Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 45(22): 13016-13028, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29069411

ABSTRACT

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


Subject(s)
Internal Ribosome Entry Sites/genetics , Pestivirus/genetics , Picornaviridae/genetics , RNA, Viral/genetics , Animals , Base Sequence , Binding Sites/genetics , Border disease virus/genetics , Border disease virus/physiology , Cell Line , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/physiology , HEK293 Cells , Host-Pathogen Interactions , Humans , Nucleic Acid Conformation , Pestivirus/physiology , Picornaviridae/physiology , RNA, Viral/chemistry , RNA, Viral/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Swine
2.
J Virol ; 90(14): 6489-6501, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27147742

ABSTRACT

UNLABELLED: In response to stress such as virus infection, cells can stall translation by storing mRNAs away in cellular compartments called stress granules (SGs). This defense mechanism favors cell survival by limiting the use of energy and nutrients until the stress is resolved. In some cases it may also block viral propagation as viruses are dependent on the host cell resources to produce viral proteins. Human norovirus is a member of the Caliciviridae family responsible for gastroenteritis outbreaks worldwide. Previous studies on caliciviruses have identified mechanisms by which they can usurp the host translational machinery, using the viral protein genome-linked VPg, or regulate host protein synthesis through the mitogen-activated protein kinase (MAPK) pathway. Here, we examined the effect of feline calicivirus (FCV) infection on SG accumulation. We show that FCV infection impairs the assembly of SGs despite an increased phosphorylation of eukaryotic initiation factor eIF2α, a hallmark of stress pathway activation. Furthermore, SGs did not accumulate in FCV-infected cells that were stressed with arsenite or hydrogen peroxide. FCV infection resulted in the cleavage of the SG-nucleating protein Ras-GTPase activating SH3 domain-binding protein (G3BP1), which is mediated by the viral 3C-like proteinase NS6(Pro) Using mutational analysis, we identified the FCV-induced cleavage site within G3BP1, which differs from the poliovirus 3C proteinase cleavage site previously identified. Finally, we showed that NS6(Pro)-mediated G3BP1 cleavage impairs SG assembly. In contrast, murine norovirus (MNV) infection did not impact arsenite-induced SG assembly or G3BP1 integrity, suggesting that related caliciviruses have distinct effects on the stress response pathway. IMPORTANCE: Human noroviruses are a major cause of viral gastroenteritis, and it is important to understand how they interact with the infected host cell. Feline calicivirus (FCV) and murine norovirus (MNV) are used as models to understand norovirus biology. Recent studies have suggested that the assembly of stress granules is central in orchestrating stress and antiviral responses to restrict viral replication. Overall, our study provides the first insight on how caliciviruses impair stress granule assembly by targeting the nucleating factor G3BP1 via the viral proteinase NS6(Pro) This work provides new insights into host-pathogen interactions that regulate stress pathways during FCV infection.


Subject(s)
Caliciviridae Infections/virology , Calicivirus, Feline/pathogenicity , Carrier Proteins/metabolism , Cytoplasmic Granules/metabolism , Host-Pathogen Interactions , Virus Replication , 3C Viral Proteases , Animals , Caliciviridae Infections/metabolism , Caliciviridae Infections/pathology , Carrier Proteins/genetics , Cats , Cysteine Endopeptidases/metabolism , Cytoplasmic Granules/virology , DNA Helicases , Eukaryotic Initiation Factor-2/metabolism , HeLa Cells , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , Viral Proteins/metabolism
3.
J Biol Chem ; 290(8): 4748-4758, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25561727

ABSTRACT

Protein synthesis is a tightly controlled process responding to several stimuli, including viral infection. As obligate intracellular parasites, viruses depend on the translation machinery of the host and can manipulate it by affecting the availability and function of specific eukaryotic initiation factors (eIFs). Human norovirus is a member of the Caliciviridae family and is responsible for gastroenteritis outbreaks. Previous studies on feline calicivirus and murine norovirus 1 (MNV1) demonstrated that the viral protein, genome-linked (VPg), acts to direct translation by hijacking the host protein synthesis machinery. Here we report that MNV1 infection modulates the MAPK pathway to activate eIF4E phosphorylation. Our results show that the activation of p38 and Mnk during MNV1 infection is important for MNV1 replication. Furthermore, phosphorylated eIF4E relocates to the polysomes, and this contributes to changes in the translational state of specific host mRNAs. We propose that global translational control of the host by eIF4E phosphorylation is a key component of the host-pathogen interaction.


Subject(s)
Caliciviridae Infections/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Host-Pathogen Interactions , Norovirus/physiology , Protein Biosynthesis , Viral Proteins/biosynthesis , Virus Replication/physiology , Animals , Caliciviridae Infections/genetics , Cats , Cell Line , Eukaryotic Initiation Factor-4E/genetics , Humans , Mice , Phosphorylation/genetics , Polyribosomes/genetics , Polyribosomes/metabolism , Protein Transport/genetics , Viral Proteins/genetics
4.
RNA ; 20(11): 1803-14, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25246653

ABSTRACT

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


Subject(s)
Herpesvirus 8, Human/genetics , RNA, Viral/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Binding Sites , Cell Line, Tumor , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Models, Molecular , Nucleic Acid Conformation , RNA, Viral/genetics , Ribosomes/metabolism , Transcription, Genetic
5.
J Biol Chem ; 289(31): 21738-50, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24928504

ABSTRACT

Viruses have evolved a variety of mechanisms to usurp the host cell translation machinery to enable translation of the viral genome in the presence of high levels of cellular mRNAs. Noroviruses, a major cause of gastroenteritis in man, have evolved a mechanism that relies on the interaction of translation initiation factors with the virus-encoded VPg protein covalently linked to the 5' end of the viral RNA. To further characterize this novel mechanism of translation initiation, we have used proteomics to identify the components of the norovirus translation initiation factor complex. This approach revealed that VPg binds directly to the eIF4F complex, with a high affinity interaction occurring between VPg and eIF4G. Mutational analyses indicated that the C-terminal region of VPg is important for the VPg-eIF4G interaction; viruses with mutations that alter or disrupt this interaction are debilitated or non-viable. Our results shed new light on the unusual mechanisms of protein-directed translation initiation.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , Genome, Viral , Norovirus/genetics , Protein Biosynthesis , Viral Proteins/physiology , Base Sequence , Chromatography, Affinity , DNA Primers , Polymerase Chain Reaction , Protein Binding , Proteomics , Viral Proteins/genetics , Viral Proteins/metabolism
6.
PLoS One ; 8(12): e82234, 2013.
Article in English | MEDLINE | ID: mdl-24376523

ABSTRACT

Internal ribosome entry site (IRES) elements found in the 5' untranslated region of mRNAs enable translation initiation in a cap-independent manner, thereby representing an alternative to cap-dependent translation in cell-free protein expression systems. However, IRES function is largely species-dependent so their utility in cell-free systems from different species is rather limited. A promising approach to overcome these limitations would be the use of IRESs that are able to recruit components of the translation initiation apparatus from diverse origins. Here, we present a solution to this technical problem and describe the ability of a number of viral IRESs to direct efficient protein expression in different eukaryotic cell-free expression systems. The IRES from the intergenic region (IGR) of the Cricket paralysis virus (CrPV) genome was shown to function efficiently in four different cell-free systems based on lysates derived from cultured Sf21, CHO and K562 cells as well as wheat germ. Our results suggest that the CrPV IGR IRES-based expression vector is universally applicable for a broad range of eukaryotic cell lysates. Sf21, CHO and K562 cell-free expression systems are particularly promising platforms for the production of glycoproteins and membrane proteins since they contain endogenous microsomes that facilitate the incorporation of membrane-spanning proteins and the formation of post-translational modifications. We demonstrate the use of the CrPV IGR IRES-based expression vector for the enhanced synthesis of various target proteins including the glycoprotein erythropoietin and the membrane proteins heparin-binding EGF-like growth factor receptor as well as epidermal growth factor receptor in the above mentioned eukaryotic cell-free systems. CrPV IGR IRES-mediated translation will facilitate the development of novel eukaryotic cell-free expression platforms as well as the high-yield synthesis of desired proteins in already established systems.


Subject(s)
5' Untranslated Regions/genetics , Eukaryotic Cells/metabolism , Glycoproteins/biosynthesis , Membrane Proteins/biosynthesis , Protein Biosynthesis/genetics , Animals , Autoradiography , Cell Line , Cell-Free System , Fluorescence , Gene Expression , Humans , Microscopy, Confocal , Viruses/genetics
7.
J Virol ; 87(10): 5318-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23487472

ABSTRACT

We report the solution structures of the VPg proteins from feline calicivirus (FCV) and murine norovirus (MNV), which have been determined by nuclear magnetic resonance spectroscopy. In both cases, the core of the protein adopts a compact helical structure flanked by flexible N and C termini. Remarkably, while the core of FCV VPg contains a well-defined three-helix bundle, the MNV VPg core has just the first two of these secondary structure elements. In both cases, the VPg cores are stabilized by networks of hydrophobic and salt bridge interactions. The Tyr residue in VPg that is nucleotidylated by the viral NS7 polymerase (Y24 in FCV, Y26 in MNV) occurs in a conserved position within the first helix of the core. Intriguingly, given its structure, VPg would appear to be unable to bind to the viral polymerase so as to place this Tyr in the active site without a major conformation change to VPg or the polymerase. However, mutations that destabilized the VPg core either had no effect on or reduced both the ability of the protein to be nucleotidylated and virus infectivity and did not reveal a clear structure-activity relationship. The precise role of the calicivirus VPg core in virus replication remains to be determined, but knowledge of its structure will facilitate future investigations.


Subject(s)
Calicivirus, Feline/chemistry , Norovirus/chemistry , Viral Proteins/chemistry , Animals , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Conformation
8.
J Virol ; 85(9): 4452-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21325406

ABSTRACT

The RNA genome of Seneca Valley virus (SVV), a recently identified picornavirus, contains an internal ribosome entry site (IRES) element which has structural and functional similarity to that from classical swine fever virus (CSFV) and hepatitis C virus, members of the Flaviviridae. The SVV IRES has an absolute requirement for the presence of a short region of virus-coding sequence to allow it to function either in cells or in rabbit reticulocyte lysate. The IRES activity does not require the translation initiation factor eIF4A or intact eIF4G. The predicted secondary structure indicates that the SVV IRES is more closely related to the CSFV IRES, including the presence of a bipartite IIId domain. Mutagenesis of the SVV IRES, coupled to functional assays, support the core elements of the IRES structure model, but surprisingly, deletion of the conserved IIId(2) domain had no effect on IRES activity, including 40S and eIF3 binding. This is the first example of a picornavirus IRES that is most closely related to the CSFV IRES and suggests the possibility of multiple, independent recombination events between the genomes of the Picornaviridae and Flaviviridae to give rise to similar IRES elements.


Subject(s)
Picornaviridae/genetics , Protein Biosynthesis , RNA, Viral/genetics , Ribosomes/metabolism , Animals , Cell Extracts , Cell Line , Classical Swine Fever Virus/genetics , DNA Mutational Analysis , Humans , Mutation , Nucleic Acid Conformation , Pestivirus/genetics , Picornaviridae/chemistry , RNA, Viral/chemistry , RNA, Viral/metabolism , Rabbits , Sequence Deletion
9.
PLoS One ; 5(11): e13924, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21085714

ABSTRACT

Double subgenomic Sindbis virus (dsSINV) vectors are widely used for the expression of proteins, peptides, and RNA sequences. These recombinant RNA viruses permit high level expression of a heterologous sequence in a wide range of animals, tissues, and cells. However, the alphavirus genome structure and replication strategy is not readily amenable to the expression of more than one heterologous sequence. The Rhopalosiphum padi virus (RhPV) genome contains two internal ribosome entry site (IRES) elements that mediate cap-independent translation of the virus nonstructural and structural proteins. Most IRES elements that have been characterized function only in mammalian cells but previous work has shown that the IRES element present in the 5' untranslated region (UTR) of the RhPV genome functions efficiently in mammalian, insect, and plant systems. To determine if the 5' RhPV IRES element could be used to express more than one heterologous sequence from a dsSINV vector, RhPV 5' IRES sequences were placed between genes for two different fluorescent marker proteins in the dsSINV, TE/3'2J/mcs. While mammalian and insect cells infected with recombinant viruses containing the RhPV sequences expressed both fluorescent marker proteins, only single marker proteins were routinely observed in cells infected with dsSINV vectors in which the RhPV IRES had been replaced by a luciferase fragment, an antisense RhPV IRES, or no intergenic sequence. Thus, we report development of a versatile tool for the expression of multiple sequences in diverse cell types.


Subject(s)
Luminescent Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Ribosomes/metabolism , Sindbis Virus/genetics , Aedes/cytology , Aedes/virology , Alphavirus/genetics , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Cells, Cultured , Chlorocebus aethiops , Genetic Vectors/genetics , Genome, Viral/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Microscopy, Fluorescence , Picornaviridae/genetics , Recombinant Fusion Proteins/genetics , Vero Cells
10.
PLoS One ; 5(3): e9562, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20224775

ABSTRACT

BACKGROUND: Positive strand RNA viruses rely heavily on host cell RNA binding proteins for various aspects of their life cycle. Such proteins interact with sequences usually present at the 5' or 3' extremities of the viral RNA genome, to regulate viral translation and/or replication. We have previously reported that the well characterized host RNA binding protein polypyrimidine tract binding protein (PTB) interacts with the 5'end of the feline calicivirus (FCV) genomic and subgenomic RNAs, playing a role in the FCV life cycle. PRINCIPAL FINDINGS: We have demonstrated that PTB interacts with at least two binding sites within the 5'end of the FCV genome. In vitro translation indicated that PTB may function as a negative regulator of FCV translation and this was subsequently confirmed as the translation of the viral subgenomic RNA in PTB siRNA treated cells was stimulated under conditions in which RNA replication could not occur. We also observed that PTB redistributes from the nucleus to the cytoplasm during FCV infection, partially localizing to viral replication complexes, suggesting that PTB binding may be involved in the switch from translation to replication. Reverse genetics studies demonstrated that synonymous mutations in the PTB binding sites result in a cell-type specific defect in FCV replication. CONCLUSIONS: Our data indicates that PTB may function to negatively regulate FCV translation initiation. To reconcile this with efficient virus replication in cells, we propose a putative model for the function of PTB in the FCV life cycle. It is possible that during the early stages of infection, viral RNA is translated in the absence of PTB, however, as the levels of viral proteins increase, the nuclear-cytoplasmic shuttling of PTB is altered, increasing the cytoplasmic levels of PTB, inhibiting viral translation. Whether PTB acts directly to repress translation initiation or via the recruitment of other factors remains to be determined but this may contribute to the stimulation of viral RNA replication via clearance of ribosomes from viral RNA.


Subject(s)
Calicivirus, Feline/metabolism , Gene Expression Regulation, Viral , Polypyrimidine Tract-Binding Protein/physiology , Animals , Binding Sites , Cats , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Genome, Viral , Green Fluorescent Proteins/metabolism , Mutation , Polypyrimidine Tract-Binding Protein/genetics , Protein Binding , Protein Biosynthesis , RNA, Small Interfering/metabolism , Ribonuclease H/metabolism , Virus Replication
11.
J Gen Virol ; 91(Pt 6): 1428-38, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20147520

ABSTRACT

For many viruses, endocytosis and exposure to the low pH within acidic endosomes is essential for infection. It has previously been reported that feline calicivirus uses clathrin-mediated endocytosis for entry into mammalian cells. Here, we report that infection of RAW264.7 macrophages by the closely related murine norovirus-1 (MNV-1) does not require the clathrin pathway, as infection was not inhibited by expression of dominant-negative Eps15 or by knockdown of the adaptin-2 complex. Further, infection was not inhibited by reagents that raise endosomal pH. RAW264.7 macrophages were shown not to express caveolin, and flotillin depletion did not inhibit infection, suggesting that caveolae and the flotillin pathway are not required for cell entry. However, MNV-1 infection was inhibited by methyl-beta-cyclodextrin and the dynamin inhibitor, dynasore. Addition of these drugs to the cells after a period of virus internalization did not inhibit infection, suggesting the involvement of cholesterol-sensitive lipid rafts and dynamin in the entry mechanism. Macropinocytosis (MPC) was shown to be active in RAW264.7 macrophages (as indicated by uptake of dextran) and could be blocked by 5-(N-ethyl-N-isopropyl) amiloride (EIPA), which is reported to inhibit this pathway. However, infection was enhanced in the presence of EIPA. Similarly, actin disruption, which also inhibits MPC, resulted in enhanced infection. These results suggest that MPC could contribute to virus degradation or that inhibition of MPC could lead to the upregulation of other endocytic pathways of virus uptake.


Subject(s)
Cholesterol/metabolism , Dynamins/metabolism , Norovirus/physiology , Virus Internalization , Animals , Cell Line , Dynamins/antagonists & inhibitors , Hydrazones/pharmacology , Macrophages/virology , Mice , beta-Cyclodextrins/pharmacology
12.
Virus Res ; 139(2): 157-65, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18755228

ABSTRACT

Translation initiation on the majority of cellular mRNAs is mediated by a cap structure on the 5' end of the mRNA and involves the binding of initiation factors to the cap, followed by recruitment of the 40S ribosomal subunit. However, a number of viral mRNAs are translated using an alternative mechanism, termed internal initiation. In this case, initiation factors and the 40S subunit bind to an internal ribosome entry site (IRES) structure within the 5' untranslated region (UTR) of the mRNA. Although there is no common feature amongst all viral IRES elements, most are believed to contain extensive secondary structure and some of these structures are important for their function. However, an IRES element from the 5' UTR of the genome of Rhopalosiphum padi virus, a Dicistrovirus, challenges this paradigm. This IRES has been shown to function in a number of different translation systems. Intriguingly, the functional region of this IRES element is largely unstructured, and multiple regions from within the 5' UTR function as efficiently as the full-length sequence. This review compares and contrasts the features of this atypical IRES element with other IRES elements and discusses its possible mechanism of action.


Subject(s)
5' Untranslated Regions , Genome, Viral , RNA Viruses/genetics , RNA, Viral/chemistry , Ribosomes/chemistry , Base Sequence , Insect Viruses/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Peptide Chain Initiation, Translational , Peptide Initiation Factors/genetics , Picornaviridae/genetics , RNA, Messenger/chemistry
13.
Prog Mol Biol Transl Sci ; 90: 313-67, 2009.
Article in English | MEDLINE | ID: mdl-20374746

ABSTRACT

Viruses do not carry their own protein biosynthesis machinery and the translation of viral proteins therefore requires that the virus usurps the machinery of the host cell. To allow optimal translation of viral proteins at the expense of cellular proteins, virus families have evolved a variety of methods to repress the host translation machinery, while allowing effective viral protein synthesis. Many viruses use noncanonical mechanisms that permit translation of their own RNAs under these conditions. Viruses have also developed mechanisms to evade host innate immune responses that would repress translation under conditions of viral infection, in particular PKR activation in response to double-stranded RNA (dsRNA). Importantly, the study of viral translation mechanisms has enormously enhanced our understanding of many aspects of the cellular protein biosynthesis pathway and its components. A number of unusual mechanisms of translation initiation that were first discovered in viruses have since been observed in cellular mRNAs, and it has become apparent that a diverse range of translation mechanisms operates in eukaryotes, allowing subtle regulation of this essential process.


Subject(s)
Mammals/metabolism , Mammals/virology , Protein Biosynthesis , Viruses/metabolism , Animals , Eukaryotic Initiation Factor-2/metabolism , Host-Pathogen Interactions , Humans , Phosphorylation , Viral Proteins/biosynthesis
14.
RNA ; 14(8): 1671-80, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18567818

ABSTRACT

The initiation of protein synthesis on mRNAs within eukaryotic cells is achieved either by a 5' cap-dependent mechanism or through internal initiation directed by an internal ribosome entry site (IRES). Picornavirus IRES elements, located in the 5' untranslated region (5'UTR), contain extensive secondary structure and multiple upstream AUG codons. These features can be expected to inhibit cap-dependent initiation of translation. However, we have now shown that certain mutant hepatitis C virus-like picornavirus IRES elements (from porcine teschovirus-1 and avian encephalomyelitis virus), which are unable to direct internal initiation, are not significant barriers to efficient translation of capped monocistronic mRNAs that contain these defective elements within their 5'UTRs. Moreover, the translation of these mRNAs is highly sensitive to the expression of an enterovirus 2A protease (which induces cleavage of eIF4G) and is also inhibited by hippuristanol, a specific inhibitor of eIF4A function, in contrast to their parental wild-type IRES elements. These results provide a possible basis for the evolution of viral IRES elements within the context of functional mRNAs that are translated by a cap-dependent mechanism.


Subject(s)
Picornaviridae/genetics , Regulatory Sequences, Ribonucleic Acid , 5' Untranslated Regions/chemistry , 5' Untranslated Regions/metabolism , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Humans , Peptide Chain Initiation, Translational , Picornaviridae/chemistry , Picornaviridae/metabolism , Protein Biosynthesis , RNA Caps/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribosomes/metabolism , Sterols/pharmacology
15.
Int J Biochem Cell Biol ; 40(12): 2675-80, 2008.
Article in English | MEDLINE | ID: mdl-18069043

ABSTRACT

Eukaryotic translation initiation factor 4E (eIF4E) is perhaps best known for its function in the initiation of protein synthesis on capped mRNAs in the cytoplasm. However, recent studies have highlighted that eIF4E has many additional functions, which include the nuclear export of specific mRNAs as well as roles in ageing and the translation of some uncapped viral RNAs. This review aims to update the reader on recent developments, including the potential of eIF4E as a therapeutic target.


Subject(s)
Eukaryotic Initiation Factor-4E , Protein Biosynthesis , Active Transport, Cell Nucleus/genetics , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Models, Biological , Models, Molecular , Protein Conformation , RNA Caps/genetics , RNA Caps/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
J Virol ; 82(4): 1993-2003, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077729

ABSTRACT

Avian encephalomyelitis virus (AEV) is a picornavirus that causes disease in poultry worldwide, and flocks must be vaccinated for protection. AEV is currently classified within the hepatovirus genus, since its proteins are most closely related to those of hepatitis A virus (HAV). We now provide evidence that the 494-nucleotide-long 5' untranslated region of the AEV genome contains an internal ribosome entry site (IRES) element that functions efficiently in vitro and in mammalian cells. Unlike the HAV IRES, the AEV IRES is relatively short and functions in the presence of cleaved eIF4G and it is also resistant to an inhibitor of eIF4A. These properties are reminiscent of the recently discovered class of IRES elements within certain other picornaviruses, such as porcine teschovirus 1 (PTV-1). Like the PTV-1 IRES, the AEV IRES shows significant similarity to the hepatitis C virus (HCV) IRES in sequence, function, and predicted secondary structure. Furthermore, mutational analysis of the predicted pseudoknot structure at the 3' end of the AEV IRES lends support to the secondary structure we present. AEV is therefore another example of a picornavirus harboring an HCV-like IRES element within its genome, and thus, its classification within the hepatovirus genus may need to be reassessed in light of these findings.


Subject(s)
Encephalomyelitis Virus, Avian/genetics , Genome, Viral , Hepacivirus/genetics , RNA, Viral/metabolism , Ribosomes/metabolism , 5' Untranslated Regions/chemistry , 5' Untranslated Regions/genetics , 5' Untranslated Regions/metabolism , Animals , Base Sequence/drug effects , Encephalomyelitis Virus, Avian/classification , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/metabolism , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Picornaviridae/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Sequence Analysis, RNA
17.
EMBO J ; 26(17): 3936-44, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17690690

ABSTRACT

The ErbB3-binding protein 1 (Ebp1) is an important regulator of transcription, affecting eukaryotic cell growth, proliferation, differentiation and survival. Ebp1 can also affect translation and cooperates with the polypyrimidine tract-binding protein (PTB) to stimulate the activity of the internal ribosome entry site (IRES) of foot-and-mouth disease virus (FMDV). We report here the crystal structure of murine Ebp1 (p48 isoform), providing the first glimpse of the architecture of this versatile regulator. The structure reveals a core domain that is homologous to methionine aminopeptidases, coupled to a C-terminal extension that contains important motifs for binding proteins and RNA. It sheds new light on the conformational differences between the p42 and p48 isoforms of Ebp1, the disposition of the key protein-interacting motif ((354)LKALL(358)) and the RNA-binding activity of Ebp1. We show that the primary RNA-binding site is formed by a Lys-rich motif in the C terminus and mediates the interaction with the FMDV IRES. We also demonstrate a specific functional requirement for Ebp1 in FMDV IRES-directed translation that is independent of a direct interaction with PTB.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Models, Molecular , Adaptor Proteins, Signal Transducing/physiology , Aminopeptidases/chemistry , Binding Sites , Foot-and-Mouth Disease Virus/genetics , Lysine/chemistry , Methionyl Aminopeptidases , Protein Biosynthesis , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/physiology , RNA, Small Interfering/genetics , RNA, Viral/chemistry , Transcriptional Activation
18.
J Gen Virol ; 88(Pt 5): 1583-1588, 2007 May.
Article in English | MEDLINE | ID: mdl-17412990

ABSTRACT

Rhopalosiphum padi virus (RhPV) is a member of the family Dicistroviridae. The genomes of viruses in this family contain two open reading frames, each preceded by distinct internal ribosome entry site (IRES) elements. The RhPV 5' IRES is functional in mammalian, insect and plant translation systems and can form 48S initiation complexes in vitro with just the mammalian initiation factors eIF2, eIF3 and eIF1. Large regions of the 5' untranslated region (UTR) can be deleted without affecting initiation-complex formation. The minimal sequences required for directing internal initiation in mammalian (rabbit reticulocyte lysate), plant (wheatgerm extract) and insect (Sf21 cells) translation systems have now been defined. A fragment (nt 426-579) from the 3' portion of the 5' UTR can direct translation in each of these translation systems. In addition, a distinct region (nt 300-429) is also active. Thus, unstructured regions within the 5' UTR seem to be critical for IRES function.


Subject(s)
5' Untranslated Regions/genetics , Aphids/virology , Insect Viruses/genetics , Peptide Chain Termination, Translational , Protein Biosynthesis , Animals , DNA, Viral/genetics , DNA, Viral/isolation & purification , Insect Viruses/isolation & purification , Mammals/virology , Plants/virology , Plasmids , Viral Proteins/biosynthesis
19.
J Gen Virol ; 87(Pt 11): 3339-3347, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17030868

ABSTRACT

The interaction of host-cell nucleic acid-binding proteins with the genomes of positive-stranded RNA viruses is known to play a role in the translation and replication of many viruses. To date, however, the characterization of similar interactions with the genomes of members of the family Caliciviridae has been limited to in vitro binding analysis. In this study, Feline calicivirus (FCV) has been used as a model system to identify and characterize the role of host-cell factors that interact with the viral RNA. It was demonstrated that polypyrimidine tract-binding protein (PTB) interacts specifically with the 5' sequences of the FCV genomic and subgenomic RNAs. Using RNA interference it was shown that PTB is required for efficient FCV replication in a temperature-dependent manner. siRNA-mediated knockdown of PTB resulted in a 15- to 100-fold reduction in virus titre, as well as a concomitant reduction in viral RNA and protein synthesis at 32 degrees C. In addition, virus-induced cytopathic effect was significantly delayed as a result of an siRNA-mediated reduction in PTB levels. A role for PTB in the calicivirus life cycle was more apparent at temperatures above and below 37 degrees C, fitting with the hypothesis that PTB functions as an RNA chaperone, potentially aiding the folding of RNA into functional structures. This is the first functional demonstration of a host-cell protein interacting with a calicivirus RNA.


Subject(s)
Calicivirus, Feline/physiology , Polypyrimidine Tract-Binding Protein/physiology , 5' Untranslated Regions/metabolism , Animals , Cell Line , Gene Deletion , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Protein Binding , RNA, Small Interfering/genetics , RNA, Viral/metabolism , Temperature , Virus Replication
20.
J Biol Chem ; 281(35): 25315-25, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16835235

ABSTRACT

Two classes of viruses, namely members of the Potyviridae and Caliciviridae, use a novel mechanism for the initiation of protein synthesis that involves the interaction of translation initiation factors with a viral protein covalently linked to the viral RNA, known as VPg. The calicivirus VPg proteins can interact directly with the initiation factors eIF4E and eIF3. Translation initiation on feline calicivirus (FCV) RNA requires eIF4E because it is inhibited by recombinant 4E-BP1. However, to date, there have been no functional studies carried out with respect to norovirus translation initiation, because of a lack of a suitable source of VPg-linked viral RNA. We have now used the recently identified murine norovirus (MNV) as a model system for norovirus translation and have extended our previous studies with FCV RNA to examine the role of the other eIF4F components in translation initiation. We now demonstrate that, as with FCV, MNV VPg interacts directly with eIF4E, although, unlike FCV RNA, translation of MNV RNA is not sensitive to 4E-BP1, eIF4E depletion, or foot-and-mouth disease virus Lb protease-mediated cleavage of eIF4G. We also demonstrate that both FCV and MNV RNA translation require the RNA helicase component of the eIF4F complex, namely eIF4A, because translation was sensitive (albeit to different degrees) to a dominant negative form and to a small molecule inhibitor of eIF4A (hippuristanol). These results suggest that calicivirus RNAs differ with respect to their requirements for the components of the eIF4F translation initiation complex.


Subject(s)
Caliciviridae/physiology , Eukaryotic Initiation Factor-4F/chemistry , Gene Expression Regulation, Viral , Animals , Genes, Dominant , Mice , Mutation , Norovirus/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Rabbits , Recombinant Proteins/chemistry , Sterols/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...