transcriptionally regulated. The IRESite analysis showed that there were two potential internal ribosome entry sites (IRESs) in the 5′-untranslated region (5′-UTR) of Bmi1. The luciferase assay demonstrated that the 5′-UTR of Bmi1 has IRES activity, which may mediate cap-independent translation. The IRES activity of the Bmi1 5′-UTR was significantly reduced after the mutation of the two IRES elements. Taken together, these results suggested that the IRES elements mediating translation is a novel post-transcriptional regulation mechanism of Bmi1.
- Published
- 2020
- Full Text
- View/download PDF
29. Consensus small interfering RNA targeted to stem-loops II and III of IRES structure of 5′ UTR effectively inhibits virus replication and translation of HCV sub-genotype 4a isolates from Saudi Arabia.
- Author
-
AlMalki, Waleed H., Shahid, Imran, Abdalla, Ashraf N., Johargy, Ayman K., Ahmed, Muhammad, and Hassan, Sajida
- Abstract
Being the most conserved region of all hepatitis C virus (HCV) genotypes and sub-genotypes, the 5′ untranslated region (5′ UTR) of HCV genome signifies it's importance as a potential target for anti-mRNA based treatment strategies like RNA interference. The advent and approval of first small interference RNA (siRNA) -based treatment of hereditary transthyretin-mediated amyloidosis for clinical use has raised the hopes to test this approach against highly susceptible viruses like HCV. We investigated the antiviral potential of consensus siRNAs targeted to stem-loops (SLs) II and III nucleotide motifs of internal ribosome entry site (IRES) structure within 5′ UTR of HCV sub-genotype 4a isolates from the Saudi population. siRNA inhibitory effects on viral replication and translation of full-length HCV genome were determined in a competent, persistent, and reproducible Huh-7 cell culture system maintained for one month. Maximal inhibition of RNA transcript levels of HCV-IRES clones and silencing of viral replication and translation of full-length virus genome was demonstrated by siRNAs targeted to SL-III nucleotide motifs of IRES in Huh-7 cells. siRNA Usi-169 decreased 5′ UTR RNA transcript levels of HCV-IRES clones up to 75% (P < 0.001) at 24 h post-transfection and 80% (P < 0.001) at 48 h treatment in Huh-7 cells. 5′ UTR-tagged GFP protein expression was significantly decreased from 70 to 80% in Huh-7 cells co-transfected with constructed vectors (i.e. pCR3.1/GFP/5′ UTR) and siRNA Usi-169 at 24 h and 48 h time-span. Viral replication was inhibited by more than 90% (P < 0.001) and HCV core (C) and hypervariable envelope glycoproteins (E1 and E2) expression was also significantly degraded by intracytoplasmic siRNA Usi-169 activity in persistent Huh-7 cell culture system. The findings unveil that siRNAs targeted to 5′ UTR-IRES of HCV sub-genotype 4a Saudi isolates show potent silencing of HCV replication and blocking of viral translation in a persistent in-vitro Huh-7 tissue culture system. Furthermore, we also elucidated that siRNA silencing of viral mRNA not only inhibits viral replication but also blocks viral translation. The results suggest that siRNA potent antiviral activity should be considered as an effective anti-mRNA based treatment strategies for further in-vivo investigations against less studied and harder-to-treat HCV sub-genotype 4a isolates in Saudi Arabia. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
30. Internal Ribosome Entry Sites Mediate Cap-Independent Translation of Bmi1 in Nasopharyngeal Carcinoma.
- Author
-
Wang, Hongbo, Zhu, Yunjia, Hu, Lijuan, Li, Yangyang, Liu, Guihong, Xia, Tianliang, Xiong, Dan, Luo, Yiling, Liu, Binliu, An, Yu, Li, Manzhi, Huang, Yuehua, Zhong, Qian, and Zeng, Musheng
- Subjects
CARCINOMA ,TRANSLATIONS ,PROTEASOME inhibitors ,CELL lines - Abstract
Bmi1 is overexpressed in multiple human cancers. We previously reported the oncogenic function and the transcription regulation mechanisms of Bmi1 in nasopharyngeal carcinoma (NPC). In this study, we observed that the mRNA and the protein levels of Bmi1 were strictly inconsistent in NPC cell lines and cancer tissues. The inhibitors of proteasome and lysosome could not enhance the protein level of Bmi1, indicating that Bmi1 may be post-transcriptionally regulated. The IRESite analysis showed that there were two potential internal ribosome entry sites (IRESs) in the 5′-untranslated region (5′-UTR) of Bmi1. The luciferase assay demonstrated that the 5′-UTR of Bmi1 has IRES activity, which may mediate cap-independent translation. The IRES activity of the Bmi1 5′-UTR was significantly reduced after the mutation of the two IRES elements. Taken together, these results suggested that the IRES elements mediating translation is a novel post-transcriptional regulation mechanism of Bmi1. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. HSP70 positively regulates translation by interacting with the IRES and stabilizes the viral structural proteins VP1 and VP3 to facilitate duck hepatitis A virus type 1 replication.
- Author
-
Jiang Y, Xu C, Cheng A, Wang M, Zhang W, Zhao X, Yang Q, Wu Y, Zhang S, Tian B, Huang J, Ou X, Sun D, He Y, Wu Z, Zhu D, Jia R, Chen S, and Liu M
- Subjects
- Animals, Viral Structural Proteins metabolism, Viral Structural Proteins genetics, Ducks, Poultry Diseases virology, Picornaviridae Infections veterinary, Picornaviridae Infections virology, Picornaviridae Infections metabolism, Capsid Proteins metabolism, Capsid Proteins genetics, Hepatitis, Viral, Animal virology, Hepatitis, Viral, Animal metabolism, Protein Biosynthesis, Hepatitis Virus, Duck physiology, Hepatitis Virus, Duck genetics, HSP70 Heat-Shock Proteins metabolism, HSP70 Heat-Shock Proteins genetics, Virus Replication, Internal Ribosome Entry Sites
- Abstract
The maintenance of viral protein homeostasis depends on the interaction between host cell proteins and viral proteins. As a molecular chaperone, heat shock protein 70 (HSP70) has been shown to play an important role in viral infection. Our results showed that HSP70 can affect translation, replication, assembly, and release during the life cycle of duck hepatitis A virus type 1 (DHAV-1). We demonstrated that HSP70 can regulate viral translation by interacting with the DHAV-1 internal ribosome entry site (IRES). In addition, HSP70 interacts with the viral capsid proteins VP1 and VP3 and promotes their stability by inhibiting proteasomal degradation, thereby facilitating the assembly of DHAV-1 virions. This study demonstrates the specific role of HSP70 in regulating DHAV-1 replication, which are helpful for understanding the pathogenesis of DHAV-1 infection and provide additional information about the role of HSP70 in infection by different kinds of picornaviruses, as well as the interaction between picornaviruses and host cells., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
32. PSPC1 Binds to HCV IRES and Prevents Ribosomal Protein S5 Binding, Inhibiting Viral RNA Translation.
- Author
-
Tripathi SK, Aneja A, Borgaonkar T, and Das S
- Subjects
- Humans, Hepatitis C virology, Hepatitis C metabolism, Host-Pathogen Interactions, Protein Binding, Virus Replication, Hepacivirus genetics, Hepacivirus physiology, Internal Ribosome Entry Sites, Protein Biosynthesis, Ribosomal Proteins metabolism, RNA, Viral metabolism, RNA, Viral genetics, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics
- Abstract
Hepatitis C virus (HCV) infects the human liver, and its chronic infection is one of the major causes of Hepatocellular carcinoma. Translation of HCV RNA is mediated by an Internal Ribosome Entry Site (IRES) element located in the 5'UTR of viral RNA. Several RNA Binding proteins of the host interact with the HCV IRES and modulate its function. Here, we demonstrate that PSPC1 (Paraspeckle Component 1), an essential paraspeckle component, upon HCV infection is relocalized and interacts with HCV IRES to prevent viral RNA translation. Competition UV-crosslinking experiments showed that PSPC1 interacts explicitly with the SLIV region of the HCV IRES, which is known to play a vital role in ribosomal loading to the HCV IRES via interaction with Ribosomal protein S5 (RPS5). Partial silencing of PSPC1 increased viral RNA translation and, consequently, HCV replication, suggesting a negative regulation by PSPC1. Interestingly, the silencing of PSPC1 protein leads to an increased interaction of RPS5 at the SLIV region, leading to an overall increase in the viral RNA in polysomes. Overall, our results showed how the host counters viral infection by relocalizing nuclear protein to the cytoplasm as a survival strategy.
- Published
- 2024
- Full Text
- View/download PDF
33. TGF-β2 Induces Ribosome Activity, Alters Ribosome Composition and Inhibits IRES-Mediated Translation in Chondrocytes.
- Author
-
van den Akker GGH, Chabronova A, Housmans BAC, van der Vloet L, Surtel DAM, Cremers A, Marchand V, Motorin Y, Caron MMJ, Peffers MJ, and Welting TJM
- Subjects
- Humans, Internal Ribosome Entry Sites, Cell Line, Chondrocytes metabolism, Chondrocytes drug effects, Ribosomes metabolism, Protein Biosynthesis, RNA, Ribosomal metabolism, RNA, Ribosomal genetics, Transforming Growth Factor beta2 metabolism, Transforming Growth Factor beta2 pharmacology
- Abstract
Alterations in cell fate are often attributed to (epigenetic) regulation of gene expression. An emerging paradigm focuses on specialized ribosomes within a cell. However, little evidence exists for the dynamic regulation of ribosome composition and function. Here, we stimulated a chondrocytic cell line with transforming growth factor beta (TGF-β2) and mapped changes in ribosome function, composition and ribosomal RNA (rRNA) epitranscriptomics. 35S Met/Cys incorporation was used to evaluate ribosome activity. Dual luciferase reporter assays were used to assess ribosomal modus. Ribosomal RNA expression and processing were determined by RT-qPCR, while RiboMethSeq and HydraPsiSeq were used to determine rRNA modification profiles. Label-free protein quantification of total cell lysates, isolated ribosomes and secreted proteins was done by LC-MS/MS. A three-day TGF-β2 stimulation induced total protein synthesis in SW1353 chondrocytic cells and human articular chondrocytes. Specifically, TGF-β2 induced cap-mediated protein synthesis, while IRES-mediated translation was not (P53 IRES) or little affected (CrPv IGR and HCV IRES). Three rRNA post-transcriptional modifications (PTMs) were affected by TGF-β2 stimulation (18S-Gm1447 downregulated, 18S-ψ1177 and 28S-ψ4598 upregulated). Proteomic analysis of isolated ribosomes revealed increased interaction with eIF2 and tRNA ligases and decreased association of eIF4A3 and heterogeneous nuclear ribonucleoprotein (HNRNP)s. In addition, thirteen core ribosomal proteins were more present in ribosomes from TGF-β2 stimulated cells, albeit with a modest fold change. A prolonged stimulation of chondrocytic cells with TGF-β2 induced ribosome activity and changed the mode of translation. These functional changes could be coupled to alterations in accessory proteins in the ribosomal proteome.
- Published
- 2024
- Full Text
- View/download PDF
34. Factor-Dependent Internal Ribosome Entry Site and -1 Programmed Frameshifting Signal in the Bemisia-Associated Dicistrovirus 2.
- Author
-
Chen Y, Chapagain S, Chien J, Pereira HS, Patel TR, Inoue-Nagata AK, and Jan E
- Subjects
- Animals, Nucleic Acid Conformation, Protein Biosynthesis, Genome, Viral, Dicistroviridae genetics, Internal Ribosome Entry Sites, Frameshifting, Ribosomal, RNA, Viral genetics, RNA, Viral metabolism, Hemiptera virology, Ribosomes metabolism
- Abstract
The dicistrovirus intergenic (IGR) IRES uses the most streamlined translation initiation mechanism: the IRES recruits ribosomes directly without using protein factors and initiates translation from a non-AUG codon. Several subtypes of dicistroviruses IRES have been identified; typically, the IRESs adopt two -to three overlapping pseudoknots with key stem-loop and unpaired regions that interact with specific domains of the ribosomal 40S and 60S subunits to direct translation. We previously predicted an atypical IGR IRES structure and a potential -1 programmed frameshift (-1 FS) signal within the genome of the whitefly Bemisia-associated dicistrovirus 2 (BaDV-2). Here, using bicistronic reporters, we demonstrate that the predicted BaDV-2 -1 FS signal can drive -1 frameshifting in vitro via a slippery sequence and a downstream stem-loop structure that would direct the translation of the viral RNA-dependent RNA polymerase. Moreover, the predicted BaDV-2 IGR can support IRES translation in vitro but does so through a mechanism that is not typical of known factorless dicistrovirus IGR IRES mechanisms. Using deletion and mutational analyses, the BaDV-2 IGR IRES is mapped within a 140-nucleotide element and initiates translation from an AUG codon. Moreover, the IRES does not bind directly to purified ribosomes and is sensitive to eIF2 and eIF4A inhibitors NSC1198983 and hippuristanol, respectively, indicating an IRES-mediated factor-dependent mechanism. Biophysical characterization suggests the BaDV-2 IGR IRES contains several stem-loops; however, mutational analysis suggests a model whereby the IRES is unstructured or adopts distinct conformations for translation initiation. In summary, we have provided evidence of the first -1 FS frameshifting signal and a novel factor-dependent IRES mechanism in this dicistrovirus family, thus highlighting the diversity of viral RNA-structure strategies to direct viral protein synthesis.
- Published
- 2024
- Full Text
- View/download PDF
35. Hyperglycemia facilitates EV71 replication: Insights into miR-206-mediated regulation of G3BP2 promoting EV71 IRES activity.
- Author
-
Lai RH, Chow YH, Lin YW, Chung NH, Nien SW, and Juang JL
- Subjects
- Animals, Humans, Mice, Disease Models, Animal, Insulin metabolism, Mice, Transgenic, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics, Viral Load, Enterovirus A, Human physiology, Enterovirus A, Human genetics, Enterovirus Infections virology, Enterovirus Infections metabolism, Hyperglycemia metabolism, Hyperglycemia virology, Internal Ribosome Entry Sites, MicroRNAs metabolism, MicroRNAs genetics, Virus Replication
- Abstract
Background: Neurotropic virus infections actively manipulate host cell metabolism to enhance virus neurovirulence. Although hyperglycemia is common during severe infections, its specific role remains unclear. This study investigates the impact of hyperglycemia on the neurovirulence of enterovirus 71 (EV71), a neurovirulent virus relying on internal ribosome entry site (IRES)-mediated translation for replication. Methods: Utilizing hSCARB2-transgenic mice, we explore the effects of hyperglycemia in EV71 infection and elucidate the underlying mechanisms. Results: Remarkably, administering insulin alone to reduce hyperglycemia in hSCARB2-transgenic mice results in a decrease in brainstem encephalitis and viral load. Conversely, induced hyperglycemia exacerbates neuropathogenesis, highlighting the pivotal role of hyperglycemia in neurovirulence. Notably, miR-206 emerges as a crucial mediator induced by viral infection, with its expression further heightened by hyperglycemia and concurrently repressed by insulin. The use of antagomiR-206 effectively mitigates EV71-induced brainstem encephalitis and reduces viral load. Mechanistically, miR-206 facilitates IRES-driven virus replication by repressing the stress granule protein G3BP2. Conclusions: Novel therapeutic approaches against severe EV71 infections involve managing hyperglycemia and targeting the miR-206-stress granule pathway to modulate virus IRES activity., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2024
- Full Text
- View/download PDF
36. Mitochondrial Ribosomal Protein MRPS15 Is a Component of Cytosolic Ribosomes and Regulates Translation in Stressed Cardiomyocytes.
- Author
-
David F, Roussel E, Froment C, Draia-Nicolau T, Pujol F, Burlet-Schiltz O, Henras AK, Lacazette E, Morfoisse F, Tatin F, Diaz JJ, Catez F, Garmy-Susini B, and Prats AC
- Subjects
- Humans, Ribosomes metabolism, Polyribosomes metabolism, Cytosol metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Internal Ribosome Entry Sites, Protein Biosynthesis, Ribosomal Proteins genetics, Ribosomal Proteins metabolism, Myocytes, Cardiac metabolism
- Abstract
Regulation of mRNA translation is a crucial step in controlling gene expression in stressed cells, impacting many pathologies, including heart ischemia. In recent years, ribosome heterogeneity has emerged as a key control mechanism driving the translation of subsets of mRNAs. In this study, we investigated variations in ribosome composition in human cardiomyocytes subjected to endoplasmic reticulum stress induced by tunicamycin treatment. Our findings demonstrate that this stress inhibits global translation in cardiomyocytes while activating internal ribosome entry site (IRES)-dependent translation. Analysis of translating ribosome composition in stressed and unstressed cardiomyocytes was conducted using mass spectrometry. We observed no significant changes in ribosomal protein composition, but several mitochondrial ribosomal proteins (MRPs) were identified in cytosolic polysomes, showing drastic variations between stressed and unstressed cells. The most notable increase in polysomes of stressed cells was observed in MRPS15. Its interaction with ribosomal proteins was confirmed by proximity ligation assay (PLA) and immunoprecipitation, suggesting its intrinsic role as a ribosomal component during stress. Knock-down or overexpression experiments of MRPS15 revealed its role as an activator of IRES-dependent translation. Furthermore, polysome profiling after immunoprecipitation with anti-MRPS15 antibody revealed that the "MRPS15 ribosome" is specialized in translating mRNAs involved in the unfolded protein response.
- Published
- 2024
- Full Text
- View/download PDF
37. Nuclear ribonucleoprotein RALY downregulates foot-and-mouth disease virus replication but antagonized by viral 3C protease.
- Author
-
Wu J, Sun C, Guan J, Abdullah SW, Wang X, Ren M, Qiao L, Sun S, and Guo H
- Subjects
- Animals, Ribonucleoproteins genetics, Ribonucleoproteins metabolism, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, RNA-Binding Proteins genetics, Ribosomes genetics, Endopeptidases metabolism, Internal Ribosome Entry Sites, 3C Viral Proteases, Ubiquitins genetics, Ubiquitins metabolism, Foot-and-Mouth Disease Virus genetics, Foot-and-Mouth Disease Virus metabolism
- Abstract
The internal ribosome entry site (IRES) element constitutes a cis-acting RNA regulatory sequence that recruits the ribosomal initiation complex in a cap-independent manner, assisted by various RNA-binding proteins and IRES trans -acting factors. Foot-and-mouth disease virus (FMDV) contains a functional IRES element and takes advantage of this element to subvert host translation machinery. Our study identified a novel mechanism wherein RALY, a member of the heterogeneous nuclear ribonucleoproteins (hnRNP) family belonging to RNA-binding proteins, binds to the domain 3 of FMDV IRES via its RNA recognition motif residue. This interaction results in the downregulation of FMDV replication by inhibiting IRES-driven translation. Furthermore, our findings reveal that the inhibitory effect exerted by RALY on FMDV replication is not attributed to the FMDV IRES-mediated assembly of translation initiation complexes but rather to the impediment of 80S ribosome complex formation after binding with 40S ribosomes. Conversely, 3C
pro of FMDV counteracts RALY-mediated inhibition by the ubiquitin-proteasome pathway. Therefore, these results indicate that RALY, as a novel critical IRES-binding protein, inhibits FMDV replication by blocking the formation of 80S ribosome, providing a deeper understanding of how viruses recruit and manipulate host factors., Importance: The translation of FMDV genomic RNA driven by IRES element is a crucial step for virus infections. Many host proteins are hijacked to regulate FMDV IRES-dependent translation, but the regulatory mechanism remains unknown. Here, we report for the first time that cellular RALY specifically interacts with the IRES of FMDV and negatively regulates viral replication by blocking 80S ribosome assembly on FMDV IRES. Conversely, RALY-mediated inhibition is antagonized by the viral 3C protease by the ubiquitin-proteasome pathway. These results would facilitate further understanding of virus-host interactions and translational control during viral infection., Competing Interests: The authors declare no conflict of interest.- Published
- 2024
- Full Text
- View/download PDF
38. Circular RNA translation: novel protein isoforms and clinical significance
- Author
-
Shuo-yang Wen, Javeria Qadir, and Burton B. Yang
- Subjects
Gene Expression Regulation ,Protein Biosynthesis ,Humans ,Protein Isoforms ,RNA ,Molecular Medicine ,RNA, Circular ,Internal Ribosome Entry Sites ,Molecular Biology - Abstract
In recent years, significant attention has focused on circular RNA (circRNA) translation to determine its clinical significance. Cap-independent translation of circRNAs driven by an internal ribosome entry site (IRES) or an N6-methyladenosine (m
- Published
- 2022
- Full Text
- View/download PDF
39. Picornavirus translation strategies
- Author
-
Salvador Abellán, Azman Embarc-Buh, Encarna Martinez-Salas, Rosario Francisco-Velilla, and UAM. Departamento de Biología Molecular
- Subjects
Virus Genome ,Messenger RNA ,Viral Protein ,RNA, Viral ,Picornaviridae ,RNA, Messenger ,Ribosomal Subunit ,Virus Gene ,Internal Ribosome Entry Sites ,Biología y Biomedicina / Biología ,Virus Replication ,General Biochemistry, Genetics and Molecular Biology ,Protein Binding - Abstract
The genome of viruses classified as picornaviruses consists of a single monocistronic positive strand RNA. The coding capacity of these RNA viruses is rather limited, and thus, they rely on the cellular machinery for their viral replication cycle. Upon the entry of the virus into susceptible cells, the viral RNA initially competes with cellular mRNAs for access to the protein synthesis machinery. Not surprisingly, picornaviruses have evolved specialized strategies that successfully allow the expression of viral gene products, which we outline in this review. The main feature of all picornavirus genomes is the presence of a heavily structured RNA element on the 5´UTR, referred to as an internal ribosome entry site (IRES) element, which directs viral protein synthesis as well and, consequently, triggers the subsequent steps required for viral replication. Here, we will summarize recent studies showing that picornavirus IRES elements consist of a modular structure, providing sites of interaction for ribosome subunits, eIFs, and a selective group of RNA-binding proteins, This work was supported by grants PID2020-115096RB-I00 (MICIN), B2017/BMD-3770 (cofinanced by Autonomous Community of Madrid and FEDER funds), and an Institutional grant from Fundación Ramón Areces
- Published
- 2022
- Full Text
- View/download PDF
40. Horizontal gene transfer as a mechanism for the promiscuous acquisition of distinct classes of IRES by avian caliciviruses
- Author
-
Yani Arhab, Anna Miścicka, Tatyana V Pestova, and Christopher U T Hellen
- Subjects
Gene Transfer, Horizontal ,AcademicSubjects/SCI00010 ,Protein Biosynthesis ,viruses ,RNA and RNA-protein complexes ,Genetics ,virus diseases ,Nucleic Acid Conformation ,RNA, Viral ,Internal Ribosome Entry Sites ,Caliciviridae ,Ribosomes - Abstract
In contrast to members of Picornaviridae which have long 5′-untranslated regions (5′UTRs) containing internal ribosomal entry sites (IRESs) that form five distinct classes, members of Caliciviridae typically have short 5′UTRs and initiation of translation on them is mediated by interaction of the viral 5′-terminal genome-linked protein (VPg) with subunits of eIF4F rather than by an IRES. The recent description of calicivirus genomes with 500–900nt long 5′UTRs was therefore unexpected and prompted us to examine them in detail. Sequence analysis and structural modelling of the atypically long 5′UTRs of Caliciviridae sp. isolate yc-13 and six other caliciviruses suggested that they contain picornavirus-like type 2 IRESs, whereas ruddy turnstone calicivirus (RTCV) and Caliciviridae sp. isolate hwf182cal1 calicivirus contain type 4 and type 5 IRESs, respectively. The suggestion that initiation on RTCV mRNA occurs by the type 4 IRES mechanism was confirmed experimentally using in vitro reconstitution. The high sequence identity between identified calicivirus IRESs and specific picornavirus IRESs suggests a common evolutionary origin. These calicivirus IRESs occur in a single phylogenetic branch of Caliciviridae and were likely acquired by horizontal gene transfer.
- Published
- 2021
- Full Text
- View/download PDF
41. Identification of an IRES within the coding region of the structural protein of human rhinovirus 16
- Author
-
Bingtian Shi, Qinqin Song, Xiaonuan Luo, Juan Song, Dong Xia, Zhiqiang Xia, Mi Liu, Wenjun Wang, Ruifang Wang, Haijun Du, Qiang Wei, and Jun Han
- Subjects
Infectious Diseases ,Rhinovirus ,Protein Biosynthesis ,Virology ,Humans ,Internal Ribosome Entry Sites ,5' Untranslated Regions ,Ribosomes - Abstract
As an alternative mechanism for cap-dependent (m7GpppN) translation, internal ribosome entry site (IRES)-dependent translation has been observed in the 5' untranslated regions (5' UTR) and coding regions of a number of viral and eukaryotic mRNAs. In this study, a series of 5' terminal truncated structural protein genes that were fused with GFP was used to screen for potential IRESs, and IRESs were identified using a bicistronic luciferase vector or GFP expression vector possessing a hairpin structure. Our results revealed that a putative IRES was located between nt 1982 and 2281 in the VP3 coding region of the human rhinovirus 16 (HRV16) genomes. We also demonstrated that effective IRES-initiated protein expression in vitro did not occur through splicing sites or cryptic promoters. We confirmed that thapsigargin (TG), an inducer of endoplasmic reticulum stress (ERS), facilitated increased IRES activity in a dose-dependent manner. Additionally, the secondary structure of the IRES was predicted online using the RNAfold web server.
- Published
- 2021
- Full Text
- View/download PDF
42. Differential bicistronic gene translation mediated by the internal ribosome entry site element of encephalomyocarditis virus
- Author
-
Chao-Lin Liu, Chia-Rui Shen, Ya-Shan Chen, Hsi-Jien Chen, and Yih-Shiou Hwang
- Subjects
Reporter gene ,Translational efficiency ,viruses ,Upstream and downstream (transduction) ,fungi ,Translation (biology) ,General Medicine ,Internal Ribosome Entry Sites ,Biology ,Cell biology ,Mice ,Internal ribosome entry site ,Cistron ,Genes, Reporter ,Gene expression ,Protein biosynthesis ,Animals ,Encephalomyocarditis virus ,Peptide Chain Initiation, Translational ,Ribosomes - Abstract
Background Internal ribosome entry sites (IRESs) allow the translation of a transcript independent of its cap structure. They are distributed in some viruses and cellular RNA. The element is applied in dual gene expression in a single vector. Although it appears the lower efficiency of IRES-mediated translation than that of cap-dependent translation, it is with the crucial needs to know the precise differences in translational efficacy between upstream cistrons (cap-dependent) and downstream cistrons (IRES-mediate, cap-independent) before applying the bicistronic vector in biomedical applications. Material and methods This study aimed to provide real examples and showed the precise differences for translational efficiency dependent upon target gene locations. We generated various bicistronic constructs with quantifiable reporter genes as upstream and downstream cistrons of the encephalomyocarditis virus (EMCV) IRES to precisely evaluate the efficacy of IRES-mediated translation in mammalian cells. Results There was no significant difference in protein production when the reporter gene was cloned as an upstream cistron. However, lower levels of protein production were obtained when the reporter gene was located downstream of the IRES. Moreover, in the presence of an upstream cistron, a markedly reduced level of protein production was observed. Conclusion Our findings demonstrate the version of the EMCV IRES that is provided in many commercial vectors is relatively less efficient than cap-dependent translation and provide valuable information regarding the utilization of IRES to facilitate the expression of more than one protein from a transcript.
- Published
- 2021
- Full Text
- View/download PDF
43. Multiple Viral Protein Genome-Linked Proteins Compensate for Viral Translation in a Positive-Sense Single-Stranded RNA Virus Infection
- Author
-
Reid Warsaba, Nikolay Stoynov, Kyung-Mee Moon, Stephane Flibotte, Leonard Foster, and Eric Jan
- Subjects
Threonine ,Immunology ,Genome, Viral ,Internal Ribosome Entry Sites ,Viral Load ,Microbiology ,Cell Line ,Genome Replication and Regulation of Viral Gene Expression ,Viral Proteins ,RNA Virus Infections ,Virology ,Insect Science ,Protein Biosynthesis ,Mutation ,Dicistroviridae ,Serine ,Animals ,RNA, Viral ,Drosophila ,5' Untranslated Regions - Abstract
Viral protein genome-linked (VPg) protein plays an essential role in protein-primed replication of plus-stranded RNA viruses. VPg is covalently linked to the 5′ end of the viral RNA genome via a phosphodiester bond typically at a conserved amino acid. Whereas most viruses have a single VPg, some viruses have multiple VPgs that are proposed to have redundant yet undefined roles in viral replication. Here, we use cricket paralysis virus (CrPV), a dicistrovirus that has four nonidentical copies of VPg, as a model to characterize the role of VPg copies in infection. Dicistroviruses contain two main open reading frames (ORFs) that are driven by distinct internal ribosome entry sites (IRESs). We systematically generated single and combinatorial deletions and mutations of VPg1 to VPg4 within the CrPV infectious clone and monitored viral yield in Drosophila S2 cells. Deletion of one to three VPg copies progressively decreased viral yield and delayed viral replication, suggesting a threshold number of VPgs for productive infection. Mass spectrometry analysis of CrPV VPg-linked RNAs revealed viral RNA linkage to either a serine or threonine in VPg, mutations of which in all VPgs attenuated infection. Mutating serine 4 in a single VPg abolished viral infection, indicating a dominant negative effect. Using viral minigenome reporters that monitor dicistrovirus 5′ untranslated (UTR) and IRES translation revealed a relationship between VPg copy number and the ratio of distinct IRES translation activities. We uncovered a novel viral strategy whereby VPg copies in dicistrovirus genomes compensate for the relative IRES translation efficiencies to promote infection. IMPORTANCE Genetic duplication is exceedingly rare in small RNA viral genomes, as there is selective pressure to prevent RNA genomes from expanding. However, some small RNA viruses encode multiple copies of a viral protein, most notably an unusual viral protein that is linked to the viral RNA genome. Here, we investigate a family of viruses that contains multiple viral protein genome-linked proteins and reveal a novel viral strategy whereby viral protein copy number counterbalances differences in viral protein synthesis mechanisms.
- Published
- 2023
44. Validating the EMCV IRES Secondary Structure with Structure-Function Analysis.
- Author
-
Maloney A and Joseph S
- Subjects
- Base Sequence, Protein Biosynthesis, RNA, Viral genetics, RNA, Viral metabolism, Internal Ribosome Entry Sites, Encephalomyocarditis virus genetics, Encephalomyocarditis virus metabolism
- Abstract
The encephalomyocarditis virus internal ribosome entry site (EMCV IRES) is a structured RNA sequence found in the 5' UTR of the genomic RNA of the encephalomyocarditis virus. The EMCV IRES structure facilitates efficient translation initiation without needing a 5' m
7 G cap or the cap-binding protein eIF4E. The secondary structure of IRES has been the subject of several previous studies, and a number of different structural models have been proposed. Though some domains of the IRES are conserved across the different secondary structure models, domain I of the IRES varies greatly across them. A literature comparison led to the identification of three regions of interest that display structural heterogeneity within past secondary structure models. To test the accuracy of the secondary structure models in these regions, we employed mutational analysis and SHAPE probing. Mutational analysis revealed that two helical regions within the identified regions of interest are important for IRES translation. These helical regions are consistent with only one of the structure predictions in the literature and do not form in EMCV IRES structures predicted using modern secondary structure prediction methods. The importance of these regions is further supported by multiple SHAPE protections when probing was performed after in vitro translation, indicating that these regions are involved in the IRES translation complex. This work validates a published structure and demonstrates the importance of domain I during EMCV IRES translation initiation.- Published
- 2024
- Full Text
- View/download PDF
45. The Repurposing of Cellular Proteins during Enterovirus A71 Infection.
- Author
-
Abedeera SM, Davila-Calderon J, Haddad C, Henry B, King J, Penumutchu S, and Tolbert BS
- Subjects
- Child, Infant, Humans, Drug Repositioning, 5' Untranslated Regions, Internal Ribosome Entry Sites, Antigens, Viral, RNA, Viral genetics, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Enterovirus Infections, Enterovirus genetics
- Abstract
Viruses pose a great threat to people's lives. Enterovirus A71 (EV-A71) infects children and infants all over the world with no FDA-approved treatment to date. Understanding the basic mechanisms of viral processes aids in selecting more efficient drug targets and designing more effective antivirals to thwart this virus. The 5'-untranslated region (5'-UTR) of the viral RNA genome is composed of a cloverleaf structure and an internal ribosome entry site (IRES). Cellular proteins that bind to the cloverleaf structure regulate viral RNA synthesis, while those that bind to the IRES also known as IRES trans-acting factors (ITAFs) regulate viral translation. In this review, we survey the cellular proteins currently known to bind the 5'-UTR and influence viral gene expression with emphasis on comparing proteins' functions and localizations pre- and post-(EV-A71) infection. A comprehensive understanding of how the host cell's machinery is hijacked and reprogrammed by the virus to facilitate its replication is crucial for developing effective antivirals.
- Published
- 2023
- Full Text
- View/download PDF
46. Rational design of eukaryotic riboswitches that up-regulate IRES-mediated translation initiation with high switching efficiency through a kinetic trapping mechanism in vitro.
- Author
-
Takahashi H, Fujikawa M, and Ogawa A
- Subjects
- Internal Ribosome Entry Sites, Ligands, Bacteria genetics, Kinetics, Riboswitch genetics
- Abstract
In general, riboswitches functioning through a cotranscriptional kinetic trapping mechanism (kt-riboswitches) show higher switching efficiencies in response to practical concentrations of their ligand molecules than eq-riboswitches, which function by an equilibrium mechanism. However, the former have been much more difficult to design due to their more complex mechanism. We here successfully developed a rational strategy for constructing eukaryotic kt-riboswitches that ligand-dependently enhance translation initiation mediated by an internal ribosome entry site (IRES). This was achieved both by utilizing some predicted structural features of a highly efficient bacterial kt-riboswitch identified through screening and by completely decoupling an aptamer domain from the IRES. Three kt-riboswitches optimized through this strategy, each responding to a different ligand, exhibited three- to sevenfold higher induction ratios (up to ∼90) than previously optimized eq-riboswitches regulating the same IRES-mediated translation in wheat germ extract. Because the IRES used functions well in various eukaryotic expression systems, these types of kt-riboswitches are expected to serve as major eukaryotic gene regulators based on RNA. In addition, the present strategy could be applied to the rational construction of other types of kt-riboswitches, including those functioning in bacterial expression systems., (© 2023 Takahashi et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2023
- Full Text
- View/download PDF
47. IRES-mediated translation of cofilin regulates axonal growth cone extension and turning.
- Author
-
Choi, Jung‐Hyun, Wang, Wei, Park, Dongkeun, Kim, Sung‐Hoon, Kim, Kyong‐Tai, and Min, Kyung‐Tai
- Subjects
- *
RIBOSOMES , *AXONAL transport , *NEURAL development , *ACTIN , *MESSENGER RNA , *SPATIOTEMPORAL processes - Abstract
In neuronal development, dynamic rearrangement of actin promotes axonal growth cone extension, and spatiotemporal translation of local mRNAs in response to guidance cues directs axonal growth cone steering, where cofilin plays a critical role. While regulation of cofilin activity is well studied, regulatory mechanism for cofilin mRNA translation in neurons is unknown. In eukaryotic cells, proteins can be synthesized by cap-dependent or cap-independent mechanism via internal ribosome entry site (IRES)-mediated translation. IRES-mediated translation has been reported in various pathophysiological conditions, but its role in normal physiological environment is poorly understood. Here, we report that 50UTR of cofilin mRNA contains an IRES element, and cofilin is predominantly translated by IRES-mediated mechanism in neurons. Furthermore, we show that IRES-mediated translation of cofilin is required for both axon extension and axonal growth cone steering. Our results provide new insights into the function of IRES-mediated translation in neuronal development. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
48. Bi-directional ribosome scanning controls the stringency of start codon selection
- Author
-
Leiming Dong, Longfei Jia, Shu-Bing Qian, Yuanhui Mao, and Yifei Gu
- Subjects
Untranslated region ,endocrine system ,Science ,General Physics and Astronomy ,Codon, Initiator ,Internal Ribosome Entry Sites ,Ribosome ,General Biochemistry, Genetics and Molecular Biology ,Article ,Eukaryotic translation ,Start codon ,Yeasts ,Humans ,RNA, Messenger ,Peptide Chain Initiation, Translational ,Physics ,Adenosine Triphosphatases ,Messenger RNA ,Multidisciplinary ,food and beverages ,Translation (biology) ,General Chemistry ,biochemical phenomena, metabolism, and nutrition ,Cell biology ,Internal ribosome entry site ,HEK293 Cells ,eIF4A ,Protein Biosynthesis ,Eukaryotic Initiation Factor-4A ,RNA ,5' Untranslated Regions ,Ribosomes ,Sequence Alignment ,RNA Helicases - Abstract
The fidelity of start codon recognition by ribosomes is paramount during protein synthesis. The current knowledge of eukaryotic translation initiation implies unidirectional 5ʹ→3ʹ migration of the pre-initiation complex (PIC) along the 5ʹ UTR. In probing translation initiation from ultra-short 5ʹ UTR, we report that an AUG triplet near the 5ʹ end can be selected via PIC backsliding. Bi-directional ribosome scanning is supported by competitive selection of closely spaced AUG codons and recognition of two initiation sites flanking an internal ribosome entry site. Transcriptome-wide PIC profiling reveals footprints with an oscillation pattern near the 5ʹ end and start codons. Depleting the RNA helicase eIF4A leads to reduced PIC oscillations and impaired selection of 5ʹ end start codons. Enhancing the ATPase activity of eIF4A promotes nonlinear PIC scanning and stimulates upstream translation initiation. The helicase-mediated PIC conformational switch may provide an operational mechanism that unifies ribosome recruitment, scanning, and start codon selection., Start codon selection is commonly thought to occur through the unidirectional scanning of the mRNA by the 40 S ribosome. Here the authors provide evidence that the pre-initiation complex can backslide on the mRNA to initiate translation at upstream AUG codons.
- Published
- 2021
49. Impacts of single nucleotide deletions from the 3′ end of Senecavirus A 5′ untranslated region on activity of viral IRES and on rescue of recombinant virus
- Author
-
Fuxiao Liu, Ning Wang, Hu Shan, and Qi Wang
- Subjects
Untranslated region ,Five prime untranslated region ,viruses ,DNA-Directed RNA Polymerases ,Picornaviridae ,Internal Ribosome Entry Sites ,Biology ,Recombinant virus ,Polymorphism, Single Nucleotide ,Molecular biology ,Virus ,Cell Line ,Viral Proteins ,Internal ribosome entry site ,Start codon ,Virology ,Complementary DNA ,Animals ,Cattle ,Cloning, Molecular ,5' Untranslated Regions ,Pseudoknot ,Gene Deletion ,Reassortant Viruses - Abstract
The 5′ untranslated region (UTR) of Senecavirus A (SVA) harbors an internal ribosome entry site (IRES), in which a pseudoknot structure is upstream of start codon AUG. Wild-type SVAs have a highly conserved 13-nt-sequence between the pseudoknot stem II (PKS-II)-forming motif and the AUG. In this study, a single nucleotide was deleted one by one from the 13-nt-sequence within a wild-type SVA minigenome. The result showed that neither mono- nor multi-nucleotide deletions abolished the IRES activity. Furthermore, a single nucleotide was deleted one by one from the 13-nt-sequence within a full-length SVA cDNA clone. The result indicated that nucleotide-deleting SVAs could be rescued from 1- to 5-nt-deleting cDNA clones, whereas only the 1- and 2-nt-deleting viruses were genetically stable during nine serial passages in vitro. Additionally, only the 1-nt-deleting SVA showed similar growth kinetics to that of the wild-type virus, suggesting that the pseudoknot-AUG distance was crucial for SVA replication.
- Published
- 2021
- Full Text
- View/download PDF
50. hnRNP‐A1 binds to the IRES of MELOE‐1 antigen to promote MELOE‐1 translation in stressed melanoma cells
- Author
-
Emmanuelle Com, François Lang, Charles Pineau, Catherine Rabu, Nathalie Labarrière, Agnès Fortun, Emilie Dupré, Floriane Briand, Maud Charpentier, and Mike Maillasson
- Subjects
Cancer Research ,Heterogeneous Nuclear Ribonucleoprotein A1 ,medicine.medical_treatment ,Internal Ribosome Entry Sites ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,IRES ,Antigens, Neoplasm ,melanoma ,Genetics ,medicine ,Humans ,long noncoding RNA ,RC254-282 ,Research Articles ,030304 developmental biology ,0303 health sciences ,Melanoma ,Endoplasmic reticulum ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Translation (biology) ,General Medicine ,Immunotherapy ,medicine.disease ,ITAF ,Neoplasm Proteins ,3. Good health ,Cell biology ,Internal ribosome entry site ,tumor antigens ,Oncology ,Protein Biosynthesis ,030220 oncology & carcinogenesis ,Unfolded protein response ,Molecular Medicine ,ER stress ,Ribosomes ,Research Article - Abstract
The major challenge in antigen‐specific immunotherapy of cancer is to select the most relevant tumor antigens to target. To this aim, understanding their mode of expression by tumor cells is critical. We previously identified a melanoma‐specific antigen, melanoma‐overexpressed antigen 1 (MELOE‐1)—coded for by a long noncoding RNA—whose internal ribosomal entry sequence (IRES)‐dependent translation is restricted to tumor cells. This restricted expression is associated with the presence of a broad‐specific T‐cell repertoire that is involved in tumor immunosurveillance in melanoma patients. In the present work, we explored the translation control of MELOE‐1 and provide evidence that heterogeneous nuclear ribonucleoprotein A1 (hnRNP‐A1) binds to the MELOE‐1 IRES and acts as an IRES trans‐activating factor (ITAF) to promote the translation of MELOE‐1 in melanoma cells. In addition, we showed that endoplasmic reticulum (ER) stress induced by thapsigargin, which promotes hnRNP‐A1 cytoplasmic translocation, enhances MELOE‐1 translation and recognition of melanoma cells by a MELOE‐1‐specific T‐cell clone. These findings suggest that pharmacological stimulation of stress pathways may enhance the efficacy of immunotherapies targeting stress‐induced tumor antigens such as MELOE‐1., Our study demonstrates that reticular stress in melanoma cells promoted the translocation of the IRES transacting factor hnRPN‐A1 to the cytoplasm. By binding to the IRES site of meloe mRNA, hnRPN‐A1 promoted the translation of the melanoma‐specific antigen MELOE‐1 and led to the expression of a new immunogenic HLA/peptide complex at the cell surface. Altogether, our data suggest that pharmacological stimulation of stress pathways may enhance the efficacy of T‐cell based immunotherapies in targeting stress‐induced tumor antigens, such as MELOE‐1 in patients with melanoma.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.