Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Cell Biochem ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38720641

ABSTRACT

Enterovirus A71 (EV-A71) belongs to the genus Enterovirus of the Picornaviridae family and often causes outbreaks in Asia. EV-A71 infection usually causes hand, foot, and mouth disease and can even affect the central nervous system, causing neurological complications or death. The 5'-untranslated region (5'-UTR) of EV-A71 contains an internal ribosome entry site (IRES) that is responsible for the translation of viral proteins. IRES-transacting factors can interact with the EV-A71 5'-UTR to regulate IRES activity. Heterogeneous nuclear ribonucleoprotein (hnRNP) A3 is a member of the hnRNP A/B protein family of RNA-binding proteins and is involved in RNA transport and modification. We found that hnRNP A3 knockdown promoted the replication of EV-A71 in neural calls. Conversely, increasing the expression of hnRNP A3 within cells inhibits the growth of EV-A71. HnRNP A3 can bind to the EV-A71 5'-UTR, and knockdown of hnRNP A3 enhances the luciferase activity of the EV-A71 5'-UTR IRES. The localization of hnRNP A3 shifts from the nucleus to the cytoplasm of infected cells during viral infection. Additionally, EV-A71 infection can increase the protein expression of hnRNP A3, and the protein level is correlated with efficient viral growth. Based on these findings, we concluded that hnRNP A3 plays a negative regulatory role in EV-A71 replication within neural cells.

2.
Virus Res ; 339: 199284, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38040125

ABSTRACT

Enterovirus D68 (EV-D68) primarily spreads through the respiratory tract and causes respiratory symptoms in children and acute flaccid myelitis (AFM). Type III interferons (IFNs) play a critical role in inhibiting viral growth in respiratory epithelial cells. However, the mechanism by which EV-D68 induces type III IFN production is not yet fully understood. In this study, we show that EV-D68 infection stimulates Calu-3 cells to secrete IFN-λ. The transfection of EV-D68 viral RNA (vRNA) stimulated IFN-λ via MDA5. Furthermore, our findings provide evidence that EV-D68 infection also induces MDA5-IRF3/IRF7-mediated IFN-λ. In addition, we discovered that EV-D68 infection downregulated MDA5 expression. Knockdown of MDA5 increased EV-D68 replication in Calu-3 cells. Finally, we demonstrated that the IFN-λ1 and IFN-λ2/3 proteins effectively inhibit EV-D68 infection in respiratory epithelial cells. In summary, our study shows that EV-D68 induces type III IFN production via the activated MDA5-IRF3/IRF7 pathway and that type III IFNs inhibit EV-D68 replication in Calu-3 cells.


Subject(s)
Enterovirus D, Human , Enterovirus Infections , Neuromuscular Diseases , Child , Humans , Enterovirus D, Human/genetics , Interferon Lambda , Respiratory System
3.
Microbes Infect ; 25(7): 105171, 2023.
Article in English | MEDLINE | ID: mdl-37321390

ABSTRACT

Mesenchymal stem cells (MSCs) comprise a primitive cell population and reside in various tissues and organs. These cells exhibit immunomodulatory activity and are effective in treating respiratory viral infections. The activation of type I and III interferons, which protect cells against viral infections, can be induced after pattern recognition receptors (PRRs) recognize viral nucleic acid species. Although certain viruses can upregulate IFN-ß expression in MSCs, the underlying mechanisms and responsiveness to different IFNs are unclear. We found that foreskin-derived fibroblast-like stromal cells (FDSCs), a kind of functional MSC, were permissive to IAV PR8, HCoV-229E, and EV-D68. Infection by IAV PR8 and HCoV-229E increased the expression of IFN-ß and IFN-λ species in FDSCs in an IRF-3-dependent manner. RIG-I was critical for detecting IAV PR8 in FDSCs, and IAV PR8 infection induced a significant increase in the expression of interferon signaling genes (ISGs). Interestingly, only IFN-ß, but not IFN-λ species, could induce the expression of ISGs, a finding supported by our observation that only IFN-ß induced STAT1 and STAT2 phosphorylation in FDSCs. We also proved that treatment with IFN-ß suppressed the propagation of IAV PR8 and promoted the survival of virus-infected FDSCs. Respiratory viruses could infect FDSCs and induce the expression of IFN-ß and IFN-λ1, but only IFN-ß could protect FDSCs against viral infection.


Subject(s)
Mesenchymal Stem Cells , Virus Diseases , Viruses , Humans , Interferons/genetics , Interferons/metabolism , Interferon Lambda , Signal Transduction , Viruses/metabolism , Mesenchymal Stem Cells/metabolism , Interferon Regulatory Factor-3/metabolism
4.
Sci Rep ; 12(1): 21425, 2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36503883

ABSTRACT

Enterovirus A71 (EV-A71) is an emerging enterovirus that can cause neurological complications. Enhanced serum IL-1ß levels were observed in EV-A71 patients with severe neurological symptoms. However, the roles of sensors in enterovirus-induced IL-1ß production are unclear. In this study, we identified that pattern recognition receptors, including RIG-I, TLR3, and TLR8, are implicated in EV-A71-triggered IL-1ß release in human macrophages. EV-A71 infection results in caspase-1 and caspase-8, which act as regulators of EV-A71-induced NLRP3 and RIG-I inflammasome activation. Moreover, knockdown of the expression of TLR3 and TLR8 decreased the released IL-1ß in an NLRP3-dependent manner. Since TLR3 and TLR8 ligands promote NLRP3 inflammasome activation via caspase-8, the alternative pathway may be involved. In summary, these results indicate that activation of the NLRP3 and RIG-I inflammasomes in EV-A71-infected macrophages is mediated by caspase-1 and caspase-8 and affected by TLRs, including TLR3 and TLR8.


Subject(s)
Enterovirus Infections , Enterovirus , Humans , Antigens, Viral , Caspase 1 , Caspase 8 , Inflammasomes , Macrophages , Toll-Like Receptor 3
5.
RNA Biol ; 18(5): 796-808, 2021 05.
Article in English | MEDLINE | ID: mdl-33406999

ABSTRACT

The pathogenic human enterovirus EV-A71 has raised serious public health concerns. A hallmark of EV-A71 infection is the distortion of host transcriptomes in favour of viral replication. While high-throughput approaches have been exploited to dissect these gene dysregulations, they do not fully capture molecular perturbations at the single-cell level and in a physiologically relevant context. In this study, we applied a single-cell RNA sequencing approach on infected differentiated enterocyte cells (C2BBe1), which model the gastrointestinal epithelium targeted initially by EV-A71. Our single-cell analysis of EV-A71-infected culture provided several lines of illuminating observations: 1) This systems approach demonstrated extensive cell-to-cell variation in a single culture upon viral infection and delineated transcriptomic differences between the EV-A71-infected and bystander cells. 2) By analysing expression profiles of known EV-A71 receptors and entry facilitation factors, we found that ANXA2 was closely correlated in expression with the viral RNA in the infected population, supporting its role in EV-A71 entry in the enteric cells. 3) We further catalogued dysregulated lncRNAs elicited by EV-A71 infection and demonstrated the functional implication of lncRNA CYTOR in promoting EV-A71 replication. Viewed together, our single-cell transcriptomic analysis illustrated at the single-cell resolution the heterogeneity of host susceptibility to EV-A71 and revealed the involvement of lncRNAs in host antiviral response.


Subject(s)
Enterovirus A, Human/pathogenicity , Host-Pathogen Interactions/genetics , Transcriptome , Cells, Cultured , Enterocytes/metabolism , Enterocytes/pathology , Enterocytes/virology , Enterovirus A, Human/genetics , Enterovirus A, Human/immunology , Enterovirus Infections/genetics , Enterovirus Infections/immunology , Enterovirus Infections/pathology , Enterovirus Infections/virology , Gene Expression Profiling , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , RNA, Long Noncoding/genetics , Single-Cell Analysis , Virus Replication/genetics
6.
Sci Rep ; 10(1): 15234, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943650

ABSTRACT

Enterovirus A71 (EV-A71), which belongs to the family Picornaviridae, can invade the central nervous system (CNS) and cause severe CNS complications or death. The EV-A71 antigen has been detected in the neurons in the brains of humans who died from EV-A71 infection. However, the effect of EV-A71 infection on human neuronal cells remains poorly understood. Human neural stem cells (NSCs) and IMR-32 neuroblastoma cells were differentiated into neuronal cells for this study. Although the neuronal cells were permissive to EV-A71 infection, EV-A71 infection did not induce an obvious cytopathic effect on the neuronal cells. EV-A71 infection did not induce apoptosis in neuronal cells. However, autophagy and autophagic flux were induced in EV-A71-infected neuronal cells. The production of autophagosomes was shown to be important for EV-A71 viral RNA (vRNA) replication in neuronal cells.


Subject(s)
Enterovirus A, Human/physiology , Enterovirus A, Human/pathogenicity , Neurons/virology , Autophagosomes/virology , Autophagy/physiology , Caspases/metabolism , Cell Differentiation , Cells, Cultured , Cytopathogenic Effect, Viral/physiology , Enterovirus A, Human/genetics , Enzyme Activation , Host Microbial Interactions/physiology , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/virology , Neurons/metabolism , Neurons/pathology , RNA, Viral/biosynthesis , RNA, Viral/genetics , Virus Replication/physiology
7.
Emerg Microbes Infect ; 9(1): 1457-1466, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32543353

ABSTRACT

Taiwan experienced two waves of imported infections with Coronavirus Disease 2019 (COVID-19). This study aimed at investigating the genomic variation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Taiwan and compared their evolutionary trajectories with the global strains. We performed culture and full-genome sequencing of SARS-CoV-2 strains followed by phylogenetic analysis. A 382-nucleotides deletion in open reading frame 8 (ORF8) was found in a Taiwanese strain isolated from a patient on February 4, 2020 who had a travel history to Wuhan. Patients in the first wave also included several sporadic, local transmission cases. Genomes of 5 strains sequenced from clustered infections were classified into a new clade with ORF1ab-V378I mutation, in addition to 3 dominant clades ORF8-L84S, ORF3a-G251V and S-D614G. This highlighted clade also included some strains isolated from patients who had a travel history to Turkey and Iran. The second wave mostly resulted from patients who had a travel history to Europe and Americas. All Taiwanese viruses were classified into various clades. Genomic surveillance of SARS-CoV-2 in Taiwan revealed a new ORF8-deletion mutant and a virus clade that may be associated with infections in the Middle East, which contributed to a better understanding of the global SARS-CoV-2 transmission dynamics.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/virology , Genome, Viral , Pneumonia, Viral/virology , Animals , Betacoronavirus/classification , Betacoronavirus/isolation & purification , COVID-19 , Cell Line , Chlorocebus aethiops , Haemophilus parainfluenzae/isolation & purification , Humans , Middle East , Open Reading Frames , Pandemics , Phylogeny , RNA, Viral , SARS-CoV-2 , Sequence Deletion , Taiwan , Travel , Vero Cells , Virus Cultivation , Whole Genome Sequencing
8.
J Infect Dis ; 222(3): 456-469, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32271384

ABSTRACT

BACKGROUND: Enterovirus A71 (EV-A71) has been noted for its tendency to lead to neurological manifestations in young children and infants. Although the alimentary tract has been identified as the primary replication site of this virus, how EV-A71 replicates in the gut and is transmitted to other organs remains unclear. METHODS: By using differentiated C2BBe1 cells as a model, we observed that intestinal epithelial cells (IECs) were permissive to EV-A71 infection, and viral particles were released in a nonlytic manner. RESULTS: The coexistence of active caspase 3 and EV-A71 protein was observed in the infected undifferentiated C2BBe1 and RD cells but not in the infected differentiated C2BBe1 cells. Furthermore, EV-A71 infection caused differentiated C2BBe1 and intestinal organoids to secrete exosomes containing viral components and have the ability to establish active infection. Inhibition of the exosome pathway decreased EV-A71 replication and release in IECs and increased the survival rates of infected animals. CONCLUSIONS: Our findings showed that EV-A71 is able to be actively replicated in enterocytes, and that the exosome pathway is involved in the nonlytic release of viral particles, which may be useful for developing antiviral strategies.


Subject(s)
Enterovirus A, Human/physiology , Epithelial Cells/metabolism , Epithelial Cells/virology , Exosomes/metabolism , Animals , Cell Differentiation , Enterovirus , Enterovirus A, Human/genetics , Enterovirus A, Human/growth & development , Enterovirus Infections/virology , Humans , Mice, Transgenic , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication
9.
Viruses ; 11(12)2019 12 04.
Article in English | MEDLINE | ID: mdl-31817126

ABSTRACT

Enterovirus 71 (EV71) can invade the central nervous system (CNS) and cause neurological disease. Accumulating evidence indicates that EV71 can directly infect neurons in the CNS. Innate immune responses in the CNS have been known to play an essential role in limiting pathogen infections. Thus, investigating the effects of EV71 infection of neural cells is important for understanding disease pathogenesis. In this study, human neural cells were infected with EV71, and interferonß (IFNß) expression was examined. Our results show that IFNß expression was upregulated in EV71-infected neural cells via pattern recognition receptors (PRRs) sensing of virus RNA. The PRRs Toll-like receptor 3 (TLR3), Toll-like receptor 8 (TLR8), and melanoma differentiation-associated gene-5 (MDA-5), but not retinoic acid-inducible gene-I (RIG-I) and Toll-like receptor 7 (TLR7), were found to be EV71-mediated IFNß induction. Although viral proteins exhibited the ability to cleave mitochondrial antiviral signaling protein (MAVS) and Toll/IL-1 receptor (TIR) domain-containing adaptor-inducing IFN-ß (TRIF) in neural cells, levels of viral protein expression were low in these cells. Furthermore, neural cells efficiently produced IFNß transcripts upon EV71 vRNA stimulation. Treating infected cells with anti-IFNß antibodies resulted in increased virus replication, indicating that IFNß release may play a role in limiting viral growth. These results indicate that EV71 infection can induce IFNß expression in neural cells through PRR pathways.


Subject(s)
Enterovirus A, Human/physiology , Enterovirus Infections/genetics , Enterovirus Infections/virology , Gene Expression , Interferon-beta/genetics , Nervous System Diseases/genetics , Nervous System Diseases/virology , Cell Line , Enterovirus Infections/metabolism , Humans , Interferon-beta/metabolism , Nervous System Diseases/metabolism , Neurons/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Toll-Like Receptors/metabolism , Virus Replication
10.
Stem Cell Res Ther ; 10(1): 387, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31843025

ABSTRACT

BACKGROUND: Neural stem cells (NSCs) residing in the central nervous system play an important role in neurogenesis. Several viruses can infect these neural progenitors and cause severe neurological diseases. The innate immune responses against the neurotropic viruses in these tissue-specific stem cells remain unclear. METHODS: Human NSCs were transfected with viral RNA mimics or infected with neurotropic virus for detecting the expression of antiviral interferons (IFNs) and downstream IFN-stimulated antiviral genes. RESULTS: NSCs are able to produce interferon-ß (IFN-ß) (type I) and λ1 (type III) after transfection with poly(I:C) and that downstream IFN-stimulated antiviral genes, such as ISG56 and MxA, and the viral RNA sensors RIG-I, MDA5, and TLR3, can be expressed in NSCs under poly(I:C) or IFN-ß stimulation. In addition, our results show that the pattern recognition receptors RIG-I and MDA5, as well as the endosomal pathogen recognition receptor TLR3, but not TLR7 and TLR8, are involved in the activation of IFN-ß transcription in NSCs. Furthermore, NSCs infected with the neurotropic viruses, Zika and Japanese encephalitis viruses, are able to induce RIG-I-mediated IFN-ß expression. CONCLUSION: Human NSCs have the ability to activate IFN signals against neurotropic viral pathogens.


Subject(s)
Interferon Type I/immunology , Neural Stem Cells/immunology , Neural Stem Cells/virology , Zika Virus Infection/immunology , Cell Line , Cells, Cultured , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Encephalitis Viruses, Japanese/immunology , Encephalitis, Japanese/genetics , Encephalitis, Japanese/immunology , Humans , Immunity, Innate , Interferon Type I/biosynthesis , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/immunology , Interferons/genetics , Interferons/immunology , Neural Stem Cells/pathology , Receptors, Immunologic , Transcription, Genetic , Transfection , Zika Virus/immunology , Zika Virus Infection/genetics , Zika Virus Infection/pathology , Interferon Lambda
11.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30814289

ABSTRACT

Infection by enteroviruses can cause severe neurological complications in humans. The interactions between the enteroviral and host proteins may facilitate the virus replication and be involved in the pathogenicity of infected individuals. It has been shown that human enteroviruses possess various mechanisms to suppress host innate immune responses in infected cells. Previous studies showed that infection by enterovirus 71 (EV71) causes the degradation of MDA5, which is a critical cytoplasmic pathogen sensor in the recognition of picornaviruses for initiating transcription of type I interferons. In the present study, we demonstrated that the RNA-dependent RNA polymerase (RdRP; also denoted 3Dpol) encoded by EV71 interacts with the caspase activation and recruitment domains (CARDs) of MDA5 and plays a role in the inhibition of MDA5-mediated beta interferon (IFN-ß) promoter activation and mRNA expression. In addition, we found that the 3Dpol protein encoded by coxsackievirus B3 also interacted with MDA5 and downregulated the antiviral signaling initiated by MDA5. These findings indicate that enteroviral RdRP may function as an antagonist against the host antiviral innate immune response.IMPORTANCE Infection by enteroviruses causes severe neurological complications in humans. Human enteroviruses possess various mechanisms to suppress the host type I interferon (IFN) response in infected cells to establish viral replication. In the present study, we found that the enteroviral 3Dpol protein (or RdRP), which is a viral RNA-dependent RNA polymerase for replicating viral RNA, plays a role in the inhibition of MDA5-mediated beta interferon (IFN-ß) promoter activation. We further demonstrated that enteroviral 3Dpol protein interacts with the caspase activation and recruitment domains (CARDs) of MDA5. These findings indicate that enteroviral RdRP functions as an antagonist against the host antiviral response.


Subject(s)
Enterovirus A, Human/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , RNA-Dependent RNA Polymerase/metabolism , Caspase Activation and Recruitment Domain/genetics , Caspase Activation and Recruitment Domain/physiology , Enterovirus/genetics , Enterovirus/metabolism , Enterovirus A, Human/genetics , Enterovirus B, Human/metabolism , Enterovirus Infections/virology , HEK293 Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon-beta/metabolism , Interferons/metabolism , Interferons/physiology , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Signal Transduction , Virus Replication
12.
PLoS One ; 13(1): e0191617, 2018.
Article in English | MEDLINE | ID: mdl-29370243

ABSTRACT

EV71 is a positive-sense single-stranded RNA virus that belongs to the Picornaviridae family. EV71 infection may cause various symptoms ranging from hand-foot-and-mouth disease to neurological pathological conditions such as aseptic meningitis, ataxia, and acute transverse myelitis. There is currently no effective treatment or vaccine available. Various compounds have been examined for their ability to restrict EV71 replication. However, most experiments have been performed in rhabdomyosarcoma or Vero cells. Since the gastrointestinal tract is the entry site for this pathogen, we anticipated that orally ingested agents may exert beneficial effects by decreasing virus replication in intestinal epithelial cells. In this study, curcumin (diferuloylmethane, C21H20O6), an active ingredient of turmeric (Curcuma longa Linn) with anti-cancer properties, was investigated for its anti-enterovirus activity. We demonstrate that curcumin treatment inhibits viral translation and increases host cell viability. Curcumin does not exert its anti-EV71 effects by modulating virus attachment or virus internal ribosome entry site (IRES) activity. Furthermore, curcumin-mediated regulation of mitogen-activated protein kinase (MAPK) signaling pathways is not involved. We found that protein kinase C delta (PKCδ) plays a role in virus translation in EV71-infected intestinal epithelial cells and that curcumin treatment decreases the phosphorylation of this enzyme. In addition, we show evidence that curcumin also limits viral translation in differentiated human intestinal epithelial cells. In summary, our data demonstrate the anti-EV71 properties of curcumin, suggesting that ingestion of this phytochemical may protect against enteroviral infections.


Subject(s)
Curcumin/metabolism , Curcumin/therapeutic use , Enterovirus Infections/drug therapy , Cell Line , Curcumin/pharmacology , Enterovirus/drug effects , Enterovirus A, Human/genetics , Enterovirus Infections/virology , Epithelial Cells/drug effects , HT29 Cells , Hand, Foot and Mouth Disease/metabolism , Host-Pathogen Interactions , Humans , Internal Ribosome Entry Sites , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/drug effects , Protein Biosynthesis/drug effects , RNA, Viral/genetics , Viral Proteins/genetics , Virus Replication/drug effects
13.
Proteomics ; 16(17): 2351-62, 2016 09.
Article in English | MEDLINE | ID: mdl-27291656

ABSTRACT

Enterovirus 71 (EV71), a single-stranded RNA virus, is one of the most serious neurotropic pathogens in the Asia-Pacific region. Through interactions with host proteins, the 5' untranslated region (5'UTR) of EV71 is important for viral replication. To gain a protein profile that interact with the EV71 5'UTR in neuronal cells, we performed a biotinylated RNA-protein pull-down assay in conjunction with LC-MS/MS analysis. A total of 109 proteins were detected and subjected to Database for Annotation, Visualization and Integrated Discovery (DAVID) analyses. These proteins were found to be highly correlated with biological processes including RNA processing/splicing, epidermal cell differentiation, and protein folding. A protein-protein interaction network was constructed using the STRING online database to illustrate the interactions of those proteins that are mainly involved in RNA processing/splicing or protein folding. Moreover, we confirmed that the far-upstream element binding protein 3 (FBP3) was able to bind to the EV71 5'UTR. The redistribution of FBP3 in subcellular compartments was observed after EV71 infection, and the decreased expression of FBP3 in host neuronal cells markedly inhibited viral replication. Our results reveal various host proteins that potentially interact with the EV71 5'UTR in neuronal cells, and we found that FBP3 could serve as a positive regulator in host cells.


Subject(s)
DNA-Binding Proteins/metabolism , Enterovirus A, Human/physiology , Enterovirus Infections/metabolism , Neurons/virology , RNA, Viral/metabolism , Transcription Factors/metabolism , Virus Replication , 5' Untranslated Regions , Cell Line , Enterovirus A, Human/genetics , Enterovirus Infections/pathology , Humans , Neurons/metabolism , Neurons/pathology , RNA, Viral/genetics
14.
Viruses ; 7(11): 6051-66, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26610549

ABSTRACT

Enteroviruses are a group of positive-sense single stranded viruses that belong to the Picornaviridae family. Most enteroviruses infect humans from the gastrointestinal tract and cause mild symptoms. However, several enteroviruses can invade the central nervous system (CNS) and result in various neurological symptoms that are correlated to mortality associated with enteroviral infections. In recent years, large outbreaks of enteroviruses occurred worldwide. Therefore, these neurotropic enteroviruses have been deemed as re-emerging pathogens. Although these viruses are becoming large threats to public health, our understanding of these viruses, especially for non-polio enteroviruses, is limited. In this article, we review recent advances in the trafficking of these pathogens from the peripheral to the central nervous system, compare their cell tropism, and discuss the effects of viral infections in their host neuronal cells.


Subject(s)
Central Nervous System Infections/epidemiology , Central Nervous System Infections/virology , Disease Outbreaks , Enterovirus Infections/epidemiology , Enterovirus Infections/virology , Enterovirus/isolation & purification , Enterovirus/pathogenicity , Enterovirus/physiology , Global Health , Host-Pathogen Interactions , Humans , Viral Tropism
15.
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
16.
J Proteome Res ; 14(4): 1818-30, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25785312

ABSTRACT

Enterovirus 71 (EV71) is a human enterovirus that has seriously affected the Asia-Pacific area for the past two decades. EV71 infection can result in mild hand-foot-and-mouth disease and herpangina and may occasionally lead to severe neurological complications in children. However, the specific biological processes that become altered during EV71 infection remain unclear. To further explore host responses upon EV71 infection, we identified proteins differentially expressed in EV71-infected human glioblastoma SF268 cells using isobaric mass tag (iTRAQ) labeling coupled with multidimensional liquid chromatography-mass spectrometry (LC-MS/MS). Network analysis of proteins altered in cells infected with EV71 revealed that the changed biological processes are related to protein and ion transport, regulation of protein degradation, and homeostatic processes. We confirmed that the levels of NEDD4L and PSMF1 were increased and reduced, respectively, in EV71-infected cells compared to mock-infected control cells. To determine the physiological relevance of our findings, we investigated the consequences of EV71 infection in cells with NEDD4L or PSMF1 depletion. We found that the depletion of NEDD4L significantly reduced the replication of EV71, whereas PSMF1 knockdown enhanced EV71 replication. Collectively, our findings provide the first evidence of proteome-wide dysregulation by EV71 infection and suggest a novel role for the host protein NEDD4L in the replication of this virus.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Enterovirus A, Human/physiology , Enterovirus Infections/physiopathology , Gene Expression Regulation/physiology , Ubiquitin-Protein Ligases/metabolism , Virus Replication/physiology , Cell Line, Tumor , Chromatography, Liquid , Computational Biology , DNA Primers , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Immunoblotting , Nedd4 Ubiquitin Protein Ligases , Proteasome Endopeptidase Complex , Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
17.
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
18.
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
19.
Cell Biol Int ; 37(12): 1308-19, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23956153

ABSTRACT

Foreskin fibroblast-like stromal cells (FDSCs) are progenitors isolated from human tissue that can differentiate into diverse cell types. Many types of stem cells can differentiate into hepatocyte-like cells, which could be used for drug testing or in liver regeneration therapy, but whether FDSCs can be converted into functional hepatocytes is unknown. FDSCs show divergent properties when cultured in distinct media, forming spheres in Dulbecco's modified Eagle's medium (DMEM) containing F12, epidermal growth factor (EGF), and basic fibroblast growth factor (b-FGF), but have fibroblast-like morphology when cultured in DMEM-based growth medium. Both cell populations express the typical mesenchymal stem cell markers CD90, CD105, and CD73, but the p75 neurotrophin receptor (p75NTR) was detected only in FDSC spheres. Both types of FDSCs can differentiate into hepatocyte-like cells, which express typical liver markers, including albumin and hepatocyte paraffin 1 (Hep Par1), along with liver-specific biological activities. When plasmids containing the human hepatitis B virus (HBV) genome were transfected transiently into FDSCs, differentiated hepatocyte-like cells secrete large amounts of HBe and HBs antigens. FDSCs could be used for clinical hepatic therapy and/or serve as a model of HBV.


Subject(s)
Cell Differentiation , Foreskin/cytology , Hepatocytes/cytology , Stromal Cells/cytology , Biomarkers/metabolism , Cells, Cultured , Child , Fibroblasts/cytology , Genes, Viral/genetics , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/metabolism , Hepatitis B virus/genetics , Hepatocytes/metabolism , Humans , Immunohistochemistry , Male , Plasmids/genetics , Plasmids/metabolism , Stromal Cells/metabolism , Transfection
20.
In Vitro Cell Dev Biol Anim ; 49(5): 307-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23636940

ABSTRACT

Stem cells are unique cell populations with the ability to undergo self-renewal and differentiation. These cells have been identified in a wide range of tissues and possess varied differentiation potentials. Tissue-specific stem cells have typically been thought to have limited differentiation capabilities. We show here that fibroblast-like cells isolated from mouse brain possess cross-germ layer differentiation abilities. These cells were found to express typical mesenchymal stem cell markers (CD44, CD29, and CD105) and were able to be passaged more than 50 times. When treated under defined conditions, the brain-derived cells were able to generate many different cell types including adipocytes, osteocytes, astrocytes, neurons, and even hepatocyte-like cells. The hepatocyte-like cells not only expressed liver cell-specific markers, but also exhibited the capacity for glycogen storage and low-density lipoprotein uptake. These results demonstrate the existence of cells in the brain with three-germ-layer differentiation potential.


Subject(s)
Brain/cytology , Cell Differentiation/physiology , Cell Lineage/physiology , Stem Cells/cytology , Animals , Antigens, Differentiation/metabolism , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred BALB C , Rosaniline Dyes , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...