Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
mBio ; 12(4): e0178121, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34372695

ABSTRACT

The 2',5'-oligoadenylate (2-5A)-dependent endoribonuclease, RNase L, is a principal mediator of the interferon (IFN) antiviral response. Therefore, the regulation of cellular levels of 2-5A is a key point of control in antiviral innate immunity. Cellular 2-5A levels are determined by IFN-inducible 2',5'-oligoadenylate synthetases (OASs) and by enzymes that degrade 2-5A. Importantly, many coronaviruses (CoVs) and rotaviruses encode 2-5A-degrading enzymes, thereby antagonizing RNase L and its antiviral effects. A-kinase-anchoring protein 7 (AKAP7), a mammalian counterpart, could possibly limit tissue damage from excessive or prolonged RNase L activation during viral infections or from self-double-stranded RNAs that activate OAS. We show that these enzymes, members of the two-histidine phosphoesterase (2H-PE) superfamily, constitute a subfamily referred here as 2',5'-PEs. 2',5'-PEs from the mouse CoV mouse hepatitis virus (MHV) (NS2), Middle East respiratory syndrome coronavirus (MERS-CoV) (NS4b), group A rotavirus (VP3), and mouse (AKAP7) were investigated for their evolutionary relationships and activities. While there was no activity against 3',5'-oligoribonucleotides, they all cleaved 2',5'-oligoadenylates efficiently but with variable activity against other 2',5'-oligonucleotides. The 2',5'-PEs are shown to be metal ion-independent enzymes that cleave trimer 2-5A (2',5'-p3A3) producing mono- or diadenylates with 2',3'-cyclic phosphate termini. Our results suggest that the elimination of 2-5A might be the sole function of viral 2',5'-PEs, thereby promoting viral escape from innate immunity by preventing or limiting the activation of RNase L. IMPORTANCE Viruses often encode accessory proteins that antagonize the host antiviral immune response. Here, we probed the evolutionary relationships and biochemical activities of two-histidine phosphoesterases (2H-PEs) that allow some coronaviruses and rotaviruses to counteract antiviral innate immunity. In addition, we investigated the mammalian enzyme AKAP7, which has homology and shared activities with the viral enzymes and might reduce self-injury. These viral and host enzymes, which we refer to as 2',5'-PEs, specifically degrade 2',5'-oligoadenylate activators of the antiviral enzyme RNase L. We show that the host and viral enzymes are metal ion independent and exclusively cleave 2',5'- and not 3',5'-phosphodiester bonds, producing cleavage products with cyclic 2',3'-phosphate termini. Our study defines 2',5'-PEs as enzymes that share characteristic conserved features with the 2H-PE superfamily but have specific and distinct biochemical cleavage activities. These findings may eventually lead to pharmacological strategies for developing antiviral drugs against coronaviruses, rotaviruses, and other viruses.


Subject(s)
A Kinase Anchor Proteins/metabolism , Adenine Nucleotides/metabolism , Endoribonucleases/metabolism , Middle East Respiratory Syndrome Coronavirus/enzymology , Murine hepatitis virus/enzymology , Oligoribonucleotides/metabolism , Rotavirus/enzymology , Animals , Humans , Immunity, Innate/immunology , Interferons/immunology , Mice
2.
Viruses ; 11(2)2019 02 20.
Article in English | MEDLINE | ID: mdl-30791582

ABSTRACT

Viral gastroenteritis is an important cause of morbidity and mortality worldwide, being particularly severe for children under the age of five. The most common viral agents of gastroenteritis are noroviruses, rotaviruses, sapoviruses, astroviruses and adenoviruses, however, no specific antiviral treatment exists today against any of these pathogens. We here discuss the feasibility of developing a broad-spectrum antiviral treatment against these diarrhea-causing viruses. This review focuses on the viral polymerase as an antiviral target, as this is the most conserved viral protein among the diverse viral families to which these viruses belong to. We describe the functional and structural similarities of the different viral polymerases, the antiviral effect of reported polymerase inhibitors and highlight common features that might be exploited in an attempt of designing such pan-polymerase inhibitor.


Subject(s)
Antiviral Agents/isolation & purification , Diarrhea/drug therapy , Diarrhea/virology , Gastroenteritis/drug therapy , Gastroenteritis/virology , RNA-Dependent RNA Polymerase/metabolism , Adenovirus Infections, Human/drug therapy , Animals , Antiviral Agents/therapeutic use , DNA Viruses/drug effects , DNA Viruses/enzymology , Humans , Norovirus/drug effects , Norovirus/enzymology , Nucleic Acid Synthesis Inhibitors/isolation & purification , Nucleic Acid Synthesis Inhibitors/therapeutic use , RNA Viruses/drug effects , RNA Viruses/enzymology , Rotavirus/drug effects , Rotavirus/enzymology , Rotavirus Infections/drug therapy
3.
Vet Microbiol ; 207: 159-163, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28757018

ABSTRACT

Group A rotaviruses (RVAs) are divided into neuraminidase (NA)-sensitive and NA-insensitive strains depending upon their binding affinity to the VP8* domain in the terminal sialic acids (SAs) of cell surface carbohydrates. Although NA-sensitive strains are known to use terminal SAs as an attachment factor, the exact nature of this attachment factor is largely unknown. Here we show that the specific linkage of SA-containing glycan to glycoprotein or glycolipid is an attachment factor used by NA-sensitive porcine G9P[7] PRG9121 and G9P[23] PRG942, bovine G6P[1] NCDV, and canine G3P[3] strains. Infectivity of porcine G9P[7] and G9P[23] strains was markedly blocked by α2,3-linkage and α2,6-linkage inhibitors, indicating that these strains bind to both α2,3- and α2,6-linked SAs. However, the infectivity of bovine G6P[1] and canine G3P[3] strains was significantly reduced by α2,6-linkage inhibitor but not by α2,3-linkage blockers, demonstrating a predilection of these strains for α2,6-linked SAs. The infectivity of four NA-sensitive strains was equally reduced by inhibitors of lipid membrane and N-linked glycoprotein but not by an inhibitor of O-linked glycoprotein, indicating that these strains utilize both glycolipid and N-linked glycoprotein. Our study demonstrates that four NA-sensitive animal strains could have a strain-dependent binding preference toward α2,6-linked SAs (P[1] NCDV and P[3] CU-1 strains) or both α2,3- and α2,6-linked SAs (P[7] PRG9121 and P[23] PRG942 strains) to the glycolipid and N-linked glycoprotein.


Subject(s)
Membrane Glycoproteins/metabolism , Neuraminidase/metabolism , Rotavirus/enzymology , Animals , Cell Line , Fibroblasts/virology , Humans , Membrane Glycoproteins/chemistry , Rotavirus/genetics , Rotavirus/metabolism , Virus Attachment
4.
J Virol ; 91(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28100623

ABSTRACT

Temperature-sensitive (ts) mutants of simian rotavirus (RV) strain SA11 have been previously created to investigate the functions of viral proteins during replication. One mutant, SA11-tsC, has a mutation that maps to the gene encoding the VP1 polymerase and shows diminished growth and RNA synthesis at 39°C compared to that at 31°C. In the present study, we sequenced all 11 genes of SA11-tsC, confirming the presence of an L138P mutation in the VP1 N-terminal domain and identifying 52 additional mutations in four other viral proteins (VP4, VP7, NSP1, and NSP2). To investigate whether the L138P mutation induces a ts phenotype in VP1 outside the SA11-tsC genetic context, we employed ectopic expression systems. Specifically, we tested whether the L138P mutation affects the ability of VP1 to localize to viroplasms, which are the sites of RV RNA synthesis, by expressing the mutant form as a green fluorescent protein (GFP) fusion protein (VP1L138P-GFP) (i) in wild-type SA11-infected cells or (ii) in uninfected cells along with viroplasm-forming proteins NSP2 and NSP5. We found that VP1L138P-GFP localized to viroplasms and interacted with NSP2 and/or NSP5 at 31°C but not at 39°C. Next, we tested the enzymatic activity of a recombinant mutant polymerase (rVP1L138P) in vitro and found that it synthesized less RNA at 39°C than at 31°C, as well as less RNA than the control at all temperatures. Together, these results provide a mechanistic basis for the ts phenotype of SA11-tsC and raise important questions about the role of leucine 138 in supporting key protein interactions and the catalytic function of the VP1 polymerase.IMPORTANCE RVs cause diarrhea in the young of many animal species, including humans. Despite their medical and economic importance, gaps in knowledge exist about how these viruses replicate inside host cells. Previously, a mutant simian RV (SA11-tsC) that replicates worse at higher temperatures was identified. This virus has an amino acid mutation in VP1, which is the enzyme responsible for copying the viral RNA genome. The mutation is located in a poorly understood region of the polymerase called the N-terminal domain. In this study, we determined that the mutation reduces the ability of VP1 to properly localize within infected cells at high temperatures, as well as reduced the ability of the enzyme to copy viral RNA in a test tube. The results of this study explain the temperature sensitivity of SA11-tsC and shed new light on functional protein-protein interaction sites of VP1.


Subject(s)
Rotavirus/enzymology , Viral Core Proteins/genetics , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Enzyme Stability , Molecular Dynamics Simulation , Mutation, Missense , Protein Binding , Protein Domains , Protein Transport , Temperature , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism
5.
J Virol ; 89(13): 6633-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25878106

ABSTRACT

UNLABELLED: Synthesis of 2'-5'-oligoadenylates (2-5A) by oligoadenylate synthetase (OAS) is an important innate cellular response that limits viral replication by activating the latent cellular RNase, RNase L, to degrade single-stranded RNA. Some rotaviruses and coronaviruses antagonize the OAS/RNase L pathway through the activity of an encoded 2H phosphoesterase domain that cleaves 2-5A. These viral 2H phosphoesterases are phylogenetically related to the cellular A kinase anchoring protein 7 (AKAP7) and share a core structure and an active site that contains two well-defined HΦ(S/T)Φ (where Φ is a hydrophobic residue) motifs, but their mechanism of substrate binding is unknown. Here, we report the structures of a viral 2H phosphoesterase, the C-terminal domain (CTD) of the group A rotavirus (RVA) VP3 protein, both alone and in complex with 2-5A. The domain forms a compact fold, with a concave ß-sheet that contains the catalytic cleft, but it lacks two α-helical regions and two ß-strands observed in AKAP7 and other 2H phosphoesterases. The cocrystal structure shows significant conformational changes in the R loop upon ligand binding. Bioinformatics and biochemical analyses reveal that conserved residues and residues required for catalytic activity and substrate binding comprise the catalytic motifs and a region on one side of the binding cleft. We demonstrate that the VP3 CTD of group B rotavirus, but not that of group G, cleaves 2-5A. These findings suggest that the VP3 CTD is a streamlined version of a 2H phosphoesterase with a ligand-binding mechanism that is shared among 2H phosphodiesterases that cleave 2-5A. IMPORTANCE: The C-terminal domain (CTD) of rotavirus VP3 is a 2H phosphoesterase that cleaves 2'-5'-oligoadenylates (2-5A), potent activators of an important innate cellular antiviral pathway. 2H phosphoesterase superfamily proteins contain two conserved catalytic motifs and a proposed core structure. Here, we present structures of a viral 2H phosphoesterase, the rotavirus VP3 CTD, alone and in complex with its substrate, 2-5A. The domain lacks two α-helical regions and ß-strands present in other 2H phosphoesterases. A loop of the protein undergoes significant structural changes upon substrate binding. Together with our bioinformatics and biochemical findings, the crystal structures suggest that the RVA VP3 CTD domain is a streamlined version of a cellular enzyme that shares a ligand-binding mechanism with other 2H phosphodiesterases that cleave 2-5A but differs from those of 2H phosphodiesterases that cleave other substrates. These findings may aid in the future design of antivirals targeting viral phosphodiesterases with cleavage specificity for 2-5A.


Subject(s)
Adenine Nucleotides/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Exoribonucleases/chemistry , Exoribonucleases/metabolism , Oligoribonucleotides/metabolism , Catalytic Domain , Crystallography, X-Ray , Protein Binding , Protein Conformation , Rotavirus/enzymology
6.
Proteins ; 83(5): 997-1002, 2015 May.
Article in English | MEDLINE | ID: mdl-25758703

ABSTRACT

In response to viral infections, the mammalian innate immune system induces the production of the second messenger 2'-5' oligoadenylate (2-5A) to activate latent ribonuclease L (RNase L) that restricts viral replication and promotes apoptosis. A subset of rotaviruses and coronaviruses encode 2',5'-phosphodiesterase enzymes that hydrolyze 2-5A, thereby inhibiting RNase L activation. We report the crystal structure of the 2',5'-phosphodiesterase domain of group A rotavirus protein VP3 at 1.39 Å resolution. The structure exhibits a 2H phosphoesterase fold and reveals conserved active site residues, providing insights into the mechanism of 2-5A degradation in viral evasion of host innate immunity.


Subject(s)
Capsid Proteins/chemistry , Phosphoric Diester Hydrolases/chemistry , Rotavirus/enzymology , Catalytic Domain , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Protein Structure, Secondary
7.
mBio ; 6(1)2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25626907

ABSTRACT

UNLABELLED: NF-κB plays a critical role in the induction and maintenance of innate and adaptive immune transcriptional programs. An associated inhibitor of κB protein (IκB) regulates NF-κB activation and contains a degron motif (DSGΦxS) that undergoes phosphorylation following pathogen recognition or other proinflammatory signals. The E3 ubiquitin ligase SCF(ß-TrCP) recognizes this phosphodegron through its ß-transducin repeat-containing protein (ß-TrCP) subunit and induces IκB degradation, allowing NF-κB to translocate to the nucleus and modulate gene expression. Rotavirus (RV), a major cause of pediatric gastroenteritis, can block NF-κB activation through the action of its nonstructural protein NSP1, a putative E3 ubiquitin ligase that mediates the degradation of ß-TrCP or other immunomodulatory proteins in a virus strain-specific manner. Here, we show that NSP1 targets ß-TrCP by mimicking the IκB phosphodegron. The NSP1 proteins of most human and porcine RV strains conserve a C-terminal phosphodegron-like (PDL) motif, DSGΦS. Deletion of this motif or mutation of its serine residues disrupts NSP1-mediated degradation of ß-TrCP and inhibition of NF-κB activation. Additionally, a point mutation within the phosphodegron-binding pocket protects ß-TrCP from NSP1-mediated turnover. Fusion of the PDL motif to an NSP1 protein known to target other immunomodulatory proteins generates a chimeric NSP1 protein that can induce ß-TrCP degradation and block NF-κB activation. Other viral proteins (Epstein-Barr virus LMP1, HIV-1 Vpu, and vaccinia virus A49) also contain a PDL motif and interact with ß-TrCP to inhibit NF-κB activation. Taken together, these data suggest that targeting ß-TrCP by molecular mimicry may be a common strategy used by human viruses to evade the host immune response. IMPORTANCE: The transcription factor NF-κB, a central regulator of the host response to infection, is a frequent target of viral antagonism. Pathogen detection activates NF-κB by inducing the phosphorylation of an associated inhibitor protein (IκB), which targets IκB for degradation by the E3 ubiquitin ligase ß-TrCP. Rotavirus, a significant cause of childhood gastroenteritis, antagonizes NF-κB through the activity of its NSP1 protein, a putative E3 ubiquitin ligase that mediates ß-TrCP turnover. Here, we show that NSP1 functions by mimicking the IκB phosphodegron recognized by ß-TrCP. Nearly all human rotavirus strains conserve this motif at the NSP1 C terminus, and its removal disrupts NSP1 antagonist activity. This sequence conserves the biochemical properties of the IκB phosphodegron and can rescue antagonist activity when fused to an NSP1 protein otherwise inactive against ß-TrCP. Other viral proteins also mimic IκB to disrupt NF-κB activation, indicating that this is an important immune evasion strategy.


Subject(s)
NF-kappa B/metabolism , Rotavirus Infections/virology , Rotavirus/enzymology , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Amino Acid Sequence , Host-Pathogen Interactions , Humans , Molecular Mimicry , Molecular Sequence Data , NF-kappa B/chemistry , NF-kappa B/genetics , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proteolysis , Rotavirus/chemistry , Rotavirus/genetics , Rotavirus Infections/genetics , Rotavirus Infections/metabolism , Sequence Alignment , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , beta-Transducin Repeat-Containing Proteins/chemistry , beta-Transducin Repeat-Containing Proteins/genetics
8.
J Mol Biol ; 425(1): 124-32, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23089332

ABSTRACT

Double-stranded RNA (dsRNA) viruses transcribe and replicate RNA within an assembled, inner capsid particle; only plus-sense mRNA emerges into the intracellular milieu. During infectious entry of a rotavirus particle, the outer layer of its three-layer structure dissociates, delivering the inner double-layered particle (DLP) into the cytosol. DLP structures determined by X-ray crystallography and electron cryomicroscopy (cryoEM) show that the RNA coils uniformly into the particle interior, avoiding a "fivefold hub" of more structured density projecting inward from the VP2 shell of the DLP along each of the twelve 5-fold axes. Analysis of the X-ray crystallographic electron density map suggested that principal contributors to the hub are the N-terminal arms of VP2, but reexamination of the cryoEM map has shown that many features come from a molecule of VP1, randomly occupying five equivalent and partly overlapping positions. We confirm here that the electron density in the X-ray map leads to the same conclusion, and we describe the functional implications of the orientation and position of the polymerase. The exit channel for the nascent transcript directs the nascent transcript toward an opening along the 5-fold axis. The template strand enters from within the particle, and the dsRNA product of the initial replication step exits in a direction tangential to the inner surface of the VP2 shell, allowing it to coil optimally within the DLP. The polymerases of reoviruses appear to have similar positions and functional orientations.


Subject(s)
Capsid/chemistry , Rotavirus/enzymology , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Image Processing, Computer-Assisted , Models, Molecular , RNA, Double-Stranded/genetics , RNA, Messenger/genetics , RNA, Viral/genetics , Rotavirus/chemistry , Rotavirus/genetics , Rotavirus/ultrastructure , Viral Core Proteins/genetics , Virus Assembly
9.
Virology ; 431(1-2): 12-20, 2012.
Article in English | MEDLINE | ID: mdl-22664357

ABSTRACT

The rotavirus RNA-dependent RNA polymerase (RdRp), VP1, contains canonical RdRp motifs and a priming loop that is hypothesized to undergo conformational rearrangements during RNA synthesis. In the absence of viral core shell protein VP2, VP1 fails to interact stably with divalent cations or nucleotides and has a retracted priming loop. To identify residues of potential import to nucleotide and divalent cation stabilization, we aligned VP1 of divergent rotaviruses and the structural homolog reovirus λ3. VP1 mutants were engineered and characterized for RNA synthetic capacity in vitro. Conserved aspartic acids in RdRp motifs A and C and arginines in motif F that likely stabilize divalent cations and nucleotides were required for efficient RNA synthesis. Mutation of individual priming loop residues diminished or enhanced RNA synthesis efficiency without obviating the need for VP2, which suggests that this structure serves as a dynamic regulatory element that links RdRp activity to particle assembly.


Subject(s)
Cations, Divalent/metabolism , Nucleotides/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Rotavirus/enzymology , Viral Core Proteins/chemistry , Viral Core Proteins/metabolism , Amino Acid Sequence , Mammalian orthoreovirus 3/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , RNA/metabolism , RNA-Dependent RNA Polymerase/genetics , Rotavirus/chemistry , Rotavirus/genetics , Sequence Alignment , Viral Core Proteins/genetics
10.
Virology ; 431(1-2): 50-7, 2012.
Article in English | MEDLINE | ID: mdl-22687427

ABSTRACT

Rotaviruses (RVs) are segmented double-stranded RNA viruses that cause gastroenteritis in mammals and birds. Within the RV genus, eight species (RVA-RVH) have been proposed. Here, we report the first RVF and RVG sequences for the viral RNA polymerase (VP1)-encoding segments and compare them to those of other RV species. Phylogenetic analyses indicate that the VP1 RNA segments and proteins resolve into two major clades, with RVA, RVC, RVD and RVF in clade A, and RVB, RVG and RVH in clade B. Plus-strand RNA of clade A viruses, and not clade B viruses, contain a 3'-proximal UGUG cassette that serves as the VP1 recognition signal. VP1 structures for a representative of each RV species were predicted using homology modeling. Structural elements involved in interactions with the UGUG cassette were conserved among VP1 of clade A, suggesting a conserved mechanism of viral RNA recognition for these viruses.


Subject(s)
DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Rotavirus/enzymology , Rotavirus/genetics , Templates, Genetic , Viral Core Proteins/genetics , Viral Core Proteins/metabolism , Animals , Birds , Cluster Analysis , Mammals , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , RNA, Viral/genetics , Sequence Analysis, DNA
11.
J Virol ; 85(5): 1958-69, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21147920

ABSTRACT

To replicate its segmented, double-stranded RNA (dsRNA) genome, the rotavirus RNA-dependent RNA polymerase, VP1, must recognize viral plus-strand RNAs (+RNAs) and guide them into the catalytic center. VP1 binds to the conserved 3' end of rotavirus +RNAs via both sequence-dependent and sequence-independent contacts. Sequence-dependent contacts permit recognition of viral +RNAs and specify an autoinhibited positioning of the template within the catalytic site. However, the contributions to dsRNA synthesis of sequence-dependent and sequence-independent VP1-RNA interactions remain unclear. To analyze the importance of VP1 residues that interact with +RNA on genome replication, we engineered mutant VP1 proteins and assayed their capacity to synthesize dsRNA in vitro. Our results showed that, individually, mutation of residues that interact specifically with RNA bases did not diminish replication levels. However, simultaneous mutations led to significantly lower levels of dsRNA product, presumably due to impaired recruitment of +RNA templates. In contrast, point mutations of sequence-independent RNA contact residues led to severely diminished replication, likely as a result of improper positioning of templates at the catalytic site. A noteworthy exception was a K419A mutation that enhanced the initiation capacity and product elongation rate of VP1. The specific chemistry of Lys419 and its position at a narrow region of the template entry tunnel appear to contribute to its capacity to moderate replication. Together, our findings suggest that distinct classes of VP1 residues interact with +RNA to mediate template recognition and dsRNA synthesis yet function in concert to promote viral RNA replication at appropriate times and rates.


Subject(s)
Genome, Viral , RNA-Dependent RNA Polymerase/metabolism , Rotavirus/enzymology , Viral Proteins/metabolism , Virus Replication , Amino Acid Sequence , Animals , Cell Line , DNA Replication , Models, Molecular , Molecular Sequence Data , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Rotavirus/chemistry , Rotavirus/genetics , Rotavirus/physiology , Templates, Genetic , Viral Proteins/chemistry , Viral Proteins/genetics
12.
Article in Chinese | MEDLINE | ID: mdl-21604574

ABSTRACT

OBJECTIVE: To confirm the activity of non structural protein 1 (NSP1) of Rotavirus (RV) as E3 ubiquitin ligase by experiments and to provide some clues for NSP1 on the pathogenic mechanisms and replication of RV. METHODS: The whole gene and RING deleted mutation gene of NSP1 were coloned into pEGFPC1 expression plasmid, and transfected into human embryonic kidney (HEK) 293 FT cells with pBlue-Script-HA-Ubiquitin. The expression of proteins were proved by using con-focal microscope and western blotting. The ubiquination of proteins were detected by co-immunoprecite. RESULTS: The cellular proteins of HEK293FT are ubiquinated by NSP1 protein and NSP1 protein was self-ubiquinated also. CONCLUSIONS: It revealed that RV NSP1 had the activity of E3 ubiquitin ligase and it may play a role on the modulate mechanisms of ubiquination.


Subject(s)
Rotavirus/enzymology , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/metabolism , HEK293 Cells , Humans , Rotavirus/genetics , Viral Nonstructural Proteins/genetics
13.
Structure ; 16(11): 1601-2, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19000811

ABSTRACT

In this issue of Structure, Lu et al. (2008) report results of structural and functional analysis of rotavirus RNA-dependent RNA polymerase, VP1. Based on their analyses of VP1 in RNA free and bound forms, the authors propose a mechanism for coordinated genome packaging and replication.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Rotavirus/genetics , Models, Molecular , RNA, Viral/chemistry , Rotavirus/enzymology
14.
Structure ; 16(11): 1678-88, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19000820

ABSTRACT

Rotavirus RNA-dependent RNA polymerase VP1 catalyzes RNA synthesis within a subviral particle. This activity depends on core shell protein VP2. A conserved sequence at the 3' end of plus-strand RNA templates is important for polymerase association and genome replication. We have determined the structure of VP1 at 2.9 A resolution, as apoenzyme and in complex with RNA. The cage-like enzyme is similar to reovirus lambda3, with four tunnels leading to or from a central, catalytic cavity. A distinguishing characteristic of VP1 is specific recognition, by conserved features of the template-entry channel, of four bases, UGUG, in the conserved 3' sequence. Well-defined interactions with these bases position the RNA so that its 3' end overshoots the initiating register, producing a stable but catalytically inactive complex. We propose that specific 3' end recognition selects rotavirus RNA for packaging and that VP2 activates the autoinhibited VP1/RNA complex to coordinate packaging and genome replication.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Rotavirus/genetics , Apoenzymes/chemistry , Base Sequence , Binding Sites , Models, Molecular , Nucleic Acid Conformation , Oligoribonucleotides/chemistry , Protein Conformation , RNA, Viral/chemistry , Rotavirus/enzymology
15.
J Gen Virol ; 88(Pt 10): 2800-2810, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17872534

ABSTRACT

The rotavirus (RV) non-structural protein 5, NSP5, is encoded by the smallest of the 11 genomic segments and localizes in 'viroplasms', cytoplasmic inclusion bodies in which viral RNA replication and packaging take place. NSP5 is essential for the replicative cycle of the virus because, in its absence, viroplasms are not formed and viral RNA replication and transcription do not occur. NSP5 is produced early in infection and undergoes a complex hyperphosphorylation process, leading to the formation of proteins differing in electrophoretic mobility. The role of hyperphosphorylation of NSP5 in the replicative cycle of rotavirus is unknown. Previous in vitro studies have suggested that the cellular kinase CK1alpha is responsible for the NSP5 hyperphosphorylation process. Here it is shown, by means of specific RNA interference, that in vivo, CK1alpha is the enzyme that initiates phosphorylation of NSP5. Lack of NSP5 hyperphosphorylation affected neither its interaction with the virus VP1 and NSP2 proteins normally found in viroplasms, nor the production of viral proteins. In contrast, the morphology of viroplasms was altered markedly in cells in which CK1alpha was depleted and a moderate decrease in the production of double-stranded RNA and infectious virus was observed. These data show that CK1alpha is the kinase that phosphorylates NSP5 in virus-infected cells and contribute to further understanding of the role of NSP5 in RV infection.


Subject(s)
Casein Kinase Ialpha/deficiency , Rotavirus/enzymology , Virus Replication/physiology , Genes, Reporter , Phosphorylation , Plasmids , RNA, Small Interfering/genetics , RNA, Viral/genetics , Rotavirus/genetics , Rotavirus/physiology , Rotavirus/ultrastructure , Transfection , Viral Nonstructural Proteins/metabolism , Viral Proteins/genetics
16.
J Virol ; 81(22): 12272-84, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17804496

ABSTRACT

Rotavirus, the major pathogen of infantile gastroenteritis, carries a nonstructural protein, NSP2, essential for viroplasm formation and genome replication/packaging. In addition to RNA-binding and helix-destabilizing properties, NSP2 exhibits nucleoside triphosphatase activity. A conserved histidine (H225) functions as the catalytic residue for this enzymatic activity, and mutation of this residue abrogates genomic double-stranded RNA synthesis without affecting viroplasm formation. To understand the structural basis of the phosphatase activity of NSP2, we performed crystallographic analyses of native NSP2 and a functionally defective H225A mutant in the presence of nucleotides. These studies showed that nucleotides bind inside a cleft between the two domains of NSP2 in a region that exhibits structural similarity to ubiquitous cellular HIT (histidine triad) proteins. Only minor conformational alterations were observed in the cleft upon nucleotide binding and hydrolysis. This hydrolysis involved the formation of a stable phosphohistidine intermediate. These observations, reminiscent of cellular nucleoside diphosphate (NDP) kinases, prompted us to investigate whether NSP2 exhibits phosphoryl-transfer activity. Bioluminometric assay showed that NSP2 exhibits an NDP kinase-like activity that transfers the bound phosphate to NDPs. However, NSP2 is distinct from the highly conserved cellular NDP kinases in both its structure and catalytic mechanism, thus making NSP2 a potential target for antiviral drug design. With structural similarities to HIT proteins, which are not known to exhibit NDP kinase activity, NSP2 represents a unique example among structure-activity relationships. The newly observed phosphoryl-transfer activity of NSP2 may be utilized for homeostasis of nucleotide pools in viroplasms during genome replication.


Subject(s)
Nucleoside-Diphosphate Kinase/chemistry , Nucleotides/chemistry , RNA-Binding Proteins/chemistry , Viral Nonstructural Proteins/chemistry , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalysis , Catalytic Domain , Humans , Kinetics , Molecular Sequence Data , Nucleoside-Diphosphate Kinase/genetics , Nucleoside-Diphosphate Kinase/physiology , Phosphorylation , Protein Conformation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , Rotavirus/enzymology , Rotavirus/physiology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Virus Replication
17.
J Biotechnol ; 122(4): 443-52, 2006 Apr 20.
Article in English | MEDLINE | ID: mdl-16290307

ABSTRACT

The production of virus-like particles (VLP) is of interest to several fields. However, little is known about their assembly when they are expressed in insect cells, as it occurs in conditions different to those of native virus. Knowledge of the localization of recombinant proteins and of the site of accumulation of VLP can increase the understanding of VLP assembly and be useful for proposing production strategies. In this work, the rotavirus proteins VP6 and the fusion protein GFPVP2 were expressed in High Five insect cells. Recombinant proteins and rotavirus-like particles (RLP) were located and visualized by confocal, epifluorescence and electron microscopy. Single-layered (sl) RLP (conformed by GFPVP2) accumulated in the cytoplasm as highly ordered aggregates. In contrast, VP6 formed fibrillar structures composed of various tubes of VP6 that were not associated to microtubules. Coexpression of GFPVP2 and VP6 altered the distribution of both proteins. VP6 formed aggregates, even when all other conditions of individual protein expression remained unchanged. Double-layered (dl) RLP were observed in dense zones of the cytoplasm, but were not in ordered aggregates. It was determined that the assembly of both slRLP and dlRLP occurs intracellularly. Accordingly, strategies for the optimum assembly of dlRLP should guarantee that each cell produces both recombinant proteins.


Subject(s)
Antigens, Viral/metabolism , Baculoviridae/metabolism , Capsid Proteins/metabolism , Cell Nucleus/metabolism , Rotavirus/metabolism , Animals , Baculoviridae/genetics , Fluorescent Antibody Technique , Genetic Vectors/metabolism , Insecta/cytology , Insecta/metabolism , Insecta/virology , Microscopy, Electron , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rotavirus/enzymology
18.
J Gen Virol ; 85(Pt 4): 929-932, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039535

ABSTRACT

The RNA segment encoding the guanylyltransferase (VP3) from 12 group A rotavirus isolates has been sequenced following RT-PCR and molecular cloning of the full-length amplicons produced. Alignment of the derived amino acid sequences including those of the four VP3 sequences available from GenBank revealed two levels of sequence divergence. Virus isolates from humans showed greater than 94% sequence identity, whereas those isolated from different mammalian species showed as low as 79% sequence identity. The exceptions were avian virus isolates, which diverged approximately 45% from those of mammalian origin, and the human virus isolates DS1 and 69M, which showed much closer (over 90%) identity to viruses of bovine origin, suggesting that these human isolates may have undergone recent reassortment events with a bovine virus. Analysis of the sequences for a putative enzymic active site has revealed that the KXTAMDXEXP and KXXGNNH motifs around amino acids 385 and 545, respectively, are conserved across both group A and C rotaviruses.


Subject(s)
Capsid Proteins/genetics , Nucleotidyltransferases/genetics , Rotavirus/enzymology , Rotavirus/genetics , Amino Acid Sequence , Animals , Base Sequence , Birds , Cattle , Conserved Sequence , DNA, Viral/genetics , Humans , Mammals , Molecular Sequence Data , Phylogeny , Rotavirus/classification , Rotavirus/isolation & purification , Sequence Homology, Amino Acid
19.
J Virol ; 76(7): 3461-70, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11884570

ABSTRACT

Rotavirus NSP5 is a nonstructural protein that localizes in cytoplasmic viroplasms of infected cells. NSP5 interacts with NSP2 and undergoes a complex posttranslational hyperphosphorylation, generating species with reduced polyacrylamide gel electrophoresis mobility. This process has been suggested to be due in part to autophosphorylation. We developed an in vitro phosphorylation assay using as a substrate an in vitro-translated NSP5 deletion mutant that was phosphorylated by extracts from MA104 cells transfected with NSP5 mutants but not by extracts from mock-transfected cells. The phosphorylated products obtained showed shifts in mobility similar to what occurs in vivo. From these and other experiments we concluded that NSP5 activates a cellular kinase(s) for its own phosphorylation. Three NSP5 regions were found to be essential for kinase(s) activation. Glutathione S-transferase-NSP5 mutants were produced in Escherichia coli and used to determine phosphoacceptor sites. These were mapped to four serines (Ser(153), Ser(155), Ser(163), and Ser(165)) within an acidic region with homology to casein kinase II (CKII) phosphorylation sites. CKII was able to phosphorylate NSP5 in vitro. NSP5 and its mutants fused to enhanced green fluorescent protein were used in transfection experiments followed by virus infection and allowed the determination of the domains essential for viroplasm localization in the context of virus infection.


Subject(s)
Phosphotransferases/metabolism , Rotavirus/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Binding Sites , Casein Kinase II , Cell Line , Enzyme Activation , Fluorescent Antibody Technique , Glutathione Transferase/genetics , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Rotavirus/enzymology , Serine/metabolism , Transfection , Viral Nonstructural Proteins/chemistry
20.
FEMS Microbiol Lett ; 198(2): 147-50, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11430406

ABSTRACT

Gene 1 (which encodes the viral RNA-dependent RNA polymerase, VP1) of an atypical human reassortant rotavirus strain, E210 (serotype G2P1B), is unrelated to genes 1 of standard human rotaviruses. To ascertain the origin of this gene, we determined a partial sequence and found that it exhibited greatest identity to gene 1 of a Taiwanese isolate, TE83, which is representative of G2 strains that caused an epidemic of gastroenteritis in 1993. Limited sequence identity to genes 1 of standard human and animal viruses was observed. This was confirmed by phylogenetic analysis. However, hybridization analysis using an E210 gene 1-specific probe indicated that a related gene was found among other Australian G2 isolates and in a Japanese strain isolated in the 1970s.


Subject(s)
DNA-Directed RNA Polymerases/genetics , Phylogeny , Rotavirus/classification , Rotavirus/genetics , Viral Core Proteins/genetics , Australia , Genes, Viral , Humans , RNA, Double-Stranded/genetics , RNA, Double-Stranded/isolation & purification , RNA, Viral/genetics , RNA, Viral/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Rotavirus/enzymology , Taiwan , Viral Structural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...