Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 17(10): e1009841, 2021 10.
Article in English | MEDLINE | ID: mdl-34648591

ABSTRACT

In general, in mammalian cells, cytosolic DNA viruses are sensed by cyclic GMP-AMP synthase (cGAS), and RNA viruses are recognized by retinoic acid-inducible gene I (RIG-I)-like receptors, triggering a series of downstream innate antiviral signaling steps in the host. We previously reported that measles virus (MeV), which possesses an RNA genome, induces rapid antiviral responses, followed by comprehensive downregulation of host gene expression in epithelial cells. Interestingly, gene ontology analysis indicated that genes encoding mitochondrial proteins are enriched among the list of downregulated genes. To evaluate mitochondrial stress after MeV infection, we first observed the mitochondrial morphology of infected cells and found that significantly elongated mitochondrial networks with a hyperfused phenotype were formed. In addition, an increased amount of mitochondrial DNA (mtDNA) in the cytosol was detected during progression of infection. Based on these results, we show that cytosolic mtDNA released from hyperfused mitochondria during MeV infection is captured by cGAS and causes consequent priming of the DNA sensing pathway in addition to canonical RNA sensing. We also ascertained the contribution of cGAS to the in vivo pathogenicity of MeV. In addition, we found that other viruses that induce downregulation of mitochondrial biogenesis as seen for MeV cause similar mitochondrial hyperfusion and cytosolic mtDNA-priming antiviral responses. These findings indicate that the mtDNA-activated cGAS pathway is critical for full innate control of certain viruses, including RNA viruses that cause mitochondrial stress.


Subject(s)
Immunity, Innate/immunology , Measles/metabolism , Mitochondria/metabolism , Nucleotidyltransferases/metabolism , Animals , Down-Regulation , Humans , Measles virus , Mice , Mice, Inbred C57BL , Mitochondria/virology , Organelle Biogenesis , RNA Virus Infections/metabolism , RNA Viruses
2.
Sci Rep ; 7(1): 12017, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28931922

ABSTRACT

Highly pathogenic avian influenza virus (HPAIV) is a serious threat not only to domestic fowls but also to humans. Vaccines inducing long-lasting immunity against HPAIV are required. In the present study, we generated recombinant measles virus (MV) expressing the hemagglutinin protein of HPAIV without the multibasic site necessary for its pathogenicity in chickens using the backbone of an MV vaccine strain (rMV-Ed-H5HA) or a wild-type MV-derived mutant (rMV-HL-Vko-H5HA). We examined protective efficacy of the candidate vaccines in the monkey infection model by the challenge with a HPAIV (H5N1). Cynomolgus monkeys inoculated with the candidate vaccines produced both anti-H5 HA and anti-MV antibodies. They recovered earlier from influenza symptoms than unvaccinated monkeys after the challenge with the HPAIV strain. Chest radiography and histopathological analyses confirmed less severe pneumonia in the vaccinated monkeys. Vaccination tended to suppress viral shedding and reduced the interleukin-6 levels in the lungs. Furthermore, the vaccination with rMV-Ed-H5HA of monkeys with pre-existing anti-MV immunity induced the production of anti-H5 HA antibodies. These results suggest that both candidate vaccines effectively reduce disease severity in naïve hosts, and that rMV-Ed-H5HA is a particularly good candidate vaccine against HPAIV infection.


Subject(s)
Influenza A Virus, H5N1 Subtype/immunology , Measles virus/immunology , Measles/immunology , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Viral/immunology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Influenza A Virus, H5N1 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Macaca fascicularis , Measles/prevention & control , Measles/virology , Measles Vaccine/administration & dosage , Measles Vaccine/immunology , Measles virus/genetics , Measles virus/physiology , Mutation , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Recombination, Genetic , Treatment Outcome , Vaccination
3.
J Gen Virol ; 98(7): 1620-1629, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28708054

ABSTRACT

In rare cases, measles virus (MV) in children leads to fatal neurological complications such as primary measles encephalitis, post-acute measles encephalitis, subacute sclerosing panencephalitis and measles inclusion-body encephalitis. To investigate the pathogenesis of MV-induced encephalitis, rodent brain-adapted MV strains CAM/RB and CAMR40 were generated. These strains acquired mutations to adapt to the rodent brain during 40 passages in rat brain. However, it is still unknown which genes confer the neurovirulence of MV. We previously established a rescue system for recombinant MVs possessing the backbone of wild-type strain HL, an avirulent strain in mice. In the present study, to identify the genes in CAMR40 that elicit neurovirulence, we generated chimeric recombinant MVs based on strain HL. As a result, recombinant wild-type MV in which the haemagglutinin (H) gene was substituted with that of CAMR40 caused a non-lethal mild disease in mice, while additional substitution of the HL phosphoprotein (P) gene with that of strain CAMR40 caused lethal severe neurological signs comparable to those of CAMR40. These results clearly indicated that, in addition to the H gene, the P gene is required for the neurovirulence of MV CAMR40.


Subject(s)
Brain/pathology , Hemagglutinins/genetics , Measles virus/genetics , Measles virus/pathogenicity , Phosphoproteins/genetics , Subacute Sclerosing Panencephalitis/pathology , Viral Proteins/genetics , Animals , Brain/virology , Callithrix , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Mice , Mice, Inbred C57BL , Subacute Sclerosing Panencephalitis/genetics , Subacute Sclerosing Panencephalitis/virology , Vero Cells
4.
J Virol ; 90(20): 9285-92, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27489268

ABSTRACT

UNLABELLED: In the current study, we generated recombinant chimeric canine distemper viruses (CDVs) by replacing the hemagglutinin (H) and/or phosphoprotein (P) gene in an avirulent strain expressing enhanced green fluorescent protein (EGFP) with those of a mouse-adapted neurovirulent strain. An in vitro experimental infection indicated that the chimeric CDVs possessing the H gene derived from the mouse-adapted CDV acquired infectivity for neural cells. These cells lack the CDV receptors that have been identified to date (SLAM and nectin-4), indicating that the H protein defines infectivity in various cell lines. The recombinant viruses were administered intracerebrally to 1-week-old mice. Fatal neurological signs of disease were observed only with a recombinant CDV that possessed both the H and P genes of the mouse-adapted strain, similar to the parental mouse-adapted strain, suggesting that both genes are important to drive virulence of CDV in mice. Using this recombinant CDV, we traced the intracerebral propagation of CDV by detecting EGFP. Widespread infection was observed in the cerebral hemispheres and brainstems of the infected mice. In addition, EGFP fluorescence in the brain slices demonstrated a sequential infectious progression in the central nervous system: CDV primarily infected the neuroependymal cells lining the ventricular wall and the neurons of the hippocampus and cortex adjacent to the ventricle, and it then progressed to an extensive infection of the brain surface, followed by the parenchyma and cortex. In the hippocampal formation, CDV spread in a unidirectional retrograde pattern along neuronal processes in the hippocampal formation from the CA1 region to the CA3 region and the dentate gyrus. Our mouse model demonstrated that the main target cells of CDV are neurons in the acute phase and that the virus spreads via neuronal transmission pathways in the hippocampal formation. IMPORTANCE: CDV is the etiological agent of distemper in dogs and other carnivores, and in many respects, the pathogenesis of CDV infection in animals resembles that of measles virus infection in humans. We successfully generated a recombinant CDV containing the H and P genes from a mouse-adapted neurovirulent strain and expressing EGFP. The recombinant CDV exhibited severe neurovirulence with high mortality, comparable to the parental mouse-adapted strain. The mouse-infectious model could become a useful tool for analyzing CDV infection of the central nervous system subsequent to passing through the blood-cerebrospinal fluid barrier and infectious progression in the target cells in acute disease.


Subject(s)
Cerebrospinal Fluid/virology , Distemper Virus, Canine/pathogenicity , Distemper/virology , Hippocampus/virology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Cell Line , Cell Line, Tumor , Cerebrospinal Fluid/metabolism , Chlorocebus aethiops , Distemper/metabolism , Dogs , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hippocampus/metabolism , Humans , Measles/metabolism , Measles/virology , Measles virus/pathogenicity , Mice , Neurons/metabolism , Neurons/virology , Receptors, Virus/metabolism , Vero Cells
5.
Mol Ther Oncolytics ; 3: 15022, 2016.
Article in English | MEDLINE | ID: mdl-27119113

ABSTRACT

Oncolytic virotherapy is a promising treatment strategy for cancer. We previously generated a recombinant measles virus (rMV-SLAMblind) that selectively uses a poliovirus receptor-related 4 (PVRL4/Nectin4) receptor, but not signaling lymphocyte activation molecule (SLAM). We demonstrated that the virus exerts therapeutic effects against human breast cancer cells. Here, we examined the applicability of rMV-SLAMblind to treating canine mammary cancers (CMCs). We found that the susceptibilities of host cells to rMV-SLAMblind were dependent on canine Nectin-4 expression. Nectin-4 was detected in four of nine CMC cell lines. The rMV-SLAMblind efficiently infected those four Nectin-4-positive cell lines and was cytotoxic for three of them (CF33, CHMm, and CTBm). In vivo experiment showed that the administration of rMV-SLAMblind greatly suppressed the progression of tumors in mice xenografted with a CMC cell line (CF33). Immunohistochemistry revealed that canine Nectin-4 was expressed in 45% of canine mammary tumors, and the tumor cells derived from one clinical specimen were efficiently infected with rMV-SLAMblind. These results suggest that rMV-SLAMblind infects CMC cells and displays antitumor activity in vitro, in xenografts, and ex vivo. Therefore, oncolytic virotherapy with rMV-SLAMblind can be a novel method for treating CMCs.

6.
Oncotarget ; 6(28): 24895-903, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26317644

ABSTRACT

Lung cancer cells, particularly those of non-small-cell lung cancer, are known to express Nectin-4. We previously generated a recombinant measles virus that uses Nectin-4 as its receptor but cannot bind its original principal receptor, signaling lymphocyte activation molecule (SLAM). This virus (rMV-SLAMblind) infects and kills breast cancer cells in vitro and in a subcutaneous xenograft model. However, it has yet to be determined whether rMV-SLAMblind is effective against other cancer types and in other tumor models that more closely represent disease. In this study, we analyzed the anti-tumor activity of this virus towards lung cancer cells using a modified variant that encodes green fluorescent protein (rMV-EGFP-SLAMblind). We found that rMV-EGFP-SLAMblind efficiently infected nine, human, lung cancer cell lines, and its infection resulted in reduced cell viability of six cell lines. Administration of the virus into subcutaneous tumors of xenotransplanted mice suppressed tumor growth. In addition, rMV-EGFP-SLAMblind could target scattered tumor masses grown in the lungs of xenotransplanted mice. These results suggest that rMV-SLAMblind is oncolytic for lung cancer and that it represents a promising tool for the treatment of this disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Measles virus/physiology , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/virology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/virology , Measles virus/genetics , Measles virus/metabolism , Mice, SCID , Microscopy, Fluorescence , Oncolytic Viruses/genetics , Oncolytic Viruses/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Xenograft Model Antitumor Assays
7.
J Virol ; 89(19): 9709-18, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26157124

ABSTRACT

UNLABELLED: Measles virus (MeV) causes several unique syndromes, including transient immunosuppression. To clarify the cellular responses to MeV infection, we previously analyzed a MeV-infected epithelial cell line and a lymphoid cell line by microarray and showed that the expression of numerous genes was up- or downregulated in the epithelial cells. In particular, there was a characteristic comprehensive downregulation of housekeeping genes during late stage infection. To identify the mechanism underlying this phenomenon, we examined the phosphorylation status of transcription factors and kinase/phosphatase activities in epithelial cells after infection. MeV infection inactivated cellular protein phosphatase 5 (PP5) that consequently inactivated DNA-dependent protein kinase, which reduced Sp1 phosphorylation levels, and c-Myc degradation, both of which downregulated the expression of many housekeeping genes. In addition, intracellular accumulation of viral nucleocapsid inactivated PP5 and subsequent downstream responses. These findings demonstrate a novel strategy of MeV during infection, which causes the collapse of host cellular functions. IMPORTANCE: Measles virus (MeV) is one of the most important pathogens in humans. We previously showed that MeV infection induces the comprehensive downregulation of housekeeping genes in epithelial cells. By examining this phenomenon, we clarified the molecular mechanism underlying the constitutive expression of housekeeping genes in cells, which is maintained by cellular protein phosphatase 5 (PP5) and DNA-dependent protein kinase. We also demonstrated that MeV targets PP5 for downregulation in epithelial cells. This is the first report to show how MeV infection triggers a reduction in overall cellular functions of infected host cells. Our findings will help uncover unique pathogenicities caused by MeV.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation/physiology , Measles virus , Measles/physiopathology , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , DNA Primers/genetics , Electrophoretic Mobility Shift Assay , HEK293 Cells , Humans , Immunoprecipitation , Luciferases , Microarray Analysis , Phosphorylation , Plasmids/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism
8.
PLoS One ; 8(3): e58414, 2013.
Article in English | MEDLINE | ID: mdl-23516477

ABSTRACT

Nipah virus (NiV) is a member of the genus Henipavirus, which emerged in Malaysia in 1998. In pigs, infection resulted in a predominantly non-lethal respiratory disease; however, infection in humans resulted in over 100 deaths. Nipah virus has continued to re-emerge in Bangladesh and India, and person-to-person transmission appeared in the outbreak. Although a number of NiV vaccine studies have been reported, there are currently no vaccines or treatments licensed for human use. In this study, we have developed a recombinant measles virus (rMV) vaccine expressing NiV envelope glycoproteins (rMV-HL-G and rMV-Ed-G). Vaccinated hamsters were completely protected against NiV challenge, while the mortality of unvaccinated control hamsters was 90%. We trialed our vaccine in a non-human primate model, African green monkeys. Upon intraperitoneal infection with NiV, monkeys showed several clinical signs of disease including severe depression, reduced ability to move and decreased food ingestion and died at 7 days post infection (dpi). Intranasal and oral inoculation induced similar clinical illness in monkeys, evident around 9 dpi, and resulted in a moribund stage around 14 dpi. Two monkeys immunized subcutaneously with rMV-Ed-G showed no clinical illness prior to euthanasia after challenge with NiV. Viral RNA was not detected in any organ samples collected from vaccinated monkeys, and no pathological changes were found upon histopathological examination. From our findings, we propose that rMV-NiV-G is an appropriate NiV vaccine candidate for use in humans.


Subject(s)
Henipavirus Infections/prevention & control , Measles virus/genetics , Nipah Virus/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Body Temperature , Body Weight , Brain/immunology , Brain/pathology , Brain/virology , Chlorocebus aethiops , Cricetinae , Gene Expression , Genetic Vectors/genetics , Henipavirus Infections/mortality , Immunization , Lung/immunology , Lung/pathology , Lung/virology , Virus Replication
9.
J Gen Virol ; 92(Pt 9): 2133-2141, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21613447

ABSTRACT

Many viruses use their host's cellular machinery to regulate the functions of viral proteins. The phosphorylation of viral proteins is known to play a role in genome transcription and replication in paramyxoviruses. The paramyxovirus nucleoprotein (N), the most abundant protein in infected cells, is a component of the N-RNA complex and supports the transcription and replication of virus mRNA and genomic RNA. Recently, we reported that the phosphorylation of measles virus N is involved in the regulation of viral RNA synthesis. In this study, we report a rapid turnover of phosphorylation in the Nipah virus N (NiV-N). The phosphorylated NiV-N was hardly detectable in steady-state cells, but was detected after inhibition of cellular protein phosphatases. We identified a phosphorylated serine residue at Ser451 of NiV-N by peptide mass fingerprinting by electrospray ionization-quadrupole time-of-flight mass spectrometry. In the NiV minigenome assay, using luciferase as a reporter gene, the substitution of Ser451 for alanine in NiV-N resulted in a reduction in luciferase activity of approximately 45 % compared with the wild-type protein. Furthermore, the substitution of Ser451 for glutamic acid, which mimics a phosphoserine, led to a more significant decrease in luciferase activity - approximately 81 %. Northern blot analysis showed that both virus transcription and replication were reduced by these mutations. These results suggest that a rapid turnover of the phosphorylation of NiV-N plays an important role in virus transcription and replication.


Subject(s)
Nipah Virus/physiology , Nucleoproteins/metabolism , Transcription, Genetic , Viral Proteins/metabolism , Virus Replication , Animals , Cell Line , Chlorocebus aethiops , Humans , Phosphorylation , RNA, Viral/metabolism , Serine/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
J Virol ; 85(5): 2247-53, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21159870

ABSTRACT

Measles is a highly contagious human disease caused by the measles virus (MeV). In this study, by proteomic analysis, we identified peroxiredoxin 1 (Prdx1) as a host factor that binds to the C-terminal region of the nucleoprotein (N; N(TAIL)) of MeV. Glutathione S-transferase (GST) pulldown experiments showed that the Prdx1-binding site overlapped with the MeV phosphoprotein (P)-binding site on N(TAIL) and that Prdx1 competed for the binding to N(TAIL) with the P protein, which is a component of RNA-dependent RNA polymerase (RdRp). Furthermore, RNA interference for Prdx1 resulted in a significant reduction in MeV growth in HEK293-SLAM cells. A minigenome assay indicated that Prdx1 suppression affected the viral RNA transcription and/or replication step. Relative quantification of viral RNA by real-time PCR (RT-PCR) showed that Prdx1 suppression not only reduced viral RNA transcription and replication but also enhanced polar attenuation in viral mRNA transcription. Surface plasmon resonance analysis showed that the binding affinity of Prdx1 to MeV-N was 40-fold lower than that of MeV-P to MeV-N, which suggested that Prdx1 might be involved in the early stage of MeV infection, when the expression level of Prdx1 was much higher than that of MeV-P. Since Prdx1 was expressed abundantly and constitutively in various cells, the results in this study indicate that Prdx1 is one of the inherent host factors implicated in MeV RNA synthesis.


Subject(s)
Measles virus/physiology , Measles/enzymology , Peroxiredoxins/metabolism , Transcription, Genetic , Virus Replication , Binding Sites , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Measles/virology , Measles virus/chemistry , Measles virus/genetics , Nucleocapsid Proteins , Nucleoproteins/chemistry , Nucleoproteins/genetics , Nucleoproteins/metabolism , Peroxiredoxins/chemistry , Peroxiredoxins/genetics , Protein Binding , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
11.
PLoS One ; 5(9): e12709, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20856799

ABSTRACT

Nipah virus (NiV) P gene encodes P protein and three accessory proteins (V, C and W). It has been reported that all four P gene products have IFN antagonist activity when the proteins were transiently expressed. However, the role of those accessory proteins in natural infection with NiV remains unknown. We generated recombinant NiVs lacking V, C or W protein, rNiV(V-), rNiV(C-), and rNiV(W-), respectively, to analyze the functions of these proteins in infected cells and the implications in in vivo pathogenicity. All the recombinants grew well in cell culture, although the maximum titers of rNiV(V-) and rNiV(C-) were lower than the other recombinants. The rNiV(V-), rNiV(C-) and rNiV(W-) suppressed the IFN response as well as the parental rNiV, thereby indicating that the lack of each accessory protein does not significantly affect the inhibition of IFN signaling in infected cells. In experimentally infected golden hamsters, rNiV(V-) and rNiV(C-) but not the rNiV(W-) virus showed a significant reduction in virulence. These results suggest that V and C proteins play key roles in NiV pathogenicity, and the roles are independent of their IFN-antagonist activity. This is the first report that identifies the molecular determinants of NiV in pathogenicity in vivo.


Subject(s)
Henipavirus Infections/virology , Nipah Virus/metabolism , Nipah Virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Humans , Mesocricetus , Nipah Virus/genetics , Vero Cells , Viral Nonstructural Proteins/genetics
12.
J Virol ; 84(19): 9793-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20668089

ABSTRACT

The interaction of Nipah virus (NiV) nucleocapsid (N) protein with phosphoprotein (P) during nucleocapsid assembly is the essential process in the viral life cycle, since only the encapsidated RNA genome can be used for replication. To identify the region responsible for N-P interaction, we utilized fluorescent protein tags to visualize NiV N and P proteins in live cells and analyzed their cellular localization. N protein fused to monomeric enhanced cyan fluorescence protein (N-ECFP) exhibited a dotted pattern in transfected cells, while P protein fused to monomeric red fluorescent protein (P-mRFP) showed diffuse distribution. When the two proteins were coexpressed, P-mRFP colocalized with N-ECFP dots. N-ECFP mutants with serial amino acid deletions were generated to search for the region(s) responsible for this N-P colocalization. We found that, in addition to the 467- to 496-amino-acid (aa) region reported previously, aa 135 to 146 were responsible for the N-P colocalization. The residues crucial for N-P interaction were further investigated by introducing alanine substitutions into the untagged N protein. Alanine scanning in the region of aa 135 to 146 has revealed that there are distinct regions essential for the interaction of N-P and the function of N. This is the first study to visualize Nipah viral proteins in live cells and to assess the essential domain of N protein for the interaction with P protein.


Subject(s)
Nipah Virus/physiology , Nucleocapsid Proteins/physiology , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nipah Virus/genetics , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Phosphoproteins/physiology , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Vero Cells , Virus Replication/physiology , Red Fluorescent Protein
13.
J Virol ; 84(9): 4183-93, 2010 May.
Article in English | MEDLINE | ID: mdl-20147391

ABSTRACT

Measles is a highly contagious human disease caused by measles virus (MeV) and remains the leading cause of death in children, particularly in developing countries. Wild-type MeV preferentially infects lymphocytes by using signaling lymphocytic activation molecule (SLAM), whose expression is restricted to hematopoietic cells, as a receptor. MeV also infects other epithelial and neuronal cells that do not express SLAM and causes pneumonia and diarrhea and, sometimes, serious symptoms such as measles encephalitis and subacute sclerosing panencephalitis. The discrepancy between the tissue tropism of MeV and the distribution of SLAM-positive cells suggests that there are unknown receptors other than SLAM for MeV. Here we identified CD147/EMMPRIN (extracellular matrix metalloproteinase inducer), a transmembrane glycoprotein, which acts as a receptor for MeV on epithelial cells. Furthermore, we found the incorporation of cyclophilin B (CypB), a cellular ligand for CD147, in MeV virions, and showed that inhibition of CypB incorporation significantly attenuated SLAM-independent infection on epithelial cells, while it had no effect on SLAM-dependent infection. To date, MeV infection was considered to be triggered by binding of its hemagglutinin (H) protein and cellular receptors. Our present study, however, indicates that MeV infection also occurs via CD147 and virion-associated CypB, independently of MeV H. Since CD147 is expressed in a variety of cells, including epithelial and neuronal cells, this molecule possibly functions as an entry receptor for MeV in SLAM-negative cells. This is the first report among members of the Mononegavirales that CD147 is used as a virus entry receptor via incorporated CypB in the virions.


Subject(s)
Basigin/physiology , Epithelial Cells/virology , Measles virus/physiology , Receptors, Virus , Virus Internalization , Amino Acid Sequence , Cell Line , Cyclophilins/analysis , Humans , Molecular Sequence Data , Virion/chemistry
14.
Virology ; 375(2): 321-30, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18374960

ABSTRACT

Measles virus (MV) causes various responses including the induction of immune responses, transient immunosuppression and establishment of long-lasting immunity. To obtain a comprehensive view of the effects of MV infection on target cells, DNA microarray analyses of two different cell-types were performed. An epithelial (293SLAM; a 293 cell line stably expressing SLAM) and lymphoid (COBL-a) cell line were inoculated with purified wild-type MV. Microarray analyses revealed significant differences in the regulation of cellular gene expression between these two different cells. In 293SLAM cells, upregulation of genes involved in the antiviral response was rapidly induced; in the later stages of infection, this was followed by regulation of many genes across a broad range of functional categories. On the other hand, in COBL-a cells, only a limited set of gene expression profiles was modulated after MV infection. Since it was reported that V protein of MV inhibited the IFN signaling pathway, we performed a microarray analysis using V knockout MV to evaluate V protein's effect on cellular gene expression. The V knockout MV displayed a similar profile to that of parental MV. In particular, in COBL-a cells infected with the virus, no alteration of cellular gene expression, including IFN signaling, was observed. Furthermore, IFN signaling analyzed in vitro was completely suppressed by MV infection in the COBL-a cells. These results reveal that MV induces different cellular responses in a cell-type specific manner. Microarray analyses will provide us useful information about potential mechanisms of MV pathogenesis.


Subject(s)
Gene Expression Regulation , Measles virus/pathogenicity , Measles/genetics , Down-Regulation , Epithelial Cells/metabolism , Gene Expression Profiling , Humans , Interferons/metabolism , Lymphocytes/metabolism , Measles/virology , Measles virus/genetics , Phosphoproteins/genetics , Point Mutation , Signal Transduction/genetics , Up-Regulation , Viral Proteins/genetics , Virulence
15.
Proteomics ; 8(9): 1871-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18442172

ABSTRACT

We report the first identification of phosphorylation sites of the nucleoprotein (N) of the family Paramyxoviridae. The N protein is known to be the most abundant protein in infected cells; it constructs the N-RNA complex (nucleocapsid) and supports transcription and replication of viral genomic RNA. To determine the role of phosphorylation of the N protein, we expressed the N protein of the HL strain of measles virus (MV) in mammalian cells and purified the nucleocapsid. After separation of the C-terminal region from the core region, phosphorylated amino acids were assayed using MALDI-TOF/TOF and ESI-Q-TOF MS analyses. Two amino acids, S479 and S510, were shown to be phosphorylated by both methods of analysis. Metabolic labeling of the N protein with (32)P demonstrated that these two sites are the major phosphorylated sites within the MV-N protein. In transcriptional analysis using negative-strand minigenomic RNA containing the ORF of the luciferase gene, mutants of each phosphorylation site showed approximately 80% reduction in luciferase activity compared with the wild-type N, suggesting that the phosphorylation of N protein is important in the activation of the transcription of viral mRNA and/or replication of the genome in vivo.


Subject(s)
Gene Expression Regulation, Viral , Nucleoproteins/chemistry , Proteomics/methods , RNA, Viral/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleocapsid Proteins , Phosphorylation , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
16.
Proc Natl Acad Sci U S A ; 103(44): 16508-13, 2006 Oct 31.
Article in English | MEDLINE | ID: mdl-17053073

ABSTRACT

Nipah virus (NiV), a paramyxovirus, was first discovered in Malaysia in 1998 in an outbreak of infection in pigs and humans and incurred a high fatality rate in humans. Fruit bats, living in vast areas extending from India to the western Pacific, were identified as the natural reservoir of the virus. However, the mechanisms that resulted in severe pathogenicity in humans (up to 70% mortality) and that enabled crossing the species barrier were not known. In this study, we established a system that enabled the rescue of replicating NiVs from a cloned DNA by cotransfection of a constructed full-length cDNA clone and supporting plasmids coding virus nucleoprotein, phosphoprotein, and polymerase with the infection of the recombinant vaccinia virus, MVAGKT7, expressing T7 RNA polymerase. The rescued NiV (rNiV), by using the newly developed reverse genetics system, showed properties in vitro that were similar to the parent virus and retained the severe pathogenicity in a previously established animal model by experimental infection. A recombinant NiV was also developed, expressing enhanced green fluorescent protein (rNiV-EGFP). Using the virus, permissibility of NiV was compared with the presence of a known cellular receptor, ephrin B2, in a number of cell lines of different origins. Interestingly, two cell lines expressing ephrin B2 were not susceptible for rNiV-EGFP, indicating that additional factors are clearly required for full NiV replication. The reverse genetics for NiV will provide a powerful tool for the analysis of the molecular mechanisms of pathogenicity and cross-species infection.


Subject(s)
Henipavirus Infections/pathology , Henipavirus Infections/virology , Nipah Virus/physiology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Ephrin-B2/genetics , Ephrin-B2/metabolism , Henipavirus Infections/genetics , Henipavirus Infections/metabolism , Humans , Nipah Virus/pathogenicity , Plasmids/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Survival Rate , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...