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










Publication year range
1.
Vaccine ; 37(15): 2158-2166, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30857932

ABSTRACT

In contrast to current ether- or detergent-disrupted "split" vaccines (SVs) for influenza, inactivated whole influenza virus particle vaccines (WPVs) retain the original virus structure and components and as such may confer similar immunity to natural infection. In a collaboration between academia and industry, the potential of WPV as a new seasonal influenza vaccine was investigated. Each of the four seasonal influenza vaccine manufacturers in Japan prepared WPVs and SVs from the same batches of purified influenza virus. Both mice and monkeys vaccinated with the WPVs exhibited superior immune responses to those vaccinated with the corresponding SVs. Vaccination with A/California/07/2009 (H1N1) WPV enabled mice to survive a lethal challenge dose of homologous virus whereas those vaccinated with SV succumbed to infection within 6 days. Furthermore, mice vaccinated with WPV induced substantial numbers of multifunctional CD8+ T cells, important for control of antigenically drifted influenza virus strains. In addition, cytokines and chemokines were detected at early time points in the sera of mice vaccinated with WPV but not in those animals vaccinated with SV. These results indicate that WPVs induce enhanced innate and adaptive immune responses compared to equivalent doses of SVs. Notably, WPV at one fifth of the dose of SV was able to induce potent immunity with limited production of IL-6, one of the pyrogenic cytokines. We thus propose that WPVs with balanced immunogenicity and safety may set a new global standard for seasonal influenza vaccines.


Subject(s)
Antibodies, Viral/blood , Influenza Vaccines/immunology , Interleukin-6/blood , Orthomyxoviridae Infections/prevention & control , Virion/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Chemokines/blood , Cytokines/blood , Female , Humans , Immunogenicity, Vaccine , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Interleukin-6/immunology , Japan , Macaca , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
2.
J Biol Chem ; 293(48): 18585-18600, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30282637

ABSTRACT

The innate immune system is important for the efficacy of vaccines, but excessive innate immune responses can cause adverse reactions after vaccination. Extracellular vesicles (EVs) are enriched in the blood and can deliver functional RNAs, such as microRNAs (miRNAs), to recipient cells, thereby mediating intercellular communication. However, the role of EVs in controlling the innate immune responses to vaccines has not been fully elucidated. Here, we found that miR-451a is abundant in human serum EVs and that its presence in blood-circulating EVs affects the innate immune responses of macrophages and dendritic cells to inactivated whole-virus vaccines (WV) against influenza. miR-451a in human serum EVs was stable for a week in healthy subjects, and its levels gradually fluctuated over several months. miR-451a within serum EVs was internalized into serum-cultured macrophages and dendritic cells and reduced endogenous 14-3-3ζ protein levels and decreased the expression of type I IFN and interleukin 6 in response to WV stimulation. miR-451a levels in blood-circulating EVs were positively correlated with intracellular miR-451a levels in mouse splenic CD11c+ cells and inversely correlated with the innate immune response to inactivated WV in vivo These findings suggest that miR-451a in circulating EVs is internalized into recipient cells in vivo and that this internalization results in an attenuation of the innate immune response to WV. Moreover, a microarray analysis identified several other miRNAs that affect the macrophage response to inactivated WV. Our results reveal that miRNAs in circulating EVs significantly modify the responses of macrophages and dendritic cells to inactivated WV.


Subject(s)
Dendritic Cells/immunology , Extracellular Vesicles/immunology , Influenza Vaccines/immunology , Macrophages/immunology , MicroRNAs/blood , 14-3-3 Proteins/metabolism , Adult , Animals , Cell Line , Dendritic Cells/metabolism , Exocytosis , Extracellular Vesicles/metabolism , Humans , Immunity, Innate , Macrophages/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction
3.
Microbiol Immunol ; 2018 May 17.
Article in English | MEDLINE | ID: mdl-29771464

ABSTRACT

Accumulation of amyloid ß (Aß40 and Aß42) in the brain is a characteristic of Alzheimer's disease (AD). Because neprilysin (NEP) is a major Aß-degrading enzyme, NEP delivery in the brain is a promising gene therapy for AD. Borna disease virus (BoDV) vector enables long-term transduction of foreign genes in the central nerve system. Here, we evaluated the proteolytic ability of NEP transduced by the BoDV vector and found that the amounts of Aß40 and Aß42 significantly decreased, which suggests that NEP expressed from the BoDV vector is functional to degrade Aß.

4.
J Biochem ; 163(1): 31-38, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29036651

ABSTRACT

Several microbial molecules with pathogen-associated molecular patterns stimulate host innate immune responses. The innate immune system plays a crucial role in activating acquired immune response via cytokine production and antigen presentation. Previous studies have shown that Aureobasidium pullulans-cultured fluid (AP-CF), which contains ß-glucan, exhibits adjuvant activity and renders mice resistance to influenza A virus infection; however, the underlying mechanism remains elusive. In this study, we investigated the innate immune response to AP-CF. We found that intraperitoneal administration of AP-CF increased the serum level of IL-18 and the number of splenic IFN-γ producing CD4+ cells during influenza A virus infection. The adjuvant effect of AP-CF was distinct from that of alum, which is known to have the ability to stimulate a Th2 immune response. In addition, AP-CF injection barely increased the number of peritoneal neutrophils and inflammatory macrophages, whereas alum injection markedly increased the number of neutrophils and inflammatory macrophages, suggesting that AP-CF is a weak inducer of inflammation compared to alum. AP-CF induced IL-18 production by DC2.4 cells, a dendritic cell line, and by peritoneal exudate cells that include peritoneal macrophages. Collectively, our findings indicate that AP-CF is an adjuvant that promotes the Th1 response during influenza A virus infection.


Subject(s)
Ascomycota/chemistry , Glucans/pharmacology , Influenza A virus/drug effects , Interleukin-18/biosynthesis , Orthomyxoviridae Infections/drug therapy , Th1 Cells/drug effects , Animals , Glucans/chemistry , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Th1 Cells/virology
5.
Microbiol Immunol ; 61(9): 380-386, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28776750

ABSTRACT

Borna disease virus (BoDV), a prototype of mammalian bornavirus, is a non-segmented, negative strand RNA virus that often causes severe neurological disorders in infected animals, including horses and sheep. Unique among animal RNA viruses, BoDV transcribes and replicates non-cytopathically in the cell nucleus, leading to establishment of long-lasting persistent infection. This striking feature of BoDV indicates its potential as an RNA virus vector system. It has previously been demonstrated by our team that recombinant BoDV (rBoDV) lacking an envelope glycoprotein (G) gene develops persistent infections in transduced cells without loss of the viral genome. In this study, a novel non-transmissive rBoDV, rBoDV ΔMG, which lacks both matrix (M) and G genes in the genome, is reported. rBoDV-ΔMG expressing green fluorescence protein (GFP), rBoDV ΔMG-GFP, was efficiently generated in Vero/MG cells stably expressing both BoDV M and G proteins. Infection with rBoDV ΔMG-GFP was persistently maintained in the parent Vero cells without propagation within cell culture. The optimal ratio of M and G for efficient viral particle production by transient transfection of M and G expression plasmids into cells persistently infected with rBoDV ΔMG-GFP was also demonstrated. These findings indicate that the rBoDV ΔMG-based BoDV vector may provide an extremely safe virus vector system and could be a novel strategy for investigating the function of M and G proteins and the host range of bornaviruses.


Subject(s)
Borna Disease/transmission , Borna disease virus/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Viral Envelope Proteins/genetics , Viral Matrix Proteins/genetics , Animals , Borna Disease/virology , Borna disease virus/pathogenicity , Cell Line , Chlorocebus aethiops , Genome, Viral/genetics , Glycoproteins/genetics , HEK293 Cells , Humans , RNA, Viral/genetics , Vero Cells , Virus Replication/genetics
6.
Front Immunol ; 7: 335, 2016.
Article in English | MEDLINE | ID: mdl-27630638

ABSTRACT

The innate immune system is essential for controlling viral infection. Hepatitis B virus (HBV) persistently infects human hepatocytes and causes hepatocellular carcinoma. However, the innate immune response to HBV infection in vivo remains unclear. Using a tree shrew animal model, we showed that HBV infection induced hepatic interferon (IFN)-γ expression during early infection. Our in vitro study demonstrated that hepatic NK cells produced IFN-γ in response to HBV only in the presence of hepatic F4/80(+) cells. Moreover, extracellular vesicles (EVs) released from HBV-infected hepatocytes contained viral nucleic acids and induced NKG2D ligand expression in macrophages by stimulating MyD88, TICAM-1, and MAVS-dependent pathways. In addition, depletion of exosomes from EVs markedly reduced NKG2D ligand expression, suggesting the importance of exosomes for NK cell activation. In contrast, infection of hepatocytes with HBV increased immunoregulatory microRNA levels in EVs and exosomes, which were transferred to macrophages, thereby suppressing IL-12p35 mRNA expression in macrophages to counteract the host innate immune response. IFN-γ increased the hepatic expression of DDX60 and augmented the DDX60-dependent degradation of cytoplasmic HBV RNA. Our results elucidated the crucial role of exosomes in antiviral innate immune response against HBV. ACCESSION NUMBER: Accession number of RNA-seq data is DRA004164 (DRA in DDBJ).

8.
Sci Rep ; 6: 31022, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27516062

ABSTRACT

Although epigenetic abnormalities have been described in Huntington's disease (HD), the causal epigenetic mechanisms driving neurodegeneration in HD cortex and striatum remain undefined. Using an epigenetic pathway-targeted drug screen, we report that inhibitors of DNA methyltransferases (DNMTs), decitabine and FdCyd, block mutant huntingtin (Htt)-induced toxicity in primary cortical and striatal neurons. In addition, knockdown of DNMT3A or DNMT1 protected neurons against mutant Htt-induced toxicity, together demonstrating a requirement for DNMTs in mutant Htt-triggered neuronal death and suggesting a neurodegenerative mechanism based on DNA methylation-mediated transcriptional repression. Inhibition of DNMTs in HD model primary cortical or striatal neurons restored the expression of several key genes, including Bdnf, an important neurotrophic factor implicated in HD. Accordingly, the Bdnf promoter exhibited aberrant cytosine methylation in mutant Htt-expressing cortical neurons. In vivo, pharmacological inhibition of DNMTs in HD mouse brains restored the mRNA levels of key striatal genes known to be downregulated in HD. Thus, disturbances in DNA methylation play a critical role in mutant Htt-induced neuronal dysfunction and death, raising the possibility that epigenetic strategies targeting abnormal DNA methylation may have therapeutic utility in HD.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Huntingtin Protein/toxicity , Huntington Disease/prevention & control , Mutant Proteins/toxicity , Animals , Brain/pathology , Cells, Cultured , DNA Methyltransferase 3A , Disease Models, Animal , Humans , Huntington Disease/pathology , Mice , Neurons/drug effects , Neurons/physiology
9.
Sci Rep ; 6: 26154, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27189575

ABSTRACT

RNA interference (RNAi) has emerged as a promising technique for gene therapy. However, the safe and long-term expression of small RNA molecules is a major concern for the application of RNAi therapies in vivo. Borna disease virus (BDV), a non-segmented, negative-strand RNA virus, establishes a persistent infection without obvious cytopathic effects. Unique among animal non-retroviral RNA viruses, BDV persistently establishes a long-lasting persistent infection in the nucleus. These features make BDV ideal for RNA virus vector persistently expressing small RNAs. Here, we demonstrated that the recombinant BDV (rBDV) containing the miR-155 precursor, rBDV-miR-155, persistently expressed miR-155 and efficiently silenced its target gene. The stem region of the miR-155 precursor in rBDV-miR-155 was replaceable by any miRNA sequences of interest and that such rBDVs efficiently silence the expression of target genes. Collectively, BDV vector would be a novel RNA virus vector enabling the long-term expression of miRNAs for RNAi therapies.


Subject(s)
Borna disease virus/genetics , Gene Expression , Genetic Vectors , MicroRNAs/biosynthesis , Molecular Biology/methods , RNA Interference , Animals , Chlorocebus aethiops , Genetic Therapy/methods , HEK293 Cells , Humans , MicroRNAs/genetics , Vero Cells
10.
Rev Med Virol ; 26(2): 90-101, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26643446

ABSTRACT

Recognition and degradation of viral RNA are essential for antiviral innate immune responses. Cytoplasmic viral RNA is recognized by retinoic acid-inducible gene I (RIG-I)-like receptors, which trigger type I interferon (IFN) production. Secreted type I IFN activates ubiquitously expressed type I IFN receptor and induces IFN-stimulated genes (ISGs). To suppress viral replication, several nucleases degrade viral RNA. RNase L is an ISG with endonuclease activity that degrades viral RNA, producing small RNA that activates RIG-I, resulting in the amplification of type I IFN production. Moreover, recent studies have elucidated novel links between viral RNA recognition and degradation. The RNA exosome is a protein complex that includes nucleases and is essential for host and viral RNA decay. Although the small RNAs produced by the RNA exosome do not activate RIG-I, several accessory factors of the RNA exosome promote RIG-I activation. Zinc-finger antiviral protein (ZAP) is an accessory factor that recognizes viral RNA and promotes viral RNA degradation via the RNA exosome. ZAPS is an alternative splicing form of ZAP and promotes RIG-I oligomerization and ATPase activity, resulting in RIG-I activation. DDX60 is another cofactor involved in the viral RNA degradation via the RNA exosome. The DDX60 protein promotes RIG-I signaling in a cell-type specific manner. These observations imply that viral RNA degradation and recognition are linked to each other. In this review, I discuss the links between recognition and degradation of viral RNA.


Subject(s)
DEAD Box Protein 58/immunology , Immunity, Innate/immunology , Interferon Type I/immunology , RNA, Viral/immunology , RNA, Viral/metabolism , Adenosine Triphosphatases/metabolism , DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , Humans , RNA-Binding Proteins/metabolism , Receptors, Immunologic
11.
Gastroenterology ; 147(2): 463-72, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24786893

ABSTRACT

BACKGROUND & AIMS: Cyclophilin inhibitors are being developed for treatment of hepatitis C virus (HCV) infection. They are believed to inhibit the HCV replication complex. We investigated whether cyclophilin inhibitors interact with interferon (IFN) signaling in cultured cells infected with HCV. METHODS: We used immunoblot assays to compare expression of IFN-stimulated genes (ISGs) and of components of IFN signaling in HCV-infected and uninfected cells. RESULTS: Incubation with IFN alfa induced expression of ISGs in noninfected cells and, to a lesser extent, in HCV-infected cells; addition of the cyclophilin inhibitor SCY-635 restored expression of ISG products in HCV-infected cells. SCY-635 reduced phosphorylation of double-strand RNA-dependent protein kinase (PKR) and its downstream factor eIF2α; the phosphorylated forms of these proteins are negative regulators of ISG translation. Cyclophilin A interacted physically with PKR; this interaction was disrupted by SCY-635. SCY-635 also suppressed PKR-mediated formation of stress granules. Cyclophilin inhibitors were found to inhibit PKR phosphorylation and stress granule formation in HCV-infected and uninfected cells. CONCLUSIONS: In cultured cells, cyclophilin inhibitors reverse the attenuation of the IFN response by HCV, in addition to their effects on HCV replication complex. Cyclophilin A regulation of PKR has been proposed as a mechanism for observed effects of cyclophilin inhibitors on IFN signaling. We found that cyclophilin inhibitors reduce phosphorylation of PKR and eIF2α during HCV infection to allow for translation of ISG products. Proteins in this pathway might be developed as targets for treatment of HCV infection.


Subject(s)
Antiviral Agents/pharmacology , Cyclophilin A/antagonists & inhibitors , Cyclosporins/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Interferon Regulatory Factors/metabolism , Liver/drug effects , eIF-2 Kinase/metabolism , Cell Line, Tumor , Cyclophilin A/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Hepacivirus/metabolism , Hepatitis C/enzymology , Hepatitis C/virology , Humans , Interferon Regulatory Factors/genetics , Interferon-alpha/metabolism , Liver/enzymology , Liver/virology , Phosphorylation , RNA, Messenger/metabolism , Signal Transduction/drug effects
12.
Hepatology ; 59(5): 1726-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24375637

ABSTRACT

UNLABELLED: Chronic hepatitis B virus (HBV) infection is a major public health problem worldwide. Although nucleos(t)ide analogs inhibiting viral reverse transcriptase are clinically available as anti-HBV agents, emergence of drug-resistant viruses highlights the need for new anti-HBV agents interfering with other targets. Here we report that cyclosporin A (CsA) can inhibit HBV entry into cultured hepatocytes. The anti-HBV effect of CsA was independent of binding to cyclophilin and calcineurin. Rather, blockade of HBV infection correlated with the ability to inhibit the transporter activity of sodium taurocholate cotransporting polypeptide (NTCP). We also found that HBV infection-susceptible cells, differentiated HepaRG cells and primary human hepatocytes expressed NTCP, while nonsusceptible cell lines did not. A series of compounds targeting NTCP could inhibit HBV infection. CsA inhibited the binding between NTCP and large envelope protein in vitro. Evaluation of CsA analogs identified a compound with higher anti-HBV potency, having a median inhibitory concentration <0.2 µM. CONCLUSION: This study provides a proof of concept for the novel strategy to identify anti-HBV agents by targeting the candidate HBV receptor, NTCP, using CsA as a structural platform.


Subject(s)
Antiviral Agents/pharmacology , Cyclosporine/pharmacology , Hepatitis B virus/drug effects , Hepatocytes/virology , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Virus Internalization/drug effects , Cells, Cultured , Hepatitis B virus/physiology , Humans , Virus Replication/drug effects
13.
J Biol Chem ; 288(44): 31715-27, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24025329

ABSTRACT

Virus infection is restricted by intracellular immune responses in host cells, and this is typically modulated by stimulation of cytokines. The cytokines and host factors that determine the host cell restriction against hepatitis B virus (HBV) infection are not well understood. We screened 36 cytokines and chemokines to determine which were able to reduce the susceptibility of HepaRG cells to HBV infection. Here, we found that pretreatment with IL-1ß and TNFα remarkably reduced the host cell susceptibility to HBV infection. This effect was mediated by activation of the NF-κB signaling pathway. A cytidine deaminase, activation-induced cytidine deaminase (AID), was up-regulated by both IL-1ß and TNFα in a variety of hepatocyte cell lines and primary human hepatocytes. Another deaminase APOBEC3G was not induced by these proinflammatory cytokines. Knockdown of AID expression impaired the anti-HBV effect of IL-1ß, and overexpression of AID antagonized HBV infection, suggesting that AID was one of the responsible factors for the anti-HBV activity of IL-1/TNFα. Although AID induced hypermutation of HBV DNA, this activity was dispensable for the anti-HBV activity. The antiviral effect of IL-1/TNFα was also observed on different HBV genotypes but not on hepatitis C virus. These results demonstrate that proinflammatory cytokines IL-1/TNFα trigger a novel antiviral mechanism involving AID to regulate host cell permissiveness to HBV infection.


Subject(s)
Cytidine Deaminase/biosynthesis , Gene Expression Regulation, Enzymologic , Hepatitis B virus/metabolism , Hepatitis B/metabolism , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , DNA, Viral/biosynthesis , DNA, Viral/genetics , DNA, Viral/immunology , Hep G2 Cells , Hepacivirus/genetics , Hepacivirus/immunology , Hepacivirus/metabolism , Hepatitis B/genetics , Hepatitis B/immunology , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mutation , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/genetics
14.
Cell Host Microbe ; 11(5): 492-503, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22607802

ABSTRACT

Bornaviruses are nonsegmented negative-strand RNA viruses that establish a persistent infection in the nucleus and occasionally integrate a DNA genome copy into the host chromosomal DNA. However, how these viruses achieve intranuclear infection remains unclear. We show that Borna disease virus (BDV), a mammalian bornavirus, closely associates with the cellular chromosome to ensure intranuclear infection. BDV generates viral factories within the nucleus using host chromatin as a scaffold. In addition, the viral ribonucleoprotein (RNP) interacts directly with the host chromosome throughout the cell cycle, using core histones as a docking platform. HMGB1, a host chromatin-remodeling DNA architectural protein, is required to stabilize RNP on chromosomes and for efficient BDV RNA transcription in the nucleus. During metaphase, the association of RNP with mitotic chromosomes allows the viral RNA to segregate into daughter cells and ensure persistent infection. Thus, bornaviruses likely evolved a chromosome-dependent life cycle to achieve stable intranuclear infection.


Subject(s)
Borna disease virus/physiology , Borna disease virus/pathogenicity , Cell Nucleus/virology , Virus Replication , Cell Cycle , Cell Line , Chromosome Segregation , HMGB1 Protein/metabolism , Histones/metabolism , Host-Pathogen Interactions , Humans , Nucleoproteins/metabolism , Protein Binding , RNA, Viral/metabolism , Transcription, Genetic , Viral Proteins/metabolism
15.
J Virol ; 85(23): 12170-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937656

ABSTRACT

Borna disease virus (BDV), a nonsegmented, negative-strand RNA virus, infects a wide variety of mammalian species and readily establishes a long-lasting, persistent infection in brain cells. Therefore, this virus could be a promising candidate as a novel RNA virus vector enabling stable gene expression in the central nervous system (CNS). Previous studies demonstrated that the 5' untranslated region of the genome is the only site for insertion and expression of a foreign gene. In this study, we established a novel BDV vector in which an additional transcription cassette has been inserted into an intercistronic noncoding region between the viral phosphoprotein (P) and matrix (M) genes. The recombinant BDV (rBDV) carrying green fluorescent protein (GFP) between the P and M genes, rBDV P/M-GFP, expressed GFP efficiently in cultured cells and rodent brains for a long period of time without attenuation. Furthermore, we generated a nonpropagating rBDV, ΔGLLP/M, which lacks the envelope glycoprotein (G) and a splicing intron within the polymerase gene (L), by the transcomplementation system with either transient or stable expression of the G gene. Interestingly, rBDV ΔGLLP/M established a persistent infection in cultured cells with stable expression of GFP in the absence of the expression of G. Using persistently infected rBDV ΔGLLP/M-infected cells, we determined the amino acid region in the cytoplasmic tail (CT) of BDV G important for the release of infectious rBDV particles and also demonstrated that the CT region may be critical for the generation of pseudotyped rBDV having vesicular stomatitis virus G protein. Our results revealed that the newly established BDV vector constitutes an alternative tool not only for stable expression of foreign genes in the CNS but also for understanding the mechanism of the release of enveloped virions.


Subject(s)
Borna disease virus/genetics , Brain/metabolism , DNA, Intergenic/genetics , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/metabolism , Viral Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Brain/virology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/virology , Chlorocebus aethiops , Fluorescent Antibody Technique, Indirect , Glycoproteins/metabolism , Green Fluorescent Proteins/genetics , Luciferases/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , Oligodendroglioma/virology , RNA, Viral , Rats , Rats, Inbred Lew , Transcription, Genetic , Vero Cells , Viral Envelope Proteins/metabolism , Viral Proteins/genetics
16.
J Vet Med Sci ; 73(9): 1243-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21566396

ABSTRACT

Borna disease virus (BDV) is a non-segmented, negative-strand RNA virus that is characterized by nuclear replication and persistent infection. A unique feature of BDV is that it releases only a small number of infectious particles from infected cells. Although these characteristics might make it difficult to obtain a large amount of recombinant viruses in a reverse genetics system, the mechanism underlying the budding or assembly of BDV particle has remained largely unknown. In this study, as a first step toward understanding the virion formation of BDV, we investigated the intracellular distribution and mobility of the fluorescent marker fusion envelope glycoprotein (G) of BDV in living cells. Expression analysis revealed that fusion proteins seem to cleave into functional subunits and localize in the endoplasmic reticulum (ER)/Golgi apparatus, as well as the authentic BDV G. Furthermore, we demonstrated using fluorescence recovery after photobleaching analysis that BDV G fluorescence shows rapid recovery in both the ER/Golgi and plasma membrane regions, indicating that BDV G fusion protein may be a useful tool to investigate not only the maturation of BDV G but also the budding and assembly of BDV particles in living cells.


Subject(s)
Borna disease virus/physiology , Gene Expression Regulation, Viral/physiology , Luminescent Proteins/metabolism , Viral Fusion Proteins/metabolism , Animals , Cell Membrane , Chlorocebus aethiops , Endoplasmic Reticulum , Fluorescence , Golgi Apparatus , Vero Cells , Viral Fusion Proteins/genetics , Red Fluorescent Protein
17.
J Virol ; 85(9): 4567-71, 2011 May.
Article in English | MEDLINE | ID: mdl-21325425

ABSTRACT

In a previous study, we demonstrated that transgenic mice that express Borna disease virus (BDV) phosphoprotein (P) in astrocytes show striking neurobehavioral abnormalities resembling those in BDV-infected animals. To understand the molecular disturbances induced by the expression of P in astrocytes, we performed microarray analysis with cultured astroglial cells transiently expressing P. We showed that expression of insulin-like growth factor binding protein 3 mRNA increases not only in P-expressing cultured cells but also in astrocytes from the cerebella of P transgenic mice (P-Tg). Furthermore, we demonstrated that insulin-like growth factor signaling is disturbed in the P-Tg cerebellum, a factor that might be involved in the increased vulnerability of Purkinje cell neurons in the brain.


Subject(s)
Astrocytes/virology , Borna disease virus/pathogenicity , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Phosphoproteins/metabolism , Viral Structural Proteins/metabolism , Animals , Cells, Cultured , Gene Expression Profiling , Mice , Mice, Transgenic , Microarray Analysis , Up-Regulation
18.
Nature ; 463(7277): 84-7, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20054395

ABSTRACT

Retroviruses are the only group of viruses known to have left a fossil record, in the form of endogenous proviruses, and approximately 8% of the human genome is made up of these elements. Although many other viruses, including non-retroviral RNA viruses, are known to generate DNA forms of their own genomes during replication, none has been found as DNA in the germline of animals. Bornaviruses, a genus of non-segmented, negative-sense RNA virus, are unique among RNA viruses in that they establish persistent infection in the cell nucleus. Here we show that elements homologous to the nucleoprotein (N) gene of bornavirus exist in the genomes of several mammalian species, including humans, non-human primates, rodents and elephants. These sequences have been designated endogenous Borna-like N (EBLN) elements. Some of the primate EBLNs contain an intact open reading frame (ORF) and are expressed as mRNA. Phylogenetic analyses showed that EBLNs seem to have been generated by different insertional events in each specific animal family. Furthermore, the EBLN of a ground squirrel was formed by a recent integration event, whereas those in primates must have been formed more than 40 million years ago. We also show that the N mRNA of a current mammalian bornavirus, Borna disease virus (BDV), can form EBLN-like elements in the genomes of persistently infected cultured cells. Our results provide the first evidence for endogenization of non-retroviral virus-derived elements in mammalian genomes and give novel insights not only into generation of endogenous elements, but also into a role of bornavirus as a source of genetic novelty in its host.


Subject(s)
Bornaviridae/genetics , Genes, Viral/genetics , Genome/genetics , Mammals/genetics , Mammals/virology , Virus Integration/genetics , Amino Acid Sequence , Animals , Borna disease virus/genetics , Borna disease virus/physiology , Bornaviridae/physiology , Cell Line , Conserved Sequence/genetics , Evolution, Molecular , Host-Pathogen Interactions/genetics , Humans , Models, Genetic , Molecular Sequence Data , Open Reading Frames/genetics , Phylogeny , Reverse Transcription , Time Factors
19.
J Virol ; 83(23): 12622-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776128

ABSTRACT

Borna disease virus (BDV) is characterized by highly neurotropic infection. BDV enters its target cells using virus surface glycoprotein (G), but the cellular molecules mediating this process remain to be elucidated. We demonstrate here that the N-terminal product of G, GP1, interacts with the 78-kDa chaperone protein BiP. BiP was found at the surface of BDV-permissive cells, and anti-BiP antibody reduced BDV infection as well as GP1 binding to the cell surface. We also reveal that BiP localizes at the synapse of neurons. These results indicate that BiP may participate in the cell surface association of BDV.


Subject(s)
Borna disease virus/physiology , Heat-Shock Proteins/metabolism , Protein Interaction Mapping , Viral Structural Proteins/metabolism , Virus Internalization , Cell Line , Endoplasmic Reticulum Chaperone BiP , Humans , Oligodendroglia/virology , Protein Binding
20.
Microbes Infect ; 11(3): 394-402, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19397879

ABSTRACT

Borna disease virus (BDV) is a non-segmented, negative-sense RNA virus and has the property of persistently infecting the cell nucleus. BDV encodes a 10-kDa non-structural protein, X, which is a negative regulator of viral polymerase activity but is essential for virus propagation. Recently, we have demonstrated that interaction of X with the viral polymerase cofactor, phosphoprotein (P), facilitates translocation of P from the nucleus to the cytoplasm. However, the mechanism by which the intracellular localization of X is controlled remains unclear. In this report, we demonstrate that BDV X interacts with the 71kDa molecular chaperon protein, Hsc70. Immunoprecipitation assays revealed that Hsc70 associates with the same region of X as P and, interestingly, that expression of P interferes competitively with the interaction between X and Hsc70. A heat shock experiment revealed that BDV X translocates into the nucleus, dependent upon the nuclear accumulation of Hsc70. Furthermore, we show that knockdown of Hsc70 by short interfering RNA decreases the nuclear localization of both X and P and markedly reduces the expression of viral genomic RNA in persistently infected cells. These data indicate that Hsc70 may be involved in viral replication by regulating the intracellular distribution of X.


Subject(s)
Borna disease virus/physiology , HSC70 Heat-Shock Proteins/metabolism , Protein Interaction Mapping , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , Virus Replication , Cell Line , Humans , Immunoprecipitation
SELECTION OF CITATIONS
SEARCH DETAIL
...