Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Virology ; 573: 1-11, 2022 08.
Article in English | MEDLINE | ID: mdl-35679629

ABSTRACT

Subacute sclerosing panencephalitis (SSPE) is a rare progressive neurodegenerative disease caused by measles virus variants (SSPE viruses) that results in eventual death. Amino acid substitution(s) in the viral fusion (F) protein are key for viral propagation in the brain in a cell-to-cell manner, a specific trait of SSPE viruses, leading to neuropathogenicity. In this study, we passaged an SSPE virus in cultured human neuronal cells and isolated an adapted virus that propagated more efficiently in neuronal cells and exhibited increased cell-to-cell fusion. Contrary to our expectation, the virus harbored mutations in the large protein, a viral RNA-dependent RNA polymerase, and in the phosphoprotein, its co-factor, rather than in the F protein. Our results imply that upregulated RNA polymerase activity, which increases F protein expression and cell-to-cell fusion, could be a viral factor that provides a growth advantage and contributes to the adaptation of SSPE viruses to neuronal cells.


Subject(s)
Neurodegenerative Diseases , Subacute Sclerosing Panencephalitis , Humans , Measles virus/physiology , SSPE Virus/genetics , SSPE Virus/metabolism , Subacute Sclerosing Panencephalitis/genetics , Subacute Sclerosing Panencephalitis/metabolism , Up-Regulation , Viral Fusion Proteins/genetics , Viral Replicase Complex Proteins
2.
Virology ; 487: 141-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26524513

ABSTRACT

Measles virus (MV) is the causative agent of subacute sclerosing panencephalitis (SSPE). We previously reported that the F gene of the SSPE Osaka-2 strain is the major determinant of MV neurovirulence. Because the sites and extents of mutations differ among SSPE strains, it is necessary to determine the mutations responsible for the SSPE-specific phenotypes of individual viral strain. In this study, recombinant viruses containing the envelope-associated genes from the SSPE Osaka-1 strain were generated in the IC323 wild-type MV background. Hamsters inoculated with MV containing the H gene of the Osaka-1 strain displayed hyperactivity and seizures, but usually recovered and survived. Hamsters inoculated with MV containing the F gene of the Osaka-1 strain displayed severe neurologic signs and died. Amino acid substitutions in the heptad repeat A and C regions of the F protein, including a methionine-to-valine substitution at amino acid 94, play major roles in neurovirulence.


Subject(s)
Amino Acid Substitution/genetics , Measles virus/genetics , Measles virus/pathogenicity , Subacute Sclerosing Panencephalitis/virology , Viral Fusion Proteins/genetics , Animals , Brain/virology , CHO Cells , Callithrix , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Humans , Measles virus/isolation & purification , Protein Structure, Tertiary , Vero Cells
3.
PLoS One ; 10(5): e0126813, 2015.
Article in English | MEDLINE | ID: mdl-25970481

ABSTRACT

Mycobacterium smegmatis has been widely used as a mycobacterial infection model. Unlike the M. smegmatis mc(2)155 strain, M. smegmatis J15cs strain has the advantage of surviving for one week in murine macrophages. In our previous report, we clarified that the J15cs strain has deleted apolar glycopeptidolipids (GPLs) in the cell wall, which may affect its morphology and survival in host cells. In this study, the gene causing the GPL deletion in the J15cs strain was identified. The mps1-2 gene (MSMEG_0400-0402) correlated with GPL biosynthesis. The J15cs strain had 18 bps deleted in the mps1 gene compared to that of the mc(2)155 strain. The mps1-complemented J15cs mutant restored the expression of GPLs. Although the J15cs strain produces a rough and dry colony, the colony morphology of this mps1-complement was smooth like the mc(2)155 strain. The length in the mps1-complemented J15cs mutant was shortened by the expression of GPLs. In addition, the GPL-restored J15cs mutant did not survive as long as the parent J15cs strain in the murine macrophage cell line J774.1 cells. The results are direct evidence that the deletion of GPLs in the J15cs strain affects bacterial size, morphology, and survival in host cells.


Subject(s)
Bacterial Proteins/physiology , Glycolipids/physiology , Glycopeptides/physiology , Mycobacterium smegmatis/physiology , Animals , Cell Line , Genes, Bacterial , Genetic Complementation Test , Host-Pathogen Interactions , Mice , Microbial Viability
4.
Virology ; 462-463: 91-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24967743

ABSTRACT

Measles virus (MV) is the causative agent of measles and its neurological complications, subacute sclerosing panencephalitis (SSPE) and measles inclusion body encephalitis (MIBE). Biased hypermutation in the M gene is a characteristic feature of SSPE and MIBE. To determine whether the M gene is the preferred target of hypermutation, an additional transcriptional unit containing a humanized Renilla reniformis green fluorescent protein (hrGFP) gene was introduced into the IC323 MV genome, and nude mice were inoculated intracerebrally with the virus. Biased hypermutation occurred in the M gene and also in the hrGFP gene when it was inserted between the leader and the N gene, but not between the H and L gene. These results indicate that biased hypermutation is usually found in a gene whose function is not essential for viral proliferation in the brain and that the location of a gene in the MV genome can affect its mutational frequency.


Subject(s)
Brain/virology , Encephalitis, Viral/virology , Genes, Reporter , Measles virus/genetics , Measles virus/isolation & purification , Measles/virology , Mutation , Animals , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Measles virus/classification , Mice , Mice, Nude , Mutant Proteins/genetics
5.
Microbiol Immunol ; 55(7): 518-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21443615

ABSTRACT

The Schwarz FF-8 (FF-8) and AIK-C measles virus vaccine strains are currently used for vaccination in Japan. Here, the complete genome nucleotide sequence of the FF-8 strain has been determined and its genome sequence found to be remarkably similar to that of the AIK-C strain. These two strains are differentiated only by two nucleotide differences in the phosphoprotein gene. Since the FF-8 strain does not possess the amino acid substitutions in the phospho- and fusion proteins which are responsible for the temperature-sensitivity and small syncytium formation phenotypes of the AIK-C strain, respectively, other unidentified common mechanisms likely attenuate both the FF-8 and AIK-C strains.


Subject(s)
Genome, Viral , Measles Vaccine/genetics , Measles virus/genetics , Phosphoproteins/genetics , Polymorphism, Genetic , Viral Proteins/genetics , Amino Acid Substitution , Humans , Japan , Molecular Sequence Data , Mutation, Missense , Point Mutation , Sequence Analysis, DNA , Vaccines, Attenuated
6.
J Virol ; 84(21): 11189-99, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719945

ABSTRACT

Measles virus (MV) is the causative agent for acute measles and subacute sclerosing panencephalitis (SSPE). Although numerous mutations have been found in the MV genome of SSPE strains, the mutations responsible for the neurovirulence have not been determined. We previously reported that the SSPE Osaka-2 strain but not the wild-type strains of MV induced acute encephalopathy when they were inoculated intracerebrally into 3-week-old hamsters. The recombinant MV system was adapted for the current study to identify the gene(s) responsible for neurovirulence in our hamster model. Recombinant viruses that contained envelope-associated genes from the Osaka-2 strain were generated on the IC323 wild-type MV background. The recombinant virus containing the M gene alone did not induce neurological disease, whereas the H gene partially contributed to neurovirulence. In sharp contrast, the recombinant virus containing the F gene alone induced lethal encephalopathy. This phenotype was related to the ability of the F protein to induce syncytium formation in Vero cells. Further study indicated that a single T461I substitution in the F protein was sufficient to transform the nonneuropathogenic wild-type MV into a lethal virus for hamsters.


Subject(s)
Measles virus/genetics , Subacute Sclerosing Panencephalitis/virology , Viral Fusion Proteins/genetics , Animals , Chlorocebus aethiops , Cricetinae , Measles virus/pathogenicity , Species Specificity , Vero Cells , Viral Fusion Proteins/physiology , Virulence/genetics
7.
J Virol ; 83(22): 11645-54, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726516

ABSTRACT

Attenuated live vaccines of measles virus (MV) have been developed from clinical isolates by serial propagation in heterologous cells, mainly chicken embryonic cells. The safety and effectiveness of these vaccines have been well established. However, the molecular mechanism of their attenuation remains a subject of investigation. The CAM-70 MV vaccine strain was developed from the Tanabe strain by serial propagation in chicken embryonic cells. In the present study, we assessed the contribution of each gene in the CAM-70 strain to efficient growth in chicken embryonic fibroblasts (CEF). We used a cloned MV IC323 based on the wild-type IC-B strain and generated a series of IC323s that possess one or more of the CAM-70 genes. Then, we examined the infection of CEF and CEF expressing human signaling lymphocyte activation molecule with the recombinant MVs. Our results demonstrated that MV needs to adapt to CEF at both the entry and postentry steps and that the CAM-70 matrix protein gene plays an important role in adaptation to CEF at the early stage of the virus replication cycle. The CAM-70 large protein gene was responsible for the efficient transcription and replication in CEF, and the CAM-70 hemagglutinin and fusion protein genes were responsible for efficient entry. Investigations focusing on these genes might elucidate unknown molecular mechanisms underlying the attenuation of MV.


Subject(s)
Hemagglutinins, Viral/genetics , Measles Vaccine/genetics , Measles virus/genetics , Animals , Blotting, Western , Cell Line , Chick Embryo/virology , Cloning, Molecular , DNA, Recombinant/genetics , Fibroblasts/virology , Flow Cytometry , Genes, Viral/physiology , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/genetics , Transduction, Genetic , Viral Matrix Proteins/genetics , Viral Proteins/genetics , Virus Internalization , Virus Replication/genetics
8.
Vaccine ; 27(29): 3838-48, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19490984

ABSTRACT

The CAM-70 measles virus (MV) vaccine strain is currently used for vaccination against measles. We examined the fusion-inducing ability of the CAM-70 hemagglutinin (H) protein and found that it was impaired in both CD46- and signaling lymphocyte activation molecule (SLAM)-expressing cells. We also generated recombinant MVs possessing H genes derived from the CAM-70 strain. The CAM-70 H protein impaired viral growth in both CD46- and SLAM-expressing cells. In peripheral blood lymphocytes (PBL) and monocyte-derived dendritic cells (Mo-DC), the CAM-70 strain did not grow efficiently. Infection with recombinant MVs revealed that impaired growth of the CAM-70 strain was attributed to the H gene only partly in PBL and largely in Mo-DC. Thus, impaired fusion-inducing ability of the H protein may be one of the underlying molecular mechanisms resulting in the attenuation of the CAM-70 strain.


Subject(s)
Antigens, CD/metabolism , Hemagglutinins, Viral/metabolism , Measles virus/physiology , Membrane Cofactor Protein/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Virus Attachment , Amino Acid Substitution/genetics , Animals , Cells, Cultured , Chlorocebus aethiops , Dendritic Cells/virology , HeLa Cells , Hemagglutinins, Viral/genetics , Humans , Lymphocytes/virology , Measles virus/genetics , Molecular Sequence Data , Protein Binding , Sequence Analysis, DNA , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells
9.
Int Immunol ; 20(9): 1169-80, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18611945

ABSTRACT

Monocyte-derived dendritic cells (mDCs) recognize viral RNA extrinsically by Toll-like receptor (TLR) 3 on the membrane and intrinsically retinoic acid-inducible gene I (RIG-I)/melanoma differentiation-associated gene 5 (MDA5) in the cytoplasm to induce type I IFNs and mDC maturation. When mDCs were treated with live or UV-irradiated respiratory syncytial virus (RSV), early ( approximately 4 h) induction of IFN-beta usually occurs in other virus infections was barely observed. Live RSV subsequently replicated to activate the cytoplasmic IFN-inducing pathway leading to robust type I IFN induction. We found that RSV initial attachment to cells blocked polyI:C-mediated IFN-beta induction, and this early IFN-beta-modulating event was abrogated by antibodies against envelope proteins of RSV, demonstrating the presence of a IFN-regulatory mode by early RSV attachment to host cells. By IFN-stimulated response element (ISRE) reporter analysis in HEK293 cells, polyI:C- or LPS-mediated ISRE activation was dose dependently inhibited by live and inactive RSV to a similar extent. Of the RSV envelope proteins, simultaneously expressed or exogenously added RSV G or soluble G (sG) proteins inhibited TLR3/4-mediated ISRE activation in HEK293 cells. sG proteins expressed in cells did not affect the RIG-I/MDA5 pathway but inhibited the TLR adaptor TRIF/TICAM-1 pathway for ISRE activation. Finally, extrinsically added sG protein suppressed the production of IFN-beta in mDCs. Although the molecular mechanism of this extrinsic functional mode of the RSV G glycoprotein (G protein) remains undetermined, G proteins may neutralize the fusion glycoprotein function that promotes IFN-mediated mDC modulation via TLR4 and may cause insufficient raising cell-mediated immunity against RSV.


Subject(s)
Interferon-beta/metabolism , Respiratory Syncytial Virus, Human/pathogenicity , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Viral Envelope Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Dendritic Cells/immunology , Gene Expression Regulation , Humans , Interferon-beta/genetics , Respiratory Syncytial Virus, Human/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Viral Envelope Proteins/immunology
10.
Virus Res ; 130(1-2): 260-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17825451

ABSTRACT

Measles virus (MV) is the causative agent of subacute sclerosing panencephalitis (SSPE) and viruses isolated from brains of the patients contain numerous mutations. We have previously demonstrated that the hemagglutinin (H) protein of MV SSPE strains can interact with the signaling lymphocyte activation molecule (SLAM) and an unidentified molecule on Vero cells, but not with CD46, as a receptor. The mechanism by which MV SSPE strains can induce cell-cell fusion in SLAM-negative Vero cells is not understood. We report here on the effect of mutations in the fusion (F) proteins of three MV SSPE strains on syncytium formation. The F proteins of the three SSPE strains were functional and co-expression with H protein from the MV wild-type or SSPE strains in this study induced formation of large syncytia in Vero cells as well as in cell lines expressing SLAM or CD46. Expression of chimeric F proteins of SSPE strains showed that amino acid substitutions in the F protein extracellular as well as cytoplasmic domain contributed to enhanced cell-cell fusion in Vero cells. These findings suggest a common molecular mechanism and a key role of the F protein for syncytium formation in cells expressing an unidentified third receptor for MV.


Subject(s)
Giant Cells/virology , Measles virus/genetics , Measles virus/pathogenicity , Subacute Sclerosing Panencephalitis/virology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Amino Acid Substitution/genetics , Animals , Chlorocebus aethiops , Humans , Measles virus/isolation & purification , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Vero Cells
11.
J Med Virol ; 79(8): 1187-93, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17597489

ABSTRACT

In 2 infants with gastroenteritis associated with Norovirus (NoV), serum immunoglobulin (Ig) G, IgM, IgA, and fecal IgA antibody responses against NoV were examined by enzyme-linked immunosorbent assay using 11 different antigenic and genetic types of NoV virus-like particles expressed in insect cells. These two cases were putative primary single NoV infections, because antibodies against NoVs were not detected in acute-phase serums. In one of two cases, long-term excretion of virus RNA for 33 days was observed. Serum IgG responses demonstrated strong seroresponse to the homologous type, and weak seroresponse to the heterologous types within the genogroup. After more than 2 years, the IgG antibody titer remained high to the homologous type and low to the heterologous type within the genogroup. IgM and IgA were specific to the homologous type. IgM was short lived and the serum IgA antibody titer remained low to the homologous type for a long period. These results improve our understanding of the humoral immune response to NoV infection.


Subject(s)
Antibodies, Viral/isolation & purification , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Gastroenteritis/immunology , Norovirus/isolation & purification , Antibodies, Viral/blood , Antibody Formation , Caliciviridae Infections/blood , Child, Preschool , Feces/virology , Gastroenteritis/virology , Humans , Immunoglobulin A/isolation & purification , Immunoglobulin M/isolation & purification , Infant , Male , Norovirus/genetics , Phylogeny
12.
Virus Res ; 123(1): 1-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16959355

ABSTRACT

Macrophages (Mø) and dendritic cells (DC) are thought to be targets of measles virus (MeV) at the early stage of infection. We compared the growth of Edmonston-derived vaccine strains and fresh clinical isolates of MeV in monocytes, monocyte-derived granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced Mø (GM-Mø) and in monocyte-derived DC (Mo-DC). Neither vaccine strains nor fresh isolates thrived in monocytes and GM-Mø and no differences were evident among them. On the other hand, infectious virus production was robust in Mo-DC infected with fresh isolates, but below the limits of detection in those infected with vaccine strains. Although the vaccine strains infected Mo-DC and replicated comparably with the fresh isolates, they accumulated far less matrix (M) protein. This was attributed to a difference in the stability of M protein produced in Mo-DC between the strains. Impaired production of infectious viruses in DC may be one cause of vaccine strain attenuation.


Subject(s)
Dendritic Cells/virology , Measles/virology , Monocytes/virology , Morbillivirus/growth & development , Animals , Cells, Cultured , Child , Child, Preschool , Dendritic Cells/metabolism , Humans , Monocytes/metabolism , Viral Matrix Proteins/metabolism
13.
J Immunol ; 175(5): 3252-61, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16116216

ABSTRACT

We generated transgenic (TG) mice that constitutively express human CD46 (huCD46) and/or TLR-inducible CD150 (huCD150), which serve as receptors for measles virus (MV). These mice were used to study the spreading and pathogenicity of GFP-expressing or intact laboratory-adapted Edmonston and wild-type Ichinose (IC) strains of MV. Irrespective of the route of administration, neither type of MV was pathogenic to these TG mice. However, in ex vivo, limited replication of IC was observed in the spleen lymphocytes from huCD46/huCD150 TG and huCD150 TG, but not in huCD46 TG and non-TG mice. In huCD150-positive TG mouse cells, CD11c-positive bone marrow-derived myeloid dendritic cells (mDC) participated in MV-mediated type I IFN induction. The level and induction profile of IFN-beta was higher in mDC than the profile of IFN-alpha. Wild-type IC induced markedly high levels of IFN-beta compared with Edmonston in mDC, as opposed to human dendritic cells. We then generated huCD46/huCD150 TG mice with type I IFN receptor (IFNAR1)-/- mice. MV-bearing mDCs spreading to draining lymph nodes were clearly observed in these triple mutant mice in vivo by i.p. MV injection. Infectious lymph nodes were also detected in the double TG mice into which MV-infected CD11c-positive mDCs were i.v. transferred. This finding suggests that in the double TG mouse model mDCs once infected facilitate systemic MV spreading and infection, which depend on mDC MV permissiveness determined by the level of type I IFN generated via IFNAR1. Although these results may not simply reflect human MV infection, the huCD150/huCD46 TG mice may serve as a useful model for the analysis of MV-dependent modulation of mDC response.


Subject(s)
Antigens, CD/physiology , CD11c Antigen/analysis , Dendritic Cells/physiology , Glycoproteins/physiology , Immunoglobulins/physiology , Measles/immunology , Membrane Glycoproteins/physiology , Animals , Chlorocebus aethiops , Dendritic Cells/virology , Humans , Interferon Type I/physiology , Membrane Cofactor Protein , Mice , Mice, Transgenic , Receptors, Cell Surface , Receptors, Interferon/physiology , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells , Virus Replication
14.
Microbiol Immunol ; 49(3): 275-83, 2005.
Article in English | MEDLINE | ID: mdl-15782001

ABSTRACT

Noroviruses (NVs) are the major cause of food- and waterborne nonbacterial gastroenteritis in Japan. Between April 2002 and March 2003, a total of 111 fecal specimens from 40 outbreaks of acute nonbacterial gastroenteritis in Osaka City, Japan were subject to NV detection. Seventy-two samples (64.9%) from 31 outbreaks (77.5%) were NV positive by a real time reverse transcription (RT)-PCR assay. To further determine the genotype of individual NV strains, we sequenced the capsid N-terminal/shell (N/S) domain of some representative strains from each outbreak. The 51 NV strains detected in this study were segregated into 15 genotypes (6 in genogroup I and 9 in genogroup II), and GII/5 genotype NV was a dominant outbreak genotype.


Subject(s)
Caliciviridae Infections/epidemiology , Caliciviridae Infections/virology , Disease Outbreaks , Gastroenteritis/epidemiology , Gastroenteritis/virology , Norovirus/classification , Norovirus/genetics , Capsid Proteins/chemistry , Capsid Proteins/genetics , Genotype , Humans , Japan/epidemiology , Molecular Sequence Data , Norovirus/isolation & purification , Phylogeny , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction
16.
Microbiol Immunol ; 48(4): 277-87, 2004.
Article in English | MEDLINE | ID: mdl-15107538

ABSTRACT

Measles virus is the causative agent of subacute sclerosing panencephalitis (SSPE). The viruses isolated from brain cells of patients with SSPE (called SSPE viruses) are defective in cell-free virus production in vitro. To investigate the cell tropism of three strains of SSPE virus (Osaka-1, Osaka-2, Osaka-3), SSPE virus-infected cell cultures were treated with cytochalasin D to prepare virus-like particles (CD-VLPs). All CD-VLPs formed syncytia after infection in CHO cells expressing CD150 but not in those expressing CD46. In addition, an antibody to CD46 did not block the infection of Vero cells by SSPE CDVLPs. The results were consistent with our previous suggestion that one or more unidentified receptors might be involved in the entry process. Infection with the CD-VLPs from three SSPE strains was further examined in different human cell lines, including those of neural origin, and was found to induce syncytia in epithelial cells (HeLa and 293T) as well as neuroblastoma cells (IMR-32 and SK-N-SH) with varying efficiency. SSPE CD-VLPs also infected glioblastoma cells (A172) and astrocytoma cells (U-251) but syncytial formation was rarely induced. These epithelial and neural cell lines were not permissive for the replication of wild-type MV. Together with our previous observations, these results suggest that the cell entry receptor is the major factor determining the cell tropism of SSPE viruses. Further studies are necessary to identify other viral and/or cellular factors that might be involved in the replication of SSPE virus in specific neural cells and in the brain.


Subject(s)
Nervous System/virology , SSPE Virus/pathogenicity , Animals , Antigens, CD , CHO Cells , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Giant Cells , Glycoproteins/metabolism , HeLa Cells , Humans , Immunoglobulins/metabolism , Nervous System/cytology , Receptors, Cell Surface , Signaling Lymphocytic Activation Molecule Family Member 1 , Subacute Sclerosing Panencephalitis/virology , Vero Cells , Virion/pathogenicity
17.
Biochem Biophys Res Commun ; 311(1): 39-48, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-14575692

ABSTRACT

PolyI:C, a synthetic double-stranded (ds)RNA, and viruses act on cells to induce IFN-beta which is a key molecule for anti-viral response. Although dsRNA is a virus-specific signature and a ligand for human Toll-like receptor 3 (TLR3), largely uncharacterized multiple pathways associate virus-mediated IFN-beta induction. Here, we demonstrated that laboratory-adapted but not wild-type strains of measles virus (MV) up-regulated TLR3 expression both in dendritic cells and epithelial cell line A549. The kinetics experiments with the laboratory MV strain revealed that TLR3 was induced late compared to IFN-beta and required new protein synthesis. Furthermore, neutralizing antibodies against IFN-beta or IFNAR (Interferon-alpha/beta receptor) suppressed MV-induced TLR3 induction, indicating that type I IFN, IFN-alpha/beta, is critical for MV-mediated TLR3 induction. Yet, a recently identified virus-inducible IFN, the IFN-lambda, did not contribute to TLR3 expression. A virus-responsive element that up-regulates TLR3 was identified in the TLR3-promoter region by reporter gene experiments. The ISRE, a recently reported site for IFN-beta induction, but not STAT binding site, located around -30bp of TLR3 promoter responded to MV to induce TLR3 expression. This further indicates the importance of type I IFN for TLR3 up-regulation in the case of viral infection. In HeLa and MRC5 cells, augmented production of IFN-beta was observed in response to dsRNA when TLR3 had been induced beforehand. Thus, the MV-induced expression of TLR3 may reflect amplified IFN production that plays a part in host defense to viral infection.


Subject(s)
Interferon-beta/immunology , Interferon-beta/metabolism , Measles virus/pathogenicity , Measles/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Up-Regulation/physiology , Cell Line , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Measles/immunology , Measles virus/classification , Measles virus/immunology , Membrane Glycoproteins/classification , Membrane Glycoproteins/immunology , Receptors, Cell Surface/classification , Receptors, Cell Surface/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Toll-Like Receptor 3 , Toll-Like Receptors , Vaccines, Attenuated
19.
J Gen Virol ; 84(Pt 8): 2133-2143, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12867645

ABSTRACT

The vaccine or Vero cell-adapted strains of measles virus (MV) have been reported to use CD46 as a cell entry receptor, while lymphotropic MVs preferentially use the signalling lymphocyte activation molecule (SLAM or CD150). In contrast to the virus obtained from patients with acute measles, little is known about the receptor that is used by defective variants of MV isolated from patients with subacute sclerosing panencephalitis (SSPE). The receptor-binding properties of SSPE strains of MV were analysed using vesicular stomatitis virus pseudotypes expressing the envelope glycoproteins of SSPE strains of MV. Such pseudotype viruses could use SLAM but not CD46 for entry. The pseudotype viruses with SSPE envelope glycoproteins could enter Vero cells, which do not express SLAM. In addition, their entry was not blocked by the monoclonal antibody to CD46, pointing to another entry receptor for SSPE strains on Vero cells. Furthermore, the unknown receptor(s), distinct from SLAM and CD46, may be present on cell lines derived from lymphoid and neural cells. Biochemical characterization of the receptor present on Vero cells and SK-N-SH neuroblastoma cells was consistent with a glycoprotein. Identification of additional entry receptors for MV will provide new insights into the mechanism of spread of MV in the central nervous system and possible reasons for differences between MVs isolated from patients with acute measles and SSPE.


Subject(s)
Glycoproteins/metabolism , Measles virus/metabolism , Receptors, Virus/metabolism , Subacute Sclerosing Panencephalitis/virology , Vesicular stomatitis Indiana virus/metabolism , Animals , Antigens, CD/metabolism , Brain/virology , Cell Line , Cricetinae , Defective Viruses/genetics , Defective Viruses/metabolism , Defective Viruses/pathogenicity , Genetic Variation , Glycoproteins/genetics , Humans , Immunoglobulins/metabolism , Measles virus/genetics , Measles virus/pathogenicity , Membrane Cofactor Protein , Membrane Glycoproteins/metabolism , Mice , Receptors, Cell Surface , Signaling Lymphocytic Activation Molecule Family Member 1 , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/pathogenicity
20.
J Clin Microbiol ; 41(4): 1756-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12682179

ABSTRACT

Surveillance of Norwalk-like virus (NLV) infections in cases of pediatric gastroenteritis between April 1996 and March 2000 showed that NLVs were an important causative agent in viral gastroenteritis cases among children between November and January in those years. The predominant type of NLV was closely related to Lordsdale virus in genogroup 2. During the 1999-2000 season, Arg320-like strains, which may be genetic recombinants, suddenly appeared and spread.


Subject(s)
Caliciviridae Infections/epidemiology , Disease Outbreaks , Gastroenteritis/epidemiology , Norovirus/classification , Population Surveillance , Caliciviridae Infections/virology , Child , Child, Preschool , Gastroenteritis/virology , Humans , Infant , Infant, Newborn , Japan/epidemiology , Molecular Sequence Data , Norovirus/genetics , Norovirus/isolation & purification , Phylogeny , Prevalence , Reverse Transcriptase Polymerase Chain Reaction , Seasons , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...