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











Publication year range
1.
Viruses ; 16(5)2024 05 03.
Article in English | MEDLINE | ID: mdl-38793607

ABSTRACT

The dengue virus is a single-stranded, positive-sense RNA virus that infects ~400 million people worldwide. Currently, there are no approved antivirals available. CRISPR-based screening methods have greatly accelerated the discovery of host factors that are essential for DENV infection and that can be targeted in host-directed antiviral interventions. In the present study, we performed a focused CRISPR (Clustered Regularly Interspaced Palindromic Repeats) library screen to discover the key host factors that are essential for DENV infection in human Huh7 cells and identified the Protein Activator of Interferon-Induced Protein Kinase (PACT) as a novel pro-viral factor for DENV. PACT is a double-stranded RNA-binding protein generally known to activate antiviral responses in virus-infected cells and block viral replication. However, in our studies, we observed that PACT plays a pro-viral role in DENV infection and specifically promotes viral RNA replication. Knockout of PACT resulted in a significant decrease in DENV RNA and protein abundances in infected cells, which was rescued upon ectopic expression of full-length PACT. An analysis of global gene expression changes indicated that several ER-associated pro-viral genes such as ERN1, DDIT3, HERPUD1, and EIF2AK3 are not upregulated in DENV-infected PACT knockout cells as compared to infected wildtype cells. Thus, our study demonstrates a novel role for PACT in promoting DENV replication, possibly through modulating the expression of ER-associated pro-viral genes.


Subject(s)
CRISPR-Cas Systems , Dengue Virus , Host-Pathogen Interactions , RNA-Binding Proteins , Virus Replication , Humans , Cell Line , Dengue/virology , Dengue Virus/physiology , Host-Pathogen Interactions/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
2.
Cell Rep ; 42(4): 112389, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37058406

ABSTRACT

Enterovirus A71 (EV-A71) causes hand, foot, and mouth disease outbreaks with neurological complications and deaths. We previously isolated an EV-A71 variant in the stool, cerebrospinal fluid, and blood of an immunocompromised patient who had a leucine-to-arginine substitution on the VP1 capsid protein, resulting in increased heparin sulfate binding. We show here that this mutation increases the virus's pathogenicity in orally infected mice with depleted B cells, which mimics the patient's immune status, and increases susceptibility to neutralizing antibodies. However, a double mutant with even greater heparin sulfate affinity is not pathogenic, suggesting that increased heparin sulfate affinity may trap virions in peripheral tissues and reduce neurovirulence. This research sheds light on the increased pathogenicity of variant with heparin sulfate (HS)-binding ability in individuals with decreased B cell immunity.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Enterovirus , Humans , Animals , Mice , Enterovirus/genetics , Enterovirus A, Human/genetics , Antigens, Viral/metabolism , Heparitin Sulfate/metabolism , Heparin/metabolism
3.
PLoS Pathog ; 18(4): e1010163, 2022 04.
Article in English | MEDLINE | ID: mdl-35482886

ABSTRACT

Arthropod-borne viruses infect both mosquito and mammalian hosts. While much is known about virus-host interactions that modulate viral gene expression in their mammalian host, much less is known about the interactions that involve inhibition, subversion or avoidance strategies in the mosquito host. A novel RNA-Protein interaction detection assay was used to detect proteins that directly or indirectly bind to dengue viral genomes in infected mosquito cells. Membrane-associated mosquito proteins Sec61A1 and Loquacious (Loqs) were found to be in complex with the viral RNA. Depletion analysis demonstrated that both Sec61A1 and Loqs have pro-viral functions in the dengue viral infectious cycle. Co-localization and pull-down assays showed that Loqs interacts with viral protein NS3 and both full-length and subgenomic viral RNAs. While Loqs coats the entire positive-stranded viral RNA, it binds selectively to the 3' end of the negative-strand of the viral genome. In-depth analyses showed that the absence of Loqs did not affect translation or turnover of the viral RNA but modulated viral replication. Loqs also displayed pro-viral functions for several flaviviruses in infected mosquito cells, suggesting a conserved role for Loqs in flavivirus-infected mosquito cells.


Subject(s)
Culicidae , Dengue , Flavivirus , Animals , Flavivirus/physiology , Mammals , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication
4.
J Virol ; 95(21): e0089721, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34379497

ABSTRACT

Enterovirus A71 (EV-A71) and many members of the Picornaviridae family are neurotropic pathogens of global concern. These viruses are primarily transmitted through the fecal-oral route, and thus suitable animal models of oral infection are needed to investigate viral pathogenesis. An animal model of oral infection was developed using transgenic mice expressing human SCARB2 (hSCARB2 Tg), murine-adapted EV-A71/MP4 virus, and EV-A71/MP4 virus with an engineered nanoluciferase gene that allows imaging of viral replication and spread in infected mice. Next-generation sequencing of EV-A71 genomes in the tissues and organs of infected mice was also performed. Oral inoculation of EV-A71/MP4 or nanoluciferase-carrying MP4 virus stably induced neurological symptoms and death in infected 21-day-old weaned mice. In vivo bioluminescence imaging of infected mice and tissue immunostaining of viral antigens indicated that orally inoculated virus can spread to the central nervous system (CNS) and other tissues. Next-generating sequencing further identified diverse mutations in viral genomes that can potentially contribute to viral pathogenesis. This study presents an EV-A71 oral infection murine model that efficiently infects weaned mice and allows tracking of viral spread, features that can facilitate research into viral pathogenesis and neuroinvasion via the natural route of infection. IMPORTANCE Enterovirus A71 (EV-A71), a positive-strand RNA virus of the Picornaviridae, poses a persistent global public health problem. EV-A71 is primarily transmitted through the fecal-oral route, and thus suitable animal models of oral infection are needed to investigate viral pathogenesis. We present an animal model of EV-A71 infection that enables the natural route of oral infection in weaned and nonimmunocompromised 21-day-old hSCARB2 transgenic mice. Our results demonstrate that severe disease and death could be stably induced, and viral invasion of the CNS could be replicated in this model, similar to severe real-world EV-A71 infections. We also developed a nanoluciferase-containing EV-A71 virus that can be used with this animal model to track viral spread after oral infection in real time. Such a model offers several advantages over existing animal models and can facilitate future research into viral spread, tissue tropism, and viral pathogenesis, all pressing issues that remain unaddressed for EV-A71 infections.


Subject(s)
Central Nervous System/virology , Enterovirus A, Human/pathogenicity , Enterovirus Infections/complications , Lysosomal Membrane Proteins/genetics , Mouth/virology , Nervous System Diseases/virology , Receptors, Scavenger/genetics , Animals , Disease Models, Animal , Enterovirus A, Human/genetics , Enterovirus Infections/pathology , Enterovirus Infections/virology , Genome, Viral , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Viral Tropism , Virus Replication , Weaning
5.
PLoS One ; 14(9): e0221048, 2019.
Article in English | MEDLINE | ID: mdl-31498791

ABSTRACT

Enterovirus 71 (EV71) induces apoptosis to promote viral particle release. Earlier work showed that EV71 utilizes its 3C protease to induce apoptosis in a caspase-3-dependent pathway, though the mechanism is unknown. However, work from Vagner, Holcik and colleagues showed that host protein heterogeneous ribonucleoprotein A1 (hnRNP A1) binds the IRES of cellular apoptotic peptidase activating factor 1 (apaf-1) mRNA to repress its translation. In this work, we show that apaf-1 expression is essential for EV71-induced apoptosis. EV71 infection or ectopic expression of 3C protease cleaves hnRNP A1, which abolishes its binding to the apaf-1 IRES. This allows IRES-dependent synthesis of apaf-1, activation of caspase-3, and apoptosis. Thus, we reveal a novel mechanism that EV71 utilizes for virus release via a 3C protease-hnRNP A1-apaf-1-caspase-3-apoptosis axis.


Subject(s)
Apoptotic Protease-Activating Factor 1/genetics , Caspase 3/genetics , Cysteine Endopeptidases/genetics , Enterovirus A, Human/genetics , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Protein Biosynthesis , Viral Proteins/genetics , 3C Viral Proteases , Animals , Apoptosis/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Cysteine Endopeptidases/metabolism , Enterovirus A, Human/metabolism , Gene Expression Regulation , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Host-Pathogen Interactions/genetics , Humans , Internal Ribosome Entry Sites , Muscle Cells/metabolism , Muscle Cells/virology , Neuroglia/metabolism , Neuroglia/virology , Protein Binding , Proteolysis , Signal Transduction , Vero Cells , Viral Proteins/metabolism
6.
Nat Microbiol ; 4(12): 2523-2537, 2019 12.
Article in English | MEDLINE | ID: mdl-31527793

ABSTRACT

Enteroviruses (EVs) comprise a large genus of positive-sense, single-stranded RNA viruses whose members cause a number of important and widespread human diseases, including poliomyelitis, myocarditis, acute flaccid myelitis and the common cold. How EVs co-opt cellular functions to promote replication and spread is incompletely understood. Here, using genome-scale CRISPR screens, we identify the actin histidine methyltransferase SET domain containing 3 (SETD3) as critically important for viral infection by a broad panel of EVs, including rhinoviruses and non-polio EVs increasingly linked to severe neurological disease such as acute flaccid myelitis (EV-D68) and viral encephalitis (EV-A71). We show that cytosolic SETD3, independent of its methylation activity, is required for the RNA replication step in the viral life cycle. Using quantitative affinity purification-mass spectrometry, we show that SETD3 specifically interacts with the viral 2A protease of multiple enteroviral species, and we map the residues in 2A that mediate this interaction. 2A mutants that retain protease activity but are unable to interact with SETD3 are severely compromised in RNA replication. These data suggest a role of the viral 2A protein in RNA replication beyond facilitating proteolytic cleavage. Finally, we show that SETD3 is essential for in vivo replication and pathogenesis in multiple mouse models for EV infection, including CV-A10, EV-A71 and EV-D68. Our results reveal a crucial role of a host protein in viral pathogenesis, and suggest targeting SETD3 as a potential mechanism for controlling viral infections.


Subject(s)
Enterovirus/metabolism , Enterovirus/pathogenicity , Histone Methyltransferases/metabolism , Methyltransferases/metabolism , Animals , CRISPR-Cas Systems , Central Nervous System Viral Diseases/virology , Disease Models, Animal , Encephalitis, Viral , Enterovirus/genetics , Enterovirus Infections/virology , Histone Methyltransferases/genetics , Mice , Myelitis/virology , Neuromuscular Diseases/virology , Proteolysis , Viral Proteins , Virus Replication
7.
Sci Rep ; 6: 24149, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27052555

ABSTRACT

Human enterovirus 71 (EV71) is a major causative agent of hand, foot, and, mouth disease, accounting for more than 65% of recent outbreaks. Following enteroviral infection, the host responses are crucial indicators for the development of a diagnosis regarding the clinical severity of EV71 infections. In this study, we implemented NanoString nCounter technology to characterize the responses of serum microRNA (miRNA) profiles to various EV71 infection diseases. Upon EV71 infection, 44 miRNAs were observed in patients with EV71 infections, with at least a 2-fold elevation and 133 miRNAs with a 2-fold reduction compared with the same miRNAs in healthy controls. Further detailed work with miR876-5p, a 9.5-fold change of upregulated miR-876-5p expression was observed in cases with severe EV71 symptoms, revealed that in vitro and in vivo knockdown of miR876-5p reduced viral RNA in cultured cells, and attenuated the severity of symptoms in EV71-infected mice. Altogether, we demonstrated that the elevated expression of circulating miR876-5p is a specific response to severe EV71 infections.


Subject(s)
Enterovirus A, Human/genetics , Enterovirus Infections/genetics , MicroRNAs/genetics , Up-Regulation , Animals , Cell Line , Cell Line, Tumor , Child , Child, Preschool , Dogs , Enterovirus A, Human/physiology , Enterovirus Infections/blood , Enterovirus Infections/virology , Female , Gene Expression Profiling/methods , Gene Knockdown Techniques , Gene Regulatory Networks , Host-Pathogen Interactions/genetics , Humans , Infant , Madin Darby Canine Kidney Cells , Male , Mice, Inbred ICR , MicroRNAs/blood , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
Wiley Interdiscip Rev RNA ; 7(5): 575-88, 2016 09.
Article in English | MEDLINE | ID: mdl-27046163

ABSTRACT

RNA virus infection in plants and invertebrates can produce virus-derived small RNAs. These RNAs share features with host endogenous small interfering RNAs (siRNAs). They can potentially mediate RNA interference (RNAi) and related RNA silencing pathways, resulting in specific antiviral defense. Although most RNA silencing components such as Dicer, Ago2, and RISC are conserved among eukaryotic hosts, whether RNA virus infection in mammals can generate functional small RNAs that act in antiviral defense remains under discussion. Here, we review recent studies on the molecular and biochemical features of viral siRNAs and other virus-derived small RNAs from infected plants, arthropods, nematodes, and vertebrates and discuss the genetic pathways for their biogenesis and their roles in antiviral activity. WIREs RNA 2016, 7:575-588. doi: 10.1002/wrna.1351 For further resources related to this article, please visit the WIREs website.


Subject(s)
Gene Silencing , Host-Pathogen Interactions , RNA Viruses/immunology , RNA, Small Untranslated/metabolism , RNA, Viral/metabolism , Animals , Invertebrates , Plants , Vertebrates
9.
Microbes Infect ; 17(8): 557-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25980760

ABSTRACT

The role of virus-derived small RNAs (vsRNAs) has been identified as an antiviral mechanism in plants, arthropods, and nematodes. Although mammalian DNA viruses have been observed to encode functional miRNAs, whether RNA virus infection generates functional vsRNAs remains under discussion. This article reviews the most recent reports regarding pathways for generating vsRNAs and the identified vsRNA activity in mammalian cells infected with RNA viruses. We also discuss several hypotheses regarding the roles of mammalian vsRNAs and comment on the potential directions for this research field.


Subject(s)
DNA Viruses/genetics , Mammals/virology , MicroRNAs/genetics , RNA, Small Interfering/genetics , Animals , Mammals/immunology , RNA Interference , RNA Viruses
10.
Antiviral Res ; 112: 18-25, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25448086

ABSTRACT

Enterovirus 71 (EV71) infections can cause hand, foot, and mouth disease with severe neurological complications. Because no clinical drug is available for treating EV71 infections, developing an efficient antiviral medication against EV71 infection is crucial. This study indicated that 6-bromo-2-[1-(2,5-dimethylphenyl)-5-methyl-1H-pyrazol-4-yl] quinoline-4-carboxylic acid (BPR-3P0128) exhibits excellent antiviral activity against EV71 (EC50 = 0.0029 µM). BPR-3P0128 inhibits viral replication during the early post infection stage, targets EV71 RNA-dependent RNA polymerase and VPg uridylylation, and also reduces viral RNA accumulation levels and inhibits viral replication of EV71.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Enterovirus A, Human/enzymology , Protein Processing, Post-Translational/drug effects , Pyrazoles/pharmacology , Quinolines/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Virus Replication/drug effects , Cell Line , Cytopathogenic Effect, Viral , Enterovirus A, Human/physiology , Humans , Inhibitory Concentration 50 , Viral Plaque Assay , Viral Proteins/metabolism
11.
Nucleic Acids Res ; 42(20): 12789-805, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25352551

ABSTRACT

The roles of virus-derived small RNAs (vsRNAs) have been studied in plants and insects. However, the generation and function of small RNAs from cytoplasmic RNA viruses in mammalian cells remain unexplored. This study describes four vsRNAs that were detected in enterovirus 71-infected cells using next-generation sequencing and northern blots. Viral infection produced substantial levels (>10(5) copy numbers per cell) of vsRNA1, one of the four vsRNAs. We also demonstrated that Dicer is involved in vsRNA1 generation in infected cells. vsRNA1 overexpression inhibited viral translation and internal ribosomal entry site (IRES) activity in infected cells. Conversely, blocking vsRNA1 enhanced viral yield and viral protein synthesis. We also present evidence that vsRNA1 targets stem-loop II of the viral 5' untranslated region and inhibits the activity of the IRES through this sequence-specific targeting. Our study demonstrates the ability of a cytoplasmic RNA virus to generate functional vsRNA in mammalian cells. In addition, we also demonstrate a potential novel mechanism for a positive-stranded RNA virus to regulate viral translation: generating a vsRNA that targets the IRES.


Subject(s)
5' Untranslated Regions , Enterovirus A, Human/genetics , Gene Expression Regulation, Viral , Protein Biosynthesis , RNA, Small Untranslated/metabolism , RNA, Viral/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Ribonuclease III/metabolism , Viral Proteins/biosynthesis
12.
Virology ; 468-470: 592-600, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25299565

ABSTRACT

Neural progenitor cells (NPCs) are stem cells that can differentiate into various neural lineage cells. The damage and loss of NPCs are associated with neurological conditions such as cognitive deficits and memory impairment. In a long-term study of patients with EV71, cognitive disorders were observed. Therefore, we hypothesized that NPCs may be permissive to EV71 infection. We demonstrated that NPCs are prone to EV71 infection and that these stem cells can support the active replication of this virus. Furthermore, EV71 infection triggers apoptosis, resulting in significant cell death in infected NPCs. However, EV71 did not replicate in the differentiated cell types that were tested. Our findings suggest that EV71 can infect NPCs and cause the depletion of these cells.


Subject(s)
Enterovirus A, Human/physiology , Enterovirus Infections/virology , Neural Stem Cells/virology , Animals , Astrocytes/cytology , Astrocytes/physiology , Cell Differentiation , Enterovirus Infections/pathology , Mice , Mice, Inbred ICR , Neurons/cytology , Neurons/physiology , Viral Plaque Assay
13.
J Virol ; 87(7): 3828-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345520

ABSTRACT

Far-upstream element-binding protein 2 (FBP2) is an internal ribosomal entry site (IRES) trans-acting factor (ITAF) that negatively regulates enterovirus 71 (EV71) translation. This study shows that EV71 infection cleaved FBP2. Live EV71 and the EV71 replicon (but not UV-inactivated virus particles) induced FBP2 cleavage, suggesting that viral replication results in FBP2 cleavage. The results also showed that virus-induced proteasome, autophagy, and caspase activity co-contribute to EV71-induced FBP2 cleavage. Using FLAG-fused FBP2, we mapped the potential cleavage fragments of FBP2 in infected cells. We also found that FBP2 altered its function when its carboxyl terminus was cleaved. This study presents a mechanism for virus-induced cellular events to cleave a negative regulator for viral IRES-driven translation.


Subject(s)
Enterovirus A, Human/metabolism , Enterovirus Infections/metabolism , Protein Biosynthesis/physiology , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Virus Internalization , Blotting, Western , Cell Line, Tumor , Humans , Plasmids/genetics , Proteasome Endopeptidase Complex/metabolism , RNA, Small Interfering/genetics , Virus Replication/physiology
14.
Future Microbiol ; 7(4): 467-79, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22439724

ABSTRACT

The single-stranded RNA virus enterovirus 71 (EV71), which belongs to the Picornaviridae family, has caused epidemics worldwide, particularly in the Asia-Pacific region. Most EV71 infections result in mild clinical symptoms, including herpangina and hand, foot and mouth disease. However, serious pathological complications have also been reported, especially for young children. The mechanisms of EV71 disease progression remain unclear. The pathogenesis of adverse clinical outcomes may relate to many factors, including cell tropism, cell death and host immune responses. This article reviews the recent advances in the identification of factors determining EV71 cell tropism, the associated mechanisms of viral infection-induced cell death and the interplay between EV71 and immunity.


Subject(s)
Enterovirus A, Human/pathogenicity , Enterovirus Infections/immunology , Enterovirus Infections/virology , Host-Pathogen Interactions , Animals , Enterovirus A, Human/immunology , Enterovirus A, Human/physiology , Humans , Immunity , Viral Tropism
15.
J Biol Chem ; 286(36): 31373-84, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21784861

ABSTRACT

Enterovirus 71 (EV71), a member of the Picornaviridae family, may cause serious clinical manifestations associated with the central nervous system. Enterovirus 3C protease is required for virus replication and can trigger host cell apoptosis via cleaving viral polyprotein precursor and cellular proteins, respectively. Although the role of the 3C protease in processing viral and cellular proteins has been established, very little is known about the modulation of EV71 3C function by host cellular factors. Here, we show that sumoylation promotes EV71 3C protein ubiquitination for degradation, correlating with a decrease of EV71 in virus replication and cell apoptosis. SUMO E2-conjugating enzyme Ubc9 was identified as an EV71 3C-interacting protein. Further studies revealed that EV71 3C can be SUMO (small ubiquitin-like modifier)-modified at residue Lys-52. Sumoylation down-regulated 3C protease activity in vitro and also 3C protein stability in cells, in agreement with data suggesting 3C K52R protein induced greater substrate cleavage and apoptosis in cells. More importantly, the recombinant EV71 3C K52R virus infection conferred more apoptotic phenotype and increased virus levels in culture cells, which also correlated with a mouse model showing increased levels of viral VP1 protein in intestine and neuron loss in the spinal cord with EV71 3C K52R recombinant viral infection. Finally, we show that EV71 3C amino acid residues 45-52 involved in Ubc9 interaction determined the extent of 3C sumoylation and protein stability. Our results uncover a previously undescribed cellular regulatory event against EV71 virus replication and host cell apoptosis by sumoylation at 3C protease.


Subject(s)
Apoptosis , Cysteine Endopeptidases/metabolism , Enterovirus/physiology , Sumoylation , Viral Proteins/metabolism , Virus Replication , 3C Viral Proteases , Animals , Cells, Cultured , Cysteine Endopeptidases/genetics , Enterovirus/enzymology , Enterovirus Infections/pathology , Enterovirus Infections/virology , Host-Pathogen Interactions , Mice , Mutation, Missense , Protein Stability , Viral Proteins/genetics
16.
Microbes Infect ; 12(7): 505-10, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20348010

ABSTRACT

Enterovirus 71 (EV71) is a neurotropic pathogen that can cause severe neural diseases and complications on infected patients. Clinical observations showed that EV71-induced immune responses may be associated with virus induced neurogenic pulmonary edema. Here reviewed studies that discovered several host molecules as potential factors for EV71 virulence.


Subject(s)
Enterovirus A, Human/pathogenicity , Enterovirus Infections/virology , Nervous System Diseases/virology , Apoptosis , Enterovirus Infections/complications , Host-Pathogen Interactions , Humans , Nervous System/virology , Sepsis/complications , Sepsis/virology
17.
J Biomed Sci ; 16: 103, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19925687

ABSTRACT

Picornaviruses cause several diseases, not only in humans but also in various animal hosts. For instance, human enteroviruses can cause hand-foot-and-mouth disease, herpangina, myocarditis, acute flaccid paralysis, acute hemorrhagic conjunctivitis, severe neurological complications, including brainstem encephalitis, meningitis and poliomyelitis, and even death. The interaction between the virus and the host is important for viral replication, virulence and pathogenicity. This article reviews studies of the functions of viral and host factors that are involved in the life cycle of picornavirus. The interactions of viral capsid proteins with host cell receptors is discussed first, and the mechanisms by which the viral and host cell factors are involved in viral replication, viral translation and the switch from translation to RNA replication are then addressed. Understanding how cellular proteins interact with viral RNA or viral proteins, as well as the roles of each in viral infection, will provide insights for the design of novel antiviral agents based on these interactions.


Subject(s)
Picornaviridae Infections/virology , Picornaviridae/physiology , Animals , Capsid/chemistry , Genome, Viral , Humans , Models, Biological , Picornaviridae/genetics , Picornaviridae Infections/metabolism , Protein Biosynthesis , RNA Interference , RNA, Viral/chemistry , Viral Proteins/chemistry , Virus Replication
18.
PLoS Pathog ; 5(9): e1000593, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19779565

ABSTRACT

Identification of novel cellular proteins as substrates to viral proteases would provide a new insight into the mechanism of cell-virus interplay. Eight nuclear proteins as potential targets for enterovirus 71 (EV71) 3C protease (3C(pro)) cleavages were identified by 2D electrophoresis and MALDI-TOF analysis. Of these proteins, CstF-64, which is a critical factor for 3' pre-mRNA processing in a cell nucleus, was selected for further study. A time-course study to monitor the expression levels of CstF-64 in EV71-infected cells also revealed that the reduction of CstF-64 during virus infection was correlated with the production of viral 3C(pro). CstF-64 was cleaved in vitro by 3C(pro) but neither by mutant 3C(pro) (in which the catalytic site was inactivated) nor by another EV71 protease 2A(pro). Serial mutagenesis was performed in CstF-64, revealing that the 3C(pro) cleavage sites are located at position 251 in the N-terminal P/G-rich domain and at multiple positions close to the C-terminus of CstF-64 (around position 500). An accumulation of unprocessed pre-mRNA and the depression of mature mRNA were observed in EV71-infected cells. An in vitro assay revealed the inhibition of the 3'-end pre-mRNA processing and polyadenylation in 3C(pro)-treated nuclear extract, and this impairment was rescued by adding purified recombinant CstF-64 protein. In summing up the above results, we suggest that 3C(pro) cleavage inactivates CstF-64 and impairs the host cell polyadenylation in vitro, as well as in virus-infected cells. This finding is, to our knowledge, the first to demonstrate that a picornavirus protein affects the polyadenylation of host mRNA.


Subject(s)
Cysteine Endopeptidases/metabolism , Enterovirus A, Human/enzymology , Enterovirus Infections/virology , RNA-Binding Proteins/metabolism , Viral Proteins/metabolism , 3C Viral Proteases , Amino Acid Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , Cleavage Stimulation Factor , Cysteine Endopeptidases/genetics , Electrophoresis, Gel, Two-Dimensional , Enterovirus A, Human/pathogenicity , Enterovirus Infections/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Poly A/metabolism , Polyadenylation , Protein Binding , Protein Interaction Mapping , RNA Precursors/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Viral Proteins/genetics
19.
J Antimicrob Chemother ; 62(6): 1169-73, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18931391

ABSTRACT

Enteroviruses (EVs) are common human pathogens that are associated with numerous disease symptoms in many organ systems of the body. Although EV infections commonly cause mild or non-symptomatic illness, some of them are associated with severe diseases such as CNS complications. The current absence of effective vaccines for most viral infection and no available antiviral drugs for the treatment of EVs highlight the urgency and significance of developing antiviral agents. Several key steps in the viral life cycle are potential targets for blocking viral replication. This article reviews recent studies of antiviral developments for EVs based on various molecular targets that interrupt viral attachment, viral translation, polyprotein processing and RNA replication.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus/drug effects , Humans , Virus Attachment/drug effects , Virus Replication/drug effects
20.
J Neurovirol ; 14(1): 53-61, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18300075

ABSTRACT

Human glioblastoma cells (SF268) develop apoptosis, as characterized by DNA fragmentation and caspase activation, upon infection with Enterovirus 71 (EV71). To determine the step in virus replication that triggers apoptosis, the authors used ultraviolet (UV)-inactivated virus, inhibitors of protein and viral RNA synthesis, and chloroquine to block virus uncoating. Activation of caspase-3 was detected 24 h after infection with EV71 but not with UV-inactivated EV71. Apoptosis was inhibited when EV71-infected cells were treated with chloroquine, guanidine HCl, or cycloheximide. In summary, the authors studied the event(s) required to induce apoptosis in EV71-infected human glioblastoma cells, a subject much less studied than the possible role of viral proapoptotic genes, concluding that EV71 induces apoptosis in the infected SF268 cell in the presence of viral protein synthesis and virus replication, whereas virus adsorption, internalization, entry, uncoating, and viral RNA replication are all not required to trigger the apoptosis.


Subject(s)
Apoptosis , Enterovirus A, Human/physiology , Viral Proteins/biosynthesis , Animals , Cell Line, Tumor/cytology , Cell Line, Tumor/virology , Chlorocebus aethiops , Chloroquine/pharmacology , Cycloheximide/pharmacology , DNA Fragmentation , Enterovirus A, Human/drug effects , Enterovirus A, Human/genetics , Enterovirus A, Human/radiation effects , Glioblastoma/pathology , Guanidine/pharmacology , Humans , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Ultraviolet Rays , Vero Cells/virology , Viral Proteins/genetics , Viral Proteins/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL