Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Front Med (Lausanne) ; 9: 867478, 2022.
Article in English | MEDLINE | ID: mdl-35463007

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) is a replication-competent human retrovirus associated with two distinct types of diseases: a malignancy of mature CD4+ T cells called adult T-cell leukemia-lymphoma (ATL) and a chronic inflammatory central nervous system disease HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). It was the first human retrovirus ever associated with a human cancer. Although most HTLV-1-infected individuals remain asymptomatic for life, a subpopulation develops ATL or HAM/TSP. Although the factors that cause these different manifestations of HTLV-1 infection are not fully understood, accumulating evidence suggests that the complex virus-host interactions, as well as the host immune response against HTLV-1 infection, appear to regulate the development of HTLV-1-associated diseases. This review outlines and discusses the current understanding, ongoing developments, and future perspectives of HTLV-1 research.

2.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: mdl-33827947

ABSTRACT

RNA viruses demonstrate a vast range of variants, called quasispecies, due to error-prone replication by viral RNA-dependent RNA polymerase. Although live attenuated vaccines are effective in preventing RNA virus infection, there is a risk of reversal to virulence after their administration. To test the hypothesis that high-fidelity viral polymerase reduces the diversity of influenza virus quasispecies, resulting in inhibition of reversal of the attenuated phenotype, we first screened for a high-fidelity viral polymerase using serial virus passages under selection with a guanosine analog ribavirin. Consequently, we identified a Leu66-to-Val single amino acid mutation in polymerase basic protein 1 (PB1). The high-fidelity phenotype of PB1-L66V was confirmed using next-generation sequencing analysis and biochemical assays with the purified influenza viral polymerase. As expected, PB1-L66V showed at least two-times-lower mutation rates and decreased misincorporation rates, compared to the wild type (WT). Therefore, we next generated an attenuated PB1-L66V virus with a temperature-sensitive (ts) phenotype based on FluMist, a live attenuated influenza vaccine (LAIV) that can restrict virus propagation by ts mutations, and examined the genetic stability of the attenuated PB1-L66V virus using serial virus passages. The PB1-L66V mutation prevented reversion of the ts phenotype to the WT phenotype, suggesting that the high-fidelity viral polymerase could contribute to generating an LAIV with high genetic stability, which would not revert to the pathogenic virus.IMPORTANCE The LAIV currently in use is prescribed for actively immunizing individuals aged 2 to 49 years. However, it is not approved for infants and elderly individuals, who actually need it the most, because it might prolong virus propagation and cause an apparent infection in these individuals, due to their weak immune systems. Recently, reversion of the ts phenotype of the LAIV strain currently in use to a pathogenic virus was demonstrated in cultured cells. Thus, the generation of mutations associated with enhanced virulence in LAIV should be considered. In this study, we isolated a novel influenza virus strain with a Leu66-to-Val single amino acid mutation in PB1 that displayed a significantly higher fidelity than the WT. We generated a novel LAIV candidate strain harboring this mutation. This strain showed higher genetic stability and no ts phenotype reversion. Thus, our high-fidelity strain might be useful for the development of a safer LAIV.


Subject(s)
Influenza A virus/genetics , Influenza A virus/physiology , Influenza Vaccines , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/genetics , Amino Acid Substitution , Animals , Antiviral Agents/pharmacology , Dogs , Drug Resistance, Viral , Influenza A virus/drug effects , Madin Darby Canine Kidney Cells , Mutation , Phenotype , Protein Engineering , RNA-Dependent RNA Polymerase/chemistry , Ribavirin/pharmacology , Vaccines, Attenuated , Viral Plaque Assay , Viral Proteins/chemistry , Viral Proteins/metabolism
3.
Virol J ; 16(1): 160, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31856855

ABSTRACT

BACKGROUND: EOS plays an important role in maintaining the suppressive function of regulatory T cells (Tregs), and induces a regulated transformation of Tregs into T helper-like cells, which are capable of secreting proinflammatory cytokines in response to specific inflammatory signals. Meanwhile, significant reduction in Treg activity along with production of proinflammatory cytokines has been reported in patients with HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). METHODS: In this study, to examine whether there is an alteration in EOS expression in peripheral blood mononuclear cells (PBMCs) derived from HTLV-1-infected individuals especially HAM/TSP, we investigated the expression of HTLV-1 tax genotype, proviral load (PVL), and the mRNA expression of tax, HBZ and EOS in HTLV-1 infected individuals including adult T-cell leukemia/lymphoma (ATL), HAM/TSP, or asymptomatic carriers. The expression levels of EOS mRNA and protein in various HTLV-1-infected or uninfected human T-cell lines were also investigated. RESULTS: EOS was highly expressed at the protein level in most HTLV-1 infected T-cell lines, and was augmented after the HTLV-1 regulatory factor Tax was induced in a Tax-inducible JPX-9 cell line. Immunoprecipitation experiments demonstrated a physical interaction between EOS and the viral regulatory protein Tax, but not HBZ. Meanwhile, there was a significant decrease in EOS mRNA levels in PBMCs of HTLV-1 infected individuals irrespective of their clinical statuses. We found an inverse correlation between EOS mRNA levels and HTLV-1 PVL in ATL patients, and positive correlations between both EOS mRNA load and PVL, and EOS and HBZ mRNA load in HAM/TSP patients, whereas this correlation was not observed in other clinical statuses. CONCLUSIONS: These findings suggest that both Tax and HBZ can alter the expression of EOS through undetermined mechanisms, and dysregulated expression of EOS in PBMCs of HTLV-1 infected individuals may contribute to the pathological progression of HTLV-1-associated diseases, such as ATL and HAM/TSP.


Subject(s)
Down-Regulation , Gene Products, tax/metabolism , HTLV-I Infections/virology , Host-Pathogen Interactions , Human T-lymphotropic virus 1/growth & development , Ikaros Transcription Factor/metabolism , Leukocytes, Mononuclear/virology , Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Profiling , Humans , Immunoprecipitation , Retroviridae Proteins/metabolism
4.
J Virol ; 93(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31462570

ABSTRACT

In various positive-sense single-stranded RNA viruses, a low-fidelity viral RNA-dependent RNA polymerase (RdRp) confers attenuated phenotypes by increasing the mutation frequency. We report a negative-sense single-stranded RNA virus RdRp mutant strain with a mutator phenotype. Based on structural data of RdRp, rational targeting of key residues, and screening of fidelity variants, we isolated a novel low-fidelity mutator strain of influenza virus that harbors a Tyr82-to-Cys (Y82C) single-amino-acid substitution in the PB1 polymerase subunit. The purified PB1-Y82C polymerase indeed showed an increased frequency of misincorporation compared with the wild-type PB1 in an in vitro biochemical assay. To further investigate the effects of position 82 on PB1 polymerase fidelity, we substituted various amino acids at this position. As a result, we isolated various novel mutators other than PB1-Y82C with higher mutation frequencies. The structural model of influenza virus polymerase complex suggested that the Tyr82 residue, which is located at the nucleoside triphosphate entrance tunnel, may influence a fidelity checkpoint. Interestingly, although the PB1-Y82C variant replicated with wild-type PB1-like kinetics in tissue culture, the 50% lethal dose of the PB1-Y82C mutant was 10 times lower than that of wild-type PB1 in embryonated chicken eggs. In conclusion, our data indicate that the Tyr82 residue of PB1 has a crucial role in regulating polymerase fidelity of influenza virus and is closely related to attenuated pathogenic phenotypes in vivoIMPORTANCE Influenza A virus rapidly acquires antigenic changes and antiviral drug resistance, which limit the effectiveness of vaccines and drug treatments, primarily owing to its high rate of evolution. Virus populations formed by quasispecies can contain resistance mutations even before a selective pressure is applied. To study the effects of the viral mutation spectrum and quasispecies, high- and low-fidelity variants have been isolated for several RNA viruses. Here, we report the discovery of a low-fidelity RdRp variant of influenza A virus that contains a substitution at Tyr82 in PB1. Viruses containing the PB1-Y82C substitution showed growth kinetics and viral RNA synthesis levels similar to those of the wild-type virus in cell culture; however, they had significantly attenuated phenotypes in a chicken egg infection experiment. These data demonstrated that decreased RdRp fidelity attenuates influenza A virus in vivo, which is a desirable feature for the development of safer live attenuated vaccine candidates.


Subject(s)
Influenza A virus/genetics , Mutation , Viral Proteins/genetics , Viral Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Dogs , HEK293 Cells , Humans , Influenza A virus/enzymology , Influenza A virus/metabolism , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Models, Molecular , Phenotype , Polymorphism, Single Nucleotide , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Virus Replication/genetics
5.
Retrovirology ; 15(1): 72, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30400920

ABSTRACT

BACKGROUND: Among human T cell leukemia virus type 1 (HTLV-1)-infected individuals, there is an association between HTLV-1 tax subgroups (subgroup-A or subgroup-B) and the risk of HAM/TSP in the Japanese population. To investigate the role of HTLV-1 subgroups in viral pathogenesis, we studied the functional difference in the subgroup-specific viral transcriptional regulators Tax and HBZ using microarray analysis, reporter gene assays, and evaluation of viral-host protein-protein interaction. RESULTS: (1) Transcriptional changes in Jurkat Tet-On human T-cells that express each subgroup of Tax or HBZ protein under the control of an inducible promoter revealed different target gene profiles; (2) the number of differentially regulated genes induced by HBZ was 2-3 times higher than that induced by Tax; (3) Tax and HBZ induced the expression of different classes of non-coding RNAs (ncRNAs); (4) the chemokine CXCL10, which has been proposed as a prognostic biomarker for HAM/TSP, was more efficiently induced by subgroup-A Tax (Tax-A) than subgroup-B Tax (Tax-B), in vitro as well as in unmanipulated (ex vivo) PBMCs obtained from HAM/TSP patients; (5) reporter gene assays indicated that although transient Tax expression in an HTLV-1-negative human T-cell line activated the CXCL10 gene promoter through the NF-κB pathway, there was no difference in the ability of each subgroup of Tax to activate the CXCL10 promoter; however, (6) chromatin immunoprecipitation assays showed that the ternary complex containing Tax-A is more efficiently recruited onto the promoter region of CXCL10, which contains two NF-κB binding sites, than that containing Tax-B. CONCLUSIONS: Our results indicate that different HTLV-1 subgroups are characterized by different patterns of host gene expression. Differential expression of pathogenesis-related genes by subgroup-specific Tax or HBZ may be associated with the onset of HAM/TSP.


Subject(s)
Gene Products, tax/genetics , HTLV-I Infections/genetics , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/pathogenicity , Paraparesis, Tropical Spastic/genetics , Trans-Activators/genetics , Adult , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line , Female , Human T-lymphotropic virus 1/classification , Humans , Jurkat Cells , Male , Microarray Analysis , Middle Aged , Paraparesis, Tropical Spastic/virology , RNA, Untranslated/genetics , Retroviridae Proteins/genetics , Risk Factors , Transcriptome , Viral Proteins/genetics
6.
Virol J ; 14(1): 234, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29202792

ABSTRACT

BACKGROUND: Chemokine (C-C motif) ligand 1 (CCL1) is produced by activated monocytes/ macrophages and T-lymphocytes, and acts as a potent attractant for Th2 cells and a subset of T-regulatory (Treg) cells. Previous reports have indicated that CCL1 is overexpressed in adult T-cell leukemia cells, mediating an autocrine anti-apoptotic loop. Because CCL1 is also known as a potent chemoattractant that plays a major role in inflammatory processes, we investigated the role of CCL1 in the pathogenesis of human T-cell leukemia virus type 1 (HTLV-1)-associated myelopathy/tropical spastic paraparesis (HAM/TSP). RESULTS: The results showed that: (1) CCL1 was preferentially expressed in HAM/TSP-derived HTLV-1-infected T-cell lines, (2) CCL1 expression was induced along with Tax expression in the Tax-inducible T-cell line JPX9, (3) transient Tax expression in an HTLV-1-negative T-cell line activated the CCL1 gene promoter, (4) plasma levels of CCL1 were significantly higher in patients with HAM/TSP than in HTLV-1-seronegative patients with multiple sclerosis and HTLV-1-infected asymptomatic healthy carriers, and (5) minocycline inhibited the production of CCL1 in HTLV-1-infected T-cell lines. CONCLUSIONS: The present results suggest that elevated CCL1 levels may be associated with the pathogenesis of HAM/TSP. Although further studies are required to determine the in vivo significance, minocycline may be considered as a potential candidate for the long-term treatment of HAM/TSP via its anti-inflammatory effects, which includes the inhibition of CCL1 expression.


Subject(s)
Chemokine CCL1/genetics , Down-Regulation/drug effects , Gene Products, tax/metabolism , Minocycline/pharmacology , Paraparesis, Tropical Spastic/physiopathology , Up-Regulation/genetics , Adult , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cell Line , Chemokine CCL1/metabolism , Flow Cytometry , Humans , Minocycline/therapeutic use , Paraparesis, Tropical Spastic/drug therapy , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Receptors, CCR8/metabolism , Transcriptional Activation/drug effects
7.
Virol J ; 14(1): 130, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28716148

ABSTRACT

BACKGROUND: Detection of specific immune responses against cancer/testis antigen NY-ESO-1 was recently reported in patients with adult T-cell leukemia/lymphoma (ATL) and human T-cell leukemia virus type 1 (HTLV-1)-infected asymptomatic carriers (ACs). However, the relationship of the responses with the HTLV-1 proviral load (PVL) and the levels of viral gene expression remain unclear. FINDINGS: We measured plasma levels of autoantibodies to NY-ESO-1 immunogenic tumor antigen in HTLV-1-infected individuals with different clinical status, and in healthy controls. Data were compared to tax and HBZ mRNA levels, and PVL. Plasma anti-NY-ESO-1 antibody was detectable in 13.7% (7/51) of ACs, 29.2% (38/130) of patients with HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP), and 18.9% (10/53) of patients with ATL. Anti-NY-ESO-1 plasma levels were significantly higher in patients with HAM/TSP than in patients with ATL or ACs. Anti-NY-ESO-1 levels were not associated with PVL or the expression levels of tax and HBZ mRNA among HTLV-1-infected individuals, regardless of clinical status. CONCLUSIONS: The present results indicate the strong humoral immune response against NY-ESO-1 in natural HTLV-1 infection, irrespective of the clinical status. The higher immunoreactivity against NY-ESO-1 is not simply associated with the levels of both HTLV-1 gene expression and the number of infected cells in vivo. Rather, it might reflect chronic and generalized immune activation in infected individuals.


Subject(s)
Antigens, Neoplasm/immunology , Autoantibodies/blood , HTLV-I Infections/pathology , Membrane Proteins/immunology , Plasma/chemistry , Testis/immunology , Humans , Male , Prevalence
8.
Sci Rep ; 7: 45043, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28344335

ABSTRACT

Influenza A virus (IAV) membrane proteins hemagglutinin (HA) and neuraminidase (NA) are determinants of virus infectivity, transmissibility, pathogenicity, host specificity, and major antigenicity. HA binds to a virus receptor, a sialoglycoprotein or sialoglycolipid, on the host cell and mediates virus attachment to the cell surface. The hydrolytic enzyme NA cleaves sialic acid from viral receptors and accelerates the release of progeny virus from host cells. In this study, we identified a novel function of HA and NA as machinery for viral motility. HAs exchanged binding partner receptors iteratively, generating virus movement on a receptor-coated glass surface instead of a cell surface. The virus movement was also dependent on NA. Virus movement mediated by HA and NA resulted in a three to four-fold increase in virus internalisation by cultured cells. We concluded that cooperation of HA and NA moves IAV particles on a cell surface and enhances virus infection of host cells.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Movement , Neuraminidase/metabolism , Virus Internalization , Animals , Cell Line, Tumor , Dogs , HEK293 Cells , Humans , Influenza A virus/metabolism , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Receptors, Virus/metabolism
9.
J Virol ; 91(6)2017 03 15.
Article in English | MEDLINE | ID: mdl-28053101

ABSTRACT

Vaccination is considered the most effective preventive means for influenza control. The development of a master virus with high growth and genetic stability, which may be used for the preparation of vaccine viruses by gene reassortment, is crucial for the enhancement of vaccine performance and efficiency of production. Here, we describe the generation of a high-fidelity and high-growth influenza vaccine master virus strain with a single V43I amino acid change in the PB1 polymerase of the high-growth A/Puerto Rico/8/1934 (PR8) master virus. The PB1-V43I mutation was introduced to increase replication fidelity in order to design an H1N1 vaccine strain with a low error rate. The PR8-PB1-V43I virus exhibited good replication compared with that of the parent PR8 virus. In order to compare the efficiency of egg adaptation and the occurrence of gene mutations leading to antigenic alterations, we constructed 6:2 genetic reassortant viruses between the A(H1N1)pdm09 and the PR8-PB1-V43I viruses; hemagglutinin (HA) and neuraminidase (NA) were from the A(H1N1)pdm09 virus, and the other genes were from the PR8 virus. Mutations responsible for egg adaptation mutations occurred in the HA of the PB1-V43I reassortant virus during serial egg passages; however, in contrast, antigenic mutations were introduced into the HA gene of the 6:2 reassortant virus possessing the wild-type PB1. This study shows that the mutant PR8 virus possessing the PB1 polymerase with the V43I substitution may be utilized as a master virus for the generation of high-growth vaccine viruses with high polymerase fidelity, low error rates of gene replication, and reduced antigenic diversity during virus propagation in eggs for vaccine production.IMPORTANCE Vaccination represents the most effective prophylactic option against influenza. The threat of emergence of influenza pandemics necessitates the ability to generate vaccine viruses rapidly. However, as the influenza virus exhibits a high mutation rate, vaccines must be updated to ensure a good match of the HA and NA antigens between the vaccine and the circulating strain. Here, we generated a genetically stable master virus of the A/Puerto Rico/8/1934 (H1N1) backbone encoding an engineered high-fidelity viral polymerase. Importantly, following the application of the high-fidelity PR8 backbone, no mutation resulting in antigenic change was introduced into the HA gene during propagation of the A(H1N1)pdm09 candidate vaccine virus. The low error rate of the present vaccine virus should decrease the risk of generating mutant viruses with increased virulence. Therefore, our findings are expected to be useful for the development of prepandemic vaccines and live attenuated vaccines with higher safety than that of the present candidate vaccines.


Subject(s)
Antigens, Viral/genetics , Influenza A virus/growth & development , Influenza Vaccines/immunology , Reassortant Viruses/growth & development , Amino Acid Substitution , Antigens, Viral/immunology , Influenza A virus/genetics , Influenza Vaccines/genetics , Point Mutation , Reassortant Viruses/genetics , Technology, Pharmaceutical/methods , Viral Proteins/genetics , Virology/methods
10.
BMC Infect Dis ; 16: 380, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27503338

ABSTRACT

BACKGROUND: Seasonally prevalent H1N1 and H3N2 influenza A viruses have evolved by antigenic drift; this evolution has resulted in the acquisition of asparagine (N)-linked glycosylation sites (NGSs) in the globular head of hemagglutinin (HA), thereby affecting the antigenic and receptor-binding properties, as well as virulence. An epidemiological survey indicated that although the traditional seasonal H1N1 strain had disappeared, H3N2 became predominant again in the seasons (2010-11 and 2011-12) immediately following the H1N1 pandemic of 2009. Interestingly, although the 2009 pandemic H1N1 strain (H1N1pdm09) lacks additional NGSs, clinically isolated H3N2 strains obtained during these seasons gained N (Asn) residues at positions 45 and 144 of HA that forms additional NGSs. METHODS: To investigate whether these NGSs are associated with re-emergence of H3N2 within the subtype, we tested the effect of amino acid substitutions on neutralizing activity by using the antisera raised against H3N2 strains with or without additional NGSs. Furthermore, because the N residue at position 144 of HA was identified as the site of mismatch between the vaccine and epidemic strains of 2011-2012, we generated mutant viruses by reverse genetics and tested the functional importance of this particular NGS for antibody-mediated neutralization by intranasal inoculation of mice. RESULTS: The results indicated that amino acid substitution at residue 144 significantly affected neutralization activity, acting as an escape mutation. CONCLUSIONS: Our data suggest that the newly acquired NGSs in the HA globular head may play an important role in the re-emergence of endemic seasonal H3N2 strain by aiding the escape from humoral immunity.


Subject(s)
Hemagglutinins/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/virology , Virulence Factors/genetics , Amino Acid Substitution , Animals , Antibodies, Neutralizing/immunology , Databases, Factual , Glycosylation , Hemagglutination Inhibition Tests , Hemagglutinins/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A virus/genetics , Influenza A virus/immunology , Influenza, Human/epidemiology , Japan/epidemiology , Mice , Mutation , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Seasons , Virulence
11.
Retrovirology ; 13: 29, 2016 Apr 27.
Article in English | MEDLINE | ID: mdl-27117327

ABSTRACT

BACKGROUND: Human T cell leukemia virus type 1 (HTLV-1) basic leucine zipper factor (HBZ), which is encoded by a minus strand mRNA, is thought to play important roles in the development of adult T-cell leukemia (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). However, a comprehensive analysis of HBZ, including mRNA and protein expression, humoral immunoreactivity against HBZ, and HTLV-1 proviral load (PVL), in HTLV-1-infected individuals with different clinical status has not been reported previously. RESULTS: In this study, using novel monoclonal antibody-based in-house enzyme-linked immunosorbent assay systems, we report the absolute quantification of HBZ protein and its plasma antibody in clinical samples from HTLV-1-infected individuals with different clinical status. The data were compared to both HBZ mRNA levels and PVL. The results showed that plasma anti-HBZ antibody was detectable only in 10.4 % (5/48) of asymptomatic carriers (ACs), 10.8 % (13/120) of HAM/TSP patients, and 16.7 % (7/42) of ATL patients. HBZ protein was detected in three out of five patients with acute ATL, but was not detected in patients with HAM/TSP (0/10) or ACs (0/4). Thus, an antibody response to HBZ was not associated with the PVL or the expression of HBZ (both at the mRNA and protein levels) or the clinical status of the infection. CONCLUSIONS: The present results emphasize the extremely low expression and immunogenicity of HBZ in natural HTLV-1 infection. However, there is a possibility that the low but distinct expression of HBZ protein in PBMCs is associated with the survival of HTLV-1-infected cells and the development of ATL.


Subject(s)
Antibodies, Viral/blood , Basic-Leucine Zipper Transcription Factors/blood , HTLV-I Infections/pathology , HTLV-I Infections/virology , Plasma/chemistry , Retroviridae Proteins/blood , Viral Load , Adult , Enzyme-Linked Immunosorbent Assay , Female , HTLV-I Infections/immunology , Humans , Male , Middle Aged , RNA, Messenger/blood , RNA, Viral/blood
12.
Vaccine ; 34(3): 328-33, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26657023

ABSTRACT

In April 2013, the first three fatal cases of human infection with an avian influenza A virus (H7N9) were reported in China. Because of a pandemic threat by this virus, we have commenced to develop candidate vaccine viruses (CVVs). Three 6:2 genetic reassortant viruses with different hemagglutinin (HA) sequences, NIIDRG-10, -10.1 and -10.2, were generated by a reverse genetics technique between the high egg-growth master virus, A/Puerto Rico/8/34 (H1N1) and A/Anhui/1/2013 (H7N9), kindly provided by the Chinese Center for Disease Control and Prevention. The different HA gene sequences of the three CVVs were derived from the original virus stock. NIIDRG-10 possesses HA, whose sequence is identical to that of the original A/Anhui/1/2013 (H7N9) in the Global Initiative on Sharing Avian Influenza Data (EPI439507), while NIIDRG-10.1 and -10.2 possess amino acid differences, A125T and N123D/N149D, respectively, compared with NIIDRG-10. NIIDRG-10 replicated in embryonated chicken eggs with low hemagglutination titer 128, whereas NIIDRG-10.1 and -10.2 grew well with hemagglutination titer 1024. These viruses reacted well with a ferret antiserum raised against the original A/Anhui/1/2013 virus. Ferret antiserum against NIIDRG-10.1 reacted well with A/Anhui/1/2013 similar to the homologous virus NIIDRG-10.1. These results indicated that NIIDRG-10.1 passed the two-way test of antigenic identity. In contrast, the ferret antiserum against NIIDRG-10.2 reacted with A/Anhui/1/2013 at an 8-fold lower hemagglutination inhibition titer than with the homologous virus NIIDRG-10.2, indicating an antigenic change. The total and HA protein yields of NIIDRG-10.1 were 14.7 and 6.9 µg/ml, respectively, similar to those levels of high-yield seed viruses of seasonal influenza vaccines. NIIDRG-10.1 was approved as one of the CVVs for H7N9 viruses by the WHO in 2013. The candidate vaccine derived from NIIDRG-10.1 is currently being evaluated in a phase II clinical study in Japan.


Subject(s)
Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Reassortant Viruses/immunology , Animals , Antibodies, Viral/blood , Clinical Trials, Phase II as Topic , Ferrets , Hemagglutination Tests , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza Vaccines/genetics , Influenza Vaccines/isolation & purification , Japan , Reassortant Viruses/genetics , Reverse Genetics
13.
J Gen Virol ; 95(Pt 12): 2677-2682, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25114028

ABSTRACT

The aim of this study was to investigate the infection and replication of swine-derived hepatitis E virus (HEV) in primary cultured human hepatocytes (PHCs). Hepatocytes were cultured from the resected normal livers of patients with metastatic tumours. These cultured hepatocytes were infected with swine-derived genotype 3 or 4 HEV. Viral replication was monitored using reverse transcriptase-quantitative PCR. The amount of HEV RNA increased in the culture media and cells following infection. Immunofluorescence staining implied that the spread of HEV infection in hepatocytes was attributed mainly to cell-to-cell transmission via the cell membrane. The sequences of the inoculated and propagated HEV were determined to examine whether sequence variation occurred during infection. Sequence analysis showed that there were no differences between inoculated and propagated HEV, demonstrating that in vitro infection and replication of swine HEV in PHCs occurred without sequence variation.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/veterinary , Hepatocytes/virology , Swine Diseases/virology , Animals , Cells, Cultured , Genome, Viral , Genotype , Hepatitis E/virology , Hepatitis E virus/classification , Hepatitis E virus/genetics , Humans , Swine
14.
ACS Chem Biol ; 9(6): 1273-83, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24689980

ABSTRACT

The heat shock response is an evolutionarily conserved, stress-responsive signaling pathway that adapts cellular proteostasis in response to pathologic insult. In metazoans, the heat shock response primarily functions through the posttranslational activation of heat shock factor 1 (HSF1), a stress-responsive transcription factor that induces the expression of cytosolic proteostasis factors including chaperones, cochaperones, and folding enzymes. HSF1 is a potentially attractive therapeutic target to ameliorate pathologic imbalances in cellular proteostasis associated with human disease, although the underlying impact of stress-independent HSF1 activation on cellular proteome composition remains to be defined. Here, we employ a highly controllable, ligand-regulated HSF1 that activates HSF1 to levels compatible with those that could be achieved using selective small molecule HSF1 activators. Using a combination of RNAseq and quantitative proteomics, we define the impact of stress-independent HSF1 activation on the composition of the cellular proteome. We show that stress-independent HSF1 activation selectively remodels cytosolic proteostasis pathways without globally influencing the composition of the cellular proteome. Furthermore, we show that stress-independent HSF1 activation decreases intracellular aggregation of a model polyglutamine-containing protein and reduces the cellular toxicity of environmental toxins like arsenite that disrupt cytosolic proteostasis. Collectively, our results reveal a proteome-level view of stress-independent HSF1 activation, providing a framework to establish therapeutic approaches to correct pathologic imbalances in cellular proteostasis through the selective targeting of HSF1.


Subject(s)
Biomarkers/analysis , DNA-Binding Proteins/metabolism , High-Throughput Nucleotide Sequencing , Proteome/analysis , Stress, Physiological , Transcription Factors/metabolism , Blotting, Western , Chromatography, Liquid , DNA-Binding Proteins/genetics , Gene Regulatory Networks , Genome, Human , HEK293 Cells , Heat Shock Transcription Factors , Humans , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Transcription Factors/genetics
15.
J Interferon Cytokine Res ; 31(11): 847-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21992152

ABSTRACT

Human myxovirus resistance gene A (MxA) is a type I interferon-inducible protein and exhibits the antiviral activity against a variety of RNA viruses, including influenza virus. Previously, we reported that MxA accelerates cell death of influenza virus-infected cells through caspase-dependent and -independent mechanisms. Similar to other viruses, influenza virus infection induces endoplasmic reticulum (ER) stress, which is one of cell death inducers. Here, we have demonstrated that MxA enhances ER stress signaling in cells infected with influenza virus. ER stress-induced events, such as expression of BiP mRNA and processing of XBP1 mRNA, were upregulated in cells expressing MxA by treatment with an ER stress inducer, tunicamycin (TM), as well as influenza virus infection. TM-induced cell death was also accelerated by MxA. Furthermore, we showed that MxA interacts with BiP and overexpression of BiP reduces MxA-promoted ER stress signaling. Because cell death in virus-infected cells is one of ultimate anti-virus mechanisms, we propose that MxA-enhanced ER stress signaling is a part of the antiviral activity of MxA by accelerating cell death.


Subject(s)
Endoplasmic Reticulum Stress , GTP-Binding Proteins/metabolism , Animals , Cell Death/drug effects , GTP-Binding Proteins/immunology , HEK293 Cells , HeLa Cells , Humans , Influenza A virus/isolation & purification , Influenza, Human/immunology , Influenza, Human/virology , Mice , Myxovirus Resistance Proteins , Signal Transduction , Swiss 3T3 Cells , Tunicamycin/pharmacology
16.
Uirusu ; 59(1): 1-11, 2009 Jun.
Article in Japanese | MEDLINE | ID: mdl-19927983

ABSTRACT

Studies using cell-free RNA synthesis systems and reverse genetics have been contributing to understanding of the molecular mechanism of replication and transcription of the influenza virus genome, which is the most essential process through the virus life cycle. Recently, it is noted that this mechanism is also involved in host range determination of the virus. In the light of the fact that viruses resistant to previously developed anti-influenza virus drugs emerge, establishment of a rational screening strategy of drugs for novel molecular targets is highly required. Further to clarify the detailed function of viral factors involved in replication and transcription of the virus genome and to devise anti-viral methods, determination of the 3D structures of viral factors should give a breakthrough. In this review, we summarize the recent accumulating information on the 3D structures of viral factors and discuss their function based on their structures.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/physiology , Genome, Viral/genetics , Orthomyxoviridae/enzymology , Orthomyxoviridae/genetics , DNA Replication/genetics , Protein Conformation , Transcription, Genetic , Virus Replication/genetics
17.
Mol Immunol ; 46(5): 858-65, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18977535

ABSTRACT

Mx, an interferon-inducible protein, is found in various vertebrates and confers resistance to several RNA viruses. At least two Mx proteins occur in vertebrates, and these proteins are key components of innate defense against viral infection. In mice and humans, the two Mx genes have different antiviral activities. Both Mx1 and Mx2 have also been detected in pigs, although only a partial sequence of porcine Mx2 has been reported, and there is no information on its antiviral activity. Here, we report the structure of the intact porcine Mx2 gene having an open reading frame of 2136 bp. We also determined the sequence of the genomic region containing the entire porcine Mx2 gene in addition to Mx1 gene. A weak constitutive expression of porcine Mx2 mRNA and endogenous Mx2 protein was observed in interferon-untreated cells. Porcine endogenous Mx2 protein showed nuclear localization. Furthermore, assays using NIH3T3 cells transfected with Mx genes showed that porcine Mx2 possessed antiviral activity against influenza, although this activity was lower than that of human MxA. This report is the first to describe the intact porcine Mx2 gene, which is a functional gene that may play a key role in the clearance of viruses in pigs.


Subject(s)
GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Swine/genetics , Swine/immunology , Animals , Cloning, Molecular , Dogs , GTP-Binding Proteins/biosynthesis , Gene Expression Regulation/immunology , Influenza A virus/genetics , Influenza A virus/metabolism , Mice , Myxovirus Resistance Proteins , NIH 3T3 Cells , Organ Specificity/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/veterinary , Swine Diseases/genetics , Swine Diseases/immunology , Swine Diseases/metabolism
18.
Rev Med Virol ; 18(4): 247-60, 2008.
Article in English | MEDLINE | ID: mdl-18383427

ABSTRACT

For replication and transcription of the influenza virus genome of eight-segmented and negative-stranded RNAs, not only viral factors but also host-derived cellular factors (host factors) are required. This paper focuses on the identification and characterisation of the host factors involved in replication and transcription of the influenza virus genome, reviewing recent progresses in the related molecular mechanisms. Functional assay systems for screening of host factors using cell-free reconstitution systems and an yeast-based influenza virus replicon system are highlighted. We have summarised the property of host factors comprehensively and provided a clue for the perspective in the determination mechanism of host range and virulence and the development of a new strategy to control the influenza virus.


Subject(s)
Host-Parasite Interactions , Orthomyxoviridae/physiology , Transcription, Genetic , Virus Replication , Animals , Humans , Saccharomyces
20.
Proc Natl Acad Sci U S A ; 104(46): 18235-40, 2007 Nov 13.
Article in English | MEDLINE | ID: mdl-17991777

ABSTRACT

Influenza viruses infect vertebrates, including mammals and birds. Influenza virus reverse-genetics systems facilitate the study of the structure and function of viral factors. In contrast, less is known about host factors involved in the replication process. Here, we developed a replication and transcription system of the negative-strand RNA genome of the influenza virus in Saccharomyces cerevisiae, which depends on viral RNAs, viral RNA polymerases, and nucleoprotein (NP). Disruption of SUB2 encoding an orthologue of human RAF-2p48/UAP56, a previously identified viral RNA synthesis stimulatory host factor, resulted in reduction of the viral RNA synthesis rate. Using a genome-wide set of yeast single-gene deletion strains, we found several host factor candidates affecting viral RNA synthesis. We found that among them, Tat-SF1, a mammalian homologue of yeast CUS2, was a stimulatory host factor in influenza virus RNA synthesis. Tat-SF1 interacted with free NP, but not with NP associated with RNA, and facilitated formation of RNA-NP complexes. These results suggest that Tat-SF1 may function as a molecular chaperone for NP, as does RAF-2p48/UAP56. This system has proven useful for further studies on the mechanism of influenza virus genome replication and transcription.


Subject(s)
Orthomyxoviridae/genetics , RNA, Viral/biosynthesis , Replicon , Saccharomyces cerevisiae/genetics , Trans-Activators/genetics , Genome, Viral , Humans , Orthomyxoviridae/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...