Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Microbiol Immunol ; 68(4): 160-164, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38414102

ABSTRACT

Immunological testing to detect neutralizing antibodies (NAbs) is important in measles (MV) infection control. Currently, the plaque reduction neutralization test is the only credible method for measuring actual virus NAbs; however, its feasibility is hampered by drawbacks, such as long turnaround times, low throughput, and the need for laboratory biosafety equipment. To solve these problems, we developed a simple and rapid MV-NAb detection system using lentivirus-based virus-like particles incorporated with the NanoLuc fragment peptide HiBiT comprising the MV fusion protein and hemagglutinin on their exterior surface. Overall, this simple, safe, and rapid method could be used to detect MV NAbs.


Subject(s)
Measles virus , Measles , Humans , Antibodies, Viral , Antibodies, Neutralizing , Hemagglutinins, Viral , Neutralization Tests
2.
Microbiol Immunol ; 67(4): 204-209, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36609846

ABSTRACT

Bovine parainfluenza virus type 3 (BPIV3) is a promising vaccine vector against various respiratory virus infections, including the human PIV3, respiratory syncytial virus, and severe acute respiratory syndrome-coronavirus 2 infections. In this study, we combined the Magnet system and reverse genetic approach to generate photocontrollable BPIV3. An optically controllable Magnet gene was inserted into the H2 region of the BPIV3 large protein gene, which encodes an RNA-dependent RNA polymerase. The generated photocontrollable BPIV3 grew in specific regions of the cell sheet only when illuminated with blue light, suggesting that spatiotemporal control can aid in safe clinical applications of BPIV3.


Subject(s)
COVID-19 , Respiratory Syncytial Virus, Human , Animals , Cattle , Humans , Parainfluenza Virus 3, Human/genetics , Cell Line , Virus Replication , Parainfluenza Virus 3, Bovine/genetics
3.
Proc Natl Acad Sci U S A ; 117(13): 7001-7003, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32165541

ABSTRACT

A novel betacoronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which caused a large respiratory outbreak in Wuhan, China in December 2019, is currently spreading across many countries globally. Here, we show that a TMPRSS2-expressing VeroE6 cell line is highly susceptible to SARS-CoV-2 infection, making it useful for isolating and propagating SARS-CoV-2. Our results reveal that, in common with SARS- and Middle East respiratory syndrome-CoV, SARS-CoV-2 infection is enhanced by TMPRSS2.


Subject(s)
Betacoronavirus/isolation & purification , Coronavirus Infections/virology , Pneumonia, Viral/virology , Serine Endopeptidases/metabolism , Animals , COVID-19 , Cell Line , Chlorocebus aethiops , Disease Outbreaks , Humans , Pandemics , RNA, Viral/metabolism , SARS-CoV-2 , Vero Cells , Virus Cultivation
4.
Proc Natl Acad Sci U S A ; 116(24): 11587-11589, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31138700

ABSTRACT

Mononegaviruses are promising tools as oncolytic vectors and transgene delivery vectors for gene therapy and regenerative medicine. By using the Magnet proteins, which reversibly heterodimerize upon blue light illumination, photocontrollable mononegaviruses (measles and rabies viruses) were generated. The Magnet proteins were inserted into the flexible domain of viral polymerase, and viruses showed strong replication and oncolytic activities only when the viral polymerases were activated by blue light illumination.


Subject(s)
Measles virus/genetics , Oncolytic Viruses/genetics , Rabies virus/genetics , Animals , Cell Line, Tumor , DNA-Directed RNA Polymerases/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Light , Mice, Inbred BALB C , Mice, Nude , Oncolytic Virotherapy/methods , Transgenes/genetics , Virus Replication/genetics
5.
Mol Ther ; 28(1): 129-141, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31677955

ABSTRACT

Recent advances in gene therapy technologies have enabled the treatment of congenital disorders and cancers and facilitated the development of innovative methods, including induced pluripotent stem cell (iPSC) production and genome editing. We recently developed a novel non-transmissible and non-integrating measles virus (MV) vector capable of transferring multiple genes simultaneously into a wide range of cells through the CD46 and CD150 receptors. The MV vector expresses four genes for iPSC generation and the GFP gene for a period of time sufficient to establish iPSCs from human fibroblasts as well as peripheral blood T cells. The transgenes were expressed differentially depending on their gene order in the vector. Human hematopoietic stem/progenitor cells were directly and efficiently reprogrammed to naive-like cells that could proliferate and differentiate into primed iPSCs by the same method used to establish primed iPSCs from other cell types. The novel MV vector has several advantages for establishing iPSCs and potential future applications in gene therapy.


Subject(s)
Cellular Reprogramming/genetics , Genetic Vectors , Genome, Viral/genetics , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Measles virus/genetics , RNA, Viral/genetics , Animals , Blood Donors , Cell Differentiation/genetics , Fibroblasts/metabolism , Genetic Therapy/methods , HEK293 Cells , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Sendai virus/genetics , T-Lymphocytes/metabolism , Transduction, Genetic , Transgenes
6.
J Virol ; 91(6)2017 03 15.
Article in English | MEDLINE | ID: mdl-28053100

ABSTRACT

Paramyxoviral RNAs are synthesized by a viral RNA-dependent RNA polymerase (RdRp) consisting of the large (L) protein and its cofactor phosphoprotein (P protein). The L protein is a multifunctional protein that catalyzes RNA synthesis, mRNA capping, and mRNA polyadenylation. Growing evidence shows that the stability of several paramyxovirus L proteins is regulated by heat shock protein 90 (Hsp90). In this study, we demonstrated that Hsp90 activity was important for mumps virus (MuV) replication. The Hsp90 activity was required for L-protein stability and activity because an Hsp90-specific inhibitor, 17-allylamino-17-demethoxygeldanamycin (17-AAG), destabilized the MuV L protein and suppressed viral RNA synthesis. However, once the L protein formed a mature polymerase complex with the P protein, Hsp90 activity was no longer required for the stability and activity of the L protein. When the Hsp90 activity was inhibited, the MuV L protein was degraded through the CHIP (C terminus of Hsp70-interacting protein)-mediated proteasomal pathway. High concentrations of 17-AAG showed strong cytotoxicity to certain cell types, but combined use of an Hsp70 inhibitor, VER155008, potentiated degradation of the L protein, allowing a sufficient reduction of 17-AAG concentration to block MuV replication with minimum cytotoxicity. Regulation of the L protein by Hsp90 and Hsp70 chaperones was also demonstrated for another paramyxovirus, the measles virus. Collectively, our data show that the Hsp90/Hsp70 chaperone machinery assists in the maturation of the paramyxovirus L protein and thereby in the formation of a mature RdRp complex and efficient viral replication.IMPORTANCE Heat shock protein 90 (Hsp90) is nearly universally required for viral protein homeostasis. Here, we report that Hsp90 activity is required for efficient propagation of mumps virus (MuV). Hsp90 functions in the maintenance of the catalytic subunit of viral polymerase, the large (L) protein, prior to formation of a mature polymerase complex with the polymerase cofactor of L, phosphoprotein. Hsp70 collaborates with Hsp90 to regulate biogenesis of the MuV L protein. The functions of these chaperones on the viral polymerase may be common among paramyxoviruses because the L protein of measles virus is also similarly regulated. Our data provide important insights into the molecular mechanisms of paramyxovirus polymerase maturation as well as a basis for the development of novel antiviral drugs.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions , Mumps virus/physiology , RNA-Dependent RNA Polymerase/metabolism , Virus Replication , Animals , Cell Line , Chlorocebus aethiops , Epithelial Cells/virology , Humans , Measles virus/physiology , Protein Stability , Proteolysis
7.
Uirusu ; 67(1): 3-16, 2017.
Article in Japanese | MEDLINE | ID: mdl-29593149

ABSTRACT

Measles virus (MeV) is exceptionally contagious and still a major cause of death in child.However, recently significant progress towards the elimination of measles has been made through increased vaccination coverage of measles-containing vaccines. The hemagglutinin (H) protein of MeV interacts with a cellular receptor, and this interaction is the first step of infection. MeV uses two different receptors, signaling lymphocyte activation molecule (SLAM) and nectin-4 expressed on immune cells and epithelial cells, respectively. The interactions of MeV with these receptors nicely explain the immune suppressive and high contagious properties of MeV. Binding of the H protein to a receptor triggers conformational changes in the fusion (F) protein, inducing fusion between viral and host plasma membranes for entry. The stalk region of the H protein plays a key role in the F protein-triggering. Recent studies of the H protein epitopes have revealed that the receptor binding site of the H protein constitutes a major neutralizing epitope. The interaction with two proteinaceous receptors probably imposes strong functional constraints on this epitope for amino acid changes. This would be a reason why measles vaccines, which are derived from MV strains isolated more than 60 years ago, are still highly effective against all MV strains currently circulating.


Subject(s)
Measles Vaccine , Measles virus , Animals , Cell Adhesion Molecules/metabolism , Epitopes , Hemagglutinins, Viral/chemistry , Hemagglutinins, Viral/metabolism , Hemagglutinins, Viral/physiology , Humans , Measles virus/pathogenicity , Protein Binding , Protein Structure, Secondary , Receptors, Virus/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , Viral Fusion Proteins/chemistry , Virus Internalization
8.
Hepatology ; 61(2): 437-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25203900

ABSTRACT

UNLABELLED: The human liver reacts to hepatitis C virus (HCV) with a balanced response consisting of host anti- and proviral activities. To explore these subtle host responses, we used oligonucleotide microarrays to investigate the differential gene expression between two groups of liver samples with high and low HCV loads (>100-fold difference). We identified and validated 26 genes that were up-regulated in livers with high HCV loads, including transmembrane protease serine 2 (TMPRSS2). Trypsin inhibitors inhibited the infection of Huh7-25-CD81 cells with cell-culture-derived HCV (HCVcc) of Japanese fulminant hepatitis 1 isolate at the postbinding and entry step, and trypsin enhanced HCVcc infection at an early stage of infection. Several major transmembrane serine proteases, in particular, furin and hepsin, were detected in Huh7-25-CD81 cells, but TMPRSS2 was not. Huh7-25-CD81 cell clones stably expressing TMPRSS2- WT (wild type) and inactive TMPRSS2-mutant genes showed positive and negative enhancement of their susceptibility to HCVcc infection, respectively. The enhanced susceptibility of TMPRSS2-WT Huh7-25-CD81 cells was confirmed by knockdown of TMPRSS2 using small interfering RNA. The cell-surface protease activity of TMPRSS2-WT cells was markedly active in the cleavage of QAR and QGR, corresponding to amino acid residues at P3 to P1. CONCLUSION: The cell-surface activity of a trypsin-like serine protease, such as TMPRSS2, activates HCV infection at the postbinding and entry stage. Host transmembrane serine proteases may be involved in the sensitivity, persistence, and pathogenesis of HCV infection and be possible targets for antiviral therapy.


Subject(s)
Hepatitis C, Chronic/metabolism , Host-Pathogen Interactions , Serine Endopeptidases/metabolism , Aged , Cell Line , Female , Gene Expression Profiling , Hepatitis C, Chronic/virology , Humans , Liver/metabolism , Liver/virology , Male , Middle Aged
9.
J Virol ; 88(10): 5608-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24600012

ABSTRACT

UNLABELLED: Proteolytic cleavage of the hemagglutinin (HA) protein is essential for influenza A virus (IAV) to acquire infectivity. This process is mediated by a host cell protease(s) in vivo. The type II transmembrane serine protease TMPRSS2 is expressed in the respiratory tract and is capable of activating a variety of respiratory viruses, including low-pathogenic (LP) IAVs possessing a single arginine residue at the cleavage site. Here we show that TMPRSS2 plays an essential role in the proteolytic activation of LP IAVs, including a recently emerged H7N9 subtype, in vivo. We generated TMPRSS2 knockout (KO) mice. The TMPRSS2 KO mice showed normal reproduction, development, and growth phenotypes. In TMPRSS2 KO mice infected with LP IAVs, cleavage of HA was severely impaired, and consequently, the majority of LP IAV progeny particles failed to gain infectivity, while the viruses were fully activated proteolytically in TMPRSS2+/+ wild-type (WT) mice. Accordingly, in contrast to WT mice, TMPRSS2 KO mice were highly tolerant of challenge infection by LP IAVs (H1N1, H3N2, and H7N9) with ≥1,000 50% lethal doses (LD50) for WT mice. On the other hand, a high-pathogenic H5N1 subtype IAV possessing a multibasic cleavage site was successfully activated in the lungs of TMPRSS2 KO mice and killed these mice, as observed for WT mice. Our results demonstrate that recently emerged H7N9 as well as seasonal IAVs mainly use the specific protease TMPRSS2 for HA cleavage in vivo and, thus, that TMPRSS2 expression is essential for IAV replication in vivo. IMPORTANCE: Influenza A virus (IAV) is a leading pathogen that infects and kills many humans every year. We clarified that the infectivity and pathogenicity of IAVs, including a recently emerged H7N9 subtype, are determined primarily by a host protease, TMPRSS2. Our data showed that TMPRSS2 is the key host protease that activates IAVs in vivo through proteolytic cleavage of their HA proteins. Hence, TMPRSS2 is a good target for the development of anti-IAV drugs. Such drugs could also be effective for many other respiratory viruses, including the recently emerged Middle East respiratory syndrome (MERS) coronavirus, because they are also activated by TMPRSS2 in vitro. Consequently, the present paper could have a large impact on the battle against respiratory virus infections and contribute greatly to human health.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H7N9 Subtype/physiology , Serine Endopeptidases/metabolism , Virus Replication , Animals , Disease Models, Animal , Female , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H5N1 Subtype/physiology , Lethal Dose 50 , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Serine Endopeptidases/deficiency , Survival Analysis
10.
J Immunol ; 191(9): 4740-7, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24078691

ABSTRACT

Infection by measles virus (MV) induces type I IFN via the retinoic acid-inducible gene I/melanoma differentiation-associated gene 5/mitochondrial antiviral signaling protein (MAVS) pathway in human cells. However, the in vivo role of the MAVS pathway in host defense against MV infection remains undetermined. CD150 transgenic (Tg) mice, which express human CD150, an entry receptor for MV, with the disrupting IFNR gene (Ifnar(-/-)), are susceptible to MV and serve as a model for MV infection. In this study, we generated CD150Tg/Mavs(-/-) mice and examined MV permissiveness compared with that in CD150Tg/Ifnar(-/-) mice. MV replicated mostly in the spleen of i.p.-infected CD150Tg/Ifnar(-/-) mice. Strikingly, CD150Tg/Mavs(-/-) mice were not permissive to MV in vivo because of substantial type I IFN induction. MV barely replicated in any other organs tested. When T cells, B cells, and dendritic cells (DCs) isolated from CD150Tg/Mavs(-/-) splenocytes were cultured with MV in vitro, only the DCs produced type I IFN. In vitro infection analysis using CD150Tg/Mavs(-/-) DC subsets revealed that CD4(+) and plasmacytoid DCs, but not CD8α(+) and CD8α(-)CD4(-) double negative DCs, were exclusively involved in type I IFN production in response to MV infection. Because CD150Tg/Mavs(-/-) mice turned permissive to MV by anti-IFNAR Ab, type I IFN produced by CD4(+) DCs and plasmacytoid DCs plays a critical role in antiviral protection for neighboring cells expressing IFNAR. Induction of type I IFN in these DC subsets was abolished by the MyD88 inhibitory peptide. Thus, production of type I IFN occurs via the MyD88-dependent and MAVS-independent signaling pathway during MV infection.


Subject(s)
Dendritic Cells/metabolism , Interferon Type I/biosynthesis , Measles virus/immunology , Measles/immunology , Myeloid Differentiation Factor 88/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/biosynthesis , Antigens, CD/metabolism , B-Lymphocytes/immunology , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cells, Cultured , Dendritic Cells/immunology , Disease Models, Animal , Humans , Measles/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/biosynthesis , Receptor, Interferon alpha-beta/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Spleen/cytology , T-Lymphocytes/immunology
11.
J Virol ; 87(6): 3583-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23283964

ABSTRACT

Here, we provide direct evidence that the receptor-binding site of measles virus (MV) hemagglutinin protein itself forms an effective conserved neutralizing epitope (CNE). Several receptor-interacting residues constitute the CNE. Thus, viral escape from neutralization has to be associated with loss of receptor-binding activity. Since interactions with both the signaling lymphocyte activation molecule (SLAM) and nectin4 are critical for MV pathogenesis, its escape, which results from loss of receptor-binding activity, should not occur in nature.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Epitopes, B-Lymphocyte/immunology , Hemagglutinins, Viral/immunology , Measles virus/immunology
12.
J Virol ; 87(1): 666-75, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115278

ABSTRACT

Effective vaccination programs have dramatically reduced the number of measles-related deaths globally. Although all the available data suggest that measles eradication is biologically feasible, a structural and biochemical basis for the single serotype nature of measles virus (MV) remains to be provided. The hemagglutinin (H) protein, which binds to two discrete proteinaceous receptors, is the major neutralizing target. Monoclonal antibodies (MAbs) recognizing distinct epitopes on the H protein were characterized using recombinant MVs encoding the H gene from different MV genotypes. The effects of various mutations on neutralization by MAbs and virus fitness were also analyzed, identifying the location of five epitopes on the H protein structure. Our data in the present study demonstrated that the H protein of MV possesses at least two conserved effective neutralizing epitopes. One, which is a previously recognized epitope, is located near the receptor-binding site (RBS), and thus MAbs that recognize this epitope blocked the receptor binding of the H protein, whereas the other epitope is located at the position distant from the RBS. Thus, a MAb that recognizes this epitope did not inhibit the receptor binding of the H protein, rather interfered with the hemagglutinin-fusion (H-F) interaction. This epitope was suggested to play a key role for formation of a higher order of an H-F protein oligomeric structure. Our data also identified one nonconserved effective neutralizing epitope. The epitope has been masked by an N-linked sugar modification in some genotype MV strains. These data would contribute to our understanding of the antigenicity of MV and support the global elimination program of measles.


Subject(s)
Antibodies, Neutralizing/immunology , Epitopes/genetics , Epitopes/immunology , Measles virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Antibodies, Monoclonal/immunology , Humans , Measles virus/genetics , Mutant Proteins/genetics , Mutant Proteins/immunology , Neutralization Tests
13.
J Virol ; 87(12): 7170-5, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23596291

ABSTRACT

A canine distemper virus (CDV) strain, CYN07-dV, associated with a lethal outbreak in monkeys, used human signaling lymphocyte activation molecule as a receptor only poorly but readily adapted to use it following a P541S substitution in the hemagglutinin protein. Since CYN07-dV had an intrinsic ability to use human nectin-4, the adapted virus became able to use both human immune and epithelial cell receptors, as well as monkey and canine ones, suggesting that CDV can potentially infect humans.


Subject(s)
Adaptation, Physiological/genetics , Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Disease Outbreaks , Distemper Virus, Canine/metabolism , Macaca/virology , Monkey Diseases/virology , Receptors, Cell Surface/metabolism , Amino Acid Substitution , Animals , Chlorocebus aethiops , Distemper/epidemiology , Distemper/virology , Distemper Virus, Canine/genetics , Distemper Virus, Canine/pathogenicity , Dogs , Epithelial Cells/metabolism , Epithelial Cells/virology , Hemagglutinins, Viral/genetics , Humans , Monkey Diseases/epidemiology , Monkey Diseases/mortality , Receptors, Virus/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells
14.
J Virol ; 87(21): 11930-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966399

ABSTRACT

Here, we show that human parainfluenza viruses and Sendai virus (SeV), like other respiratory viruses, use TMPRSS2 for their activation. The membrane fusion proteins of respiratory viruses often possess serine and glutamine residues at the P2 and P3 positions, respectively, but these residues were not critical for cleavage by TMPRSS2. However, mutations of these residues affected SeV growth in specific epithelial cell lines, suggesting the importance of these residues for SeV replication in epithelia.


Subject(s)
Host-Pathogen Interactions , Paramyxovirinae/physiology , Serine Endopeptidases/metabolism , Virus Replication , Animals , Cell Line , Epithelial Cells/virology , Humans , Viral Load , Viral Plaque Assay
15.
Methods Mol Biol ; 2808: 35-56, 2024.
Article in English | MEDLINE | ID: mdl-38743361

ABSTRACT

Mononegaviruses are promising tools as oncolytic and transgene vectors for gene therapy and regenerative medicine. However, when mononegaviruses are used for therapeutic applications, the viral activity must be strictly controlled due to concerns about toxicity and severe side effects. With this technology, mononegavirus vectors can be grown where they are intended and can be easily removed when they are no longer needed. In particular, a photoswitch protein called Magnet (consisting of two magnet domains) is incorporated into the hinge region between the connector and methyltransferase domains of the mononegavirus polymerase protein (L protein) to disrupt the L protein functions. Blue light (470 ± 20 nm) irradiation causes the dimerization of the two magnet domains, and the L protein is restored to activity, allowing viral gene expression and virus replication. Since the magnet domains' dimerization is reversible, viral gene expression and replication cease when blue light irradiation is stopped.


Subject(s)
Gene Expression Regulation, Viral , Virus Replication , Virus Replication/genetics , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Light , Animals , Genetic Vectors/genetics
16.
Nat Commun ; 13(1): 6100, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36243815

ABSTRACT

In cultured cells, SARS-CoV-2 infects cells via multiple pathways using different host proteases. Recent studies have shown that the furin and TMPRSS2 (furin/TMPRSS2)-dependent pathway plays a minor role in infection of the Omicron variant. Here, we confirm that Omicron uses the furin/TMPRSS2-dependent pathway inefficiently and enters cells mainly using the cathepsin-dependent endocytosis pathway in TMPRSS2-expressing VeroE6/TMPRSS2 and Calu-3 cells. This is the case despite efficient cleavage of the spike protein of Omicron. However, in the airways of TMPRSS2-knockout mice, Omicron infection is significantly reduced. We furthermore show that propagation of the mouse-adapted SARS-CoV-2 QHmusX strain and human clinical isolates of Beta and Gamma is reduced in TMPRSS2-knockout mice. Therefore, the Omicron variant isn't an exception in using TMPRSS2 in vivo, and analysis with TMPRSS2-knockout mice is important when evaluating SARS-CoV-2 variants. In conclusion, this study shows that TMPRSS2 is critically important for SARS-CoV-2 infection of murine airways, including the Omicron variant.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Humans , Mice , Cathepsins , Furin/genetics , Furin/metabolism , Mice, Knockout , Peptide Hydrolases , Serine Endopeptidases/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization
17.
J Biol Chem ; 285(27): 20882-90, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20435897

ABSTRACT

Measles virus (MV), an enveloped negative-strand RNA virus, remains a major cause of morbidity and mortality in developing countries. MV predominantly infects immune cells by using signaling lymphocyte activation molecule (SLAM; also called CD150) as a receptor, but it also infects polarized epithelial cells, forming tight junctions in a SLAM-independent manner. Although the ability of MV to infect polarized epithelial cells is thought to be important for its transmission, the epithelial cell receptor for MV has not been identified. A transcriptional repressor, Snail, induces epithelial-mesenchymal transition (EMT), in which epithelial cells lose epithelial cell phenotypes, such as adherens and tight junctions. In this study, EMT was induced by expressing Snail in a lung adenocarcinoma cell line, II-18, which is highly susceptible to wild-type MV. Snail-expressing II-18 cells lost adherens and tight junctions. Microarray analysis confirmed the induction of EMT in II-18 cells and suggested a novel function of Snail in protein degradation and distribution. Importantly, wild-type MV no longer entered EMT-induced II-18 cells, suggesting that the epithelial cell receptor is down-regulated by the induction of EMT. Other polarized cell lines, NCI-H358 and HT-29, also lost susceptibility to wild-type MV when EMT was induced. However, the complete formation of tight junctions rather reduced MV entry into HT-29 cells. Taken together, these data suggest that the unidentified epithelial cell receptor for MV is involved in the formation of epithelial intercellular junctions.


Subject(s)
Epithelial Cells/cytology , Measles virus/pathogenicity , Measles/prevention & control , Mesoderm/cytology , Animals , Cell Line , Disease Susceptibility , Epithelial Cells/physiology , Epithelial Cells/virology , Flow Cytometry , Genetic Predisposition to Disease , Genetic Vectors , Genome, Viral , Humans , Lymphocyte Activation , Macaca mulatta/virology , Measles/immunology , Measles/transmission , Measles/veterinary , Measles virus/genetics , Membrane Cofactor Protein/physiology , Mesoderm/physiology , Mesoderm/virology , Oligonucleotide Array Sequence Analysis , Plasmids , Receptors, Virus/physiology , Tight Junctions/physiology , Virus Shedding
18.
Uirusu ; 61(2): 249-55, 2011 Dec.
Article in Japanese | MEDLINE | ID: mdl-22916571

ABSTRACT

Measles is a highly contagious acute viral disease characterized by a maculopapular rash. It causes severe and temporary immune suppression and is often accompanied by secondary bacterial infections. In 2000, signaling lymphocyte activation molecule (SLAM) was identified as a receptor for measles virus (MV). Observations that SLAM is expressed on cells of the immune system provided a good explanation for the lymphotropic and immunosuppressive nature of MV. However, molecular mechanisms of highly contagious nature of MV have remained unclear. Previously we have demonstrated that MV has an intrinsic ability to infect polarized epithelial cells by using a receptor other than SLAM. Recently, nectin4, a cellular adhesion junction molecule, was identified as the epithelial cell receptor for MV. Understanding the molecular mechanisms of MV to infect both epithelial and immune cells provides a deep insight into measles pathogenesis.


Subject(s)
Antigens, CD , Cell Adhesion Molecules , Measles virus/genetics , Measles virus/pathogenicity , Measles/virology , Membrane Cofactor Protein , Receptors, Cell Surface , Animals , Epithelial Cells/virology , Humans , Immune System/cytology , Immune System/virology , Measles/immunology , Measles virus/immunology , Signaling Lymphocytic Activation Molecule Family Member 1
19.
FEBS J ; 287(1): 145-159, 2020 01.
Article in English | MEDLINE | ID: mdl-31287622

ABSTRACT

The measles virus (MV) is a major cause of childhood morbidity and mortality worldwide. We previously established a mouse monoclonal antibody, 2F4, which shows high neutralizing titers against eight different genotypes of MV. However, the molecular basis for the neutralizing activity of the 2F4 antibody remains incompletely understood. Here, we have evaluated the binding characteristics of a Fab fragment of the 2F4 antibody. Using the MV infectious assay, we demonstrated that 2F4 Fab inhibits viral entry via either of two cellular receptors, SLAM and Nectin4. Surface plasmon resonance (SPR) analysis of recombinant proteins indicated that 2F4 Fab interacts with MV hemagglutinin (MV-H) with a KD value at the nm level. Furthermore, we designed a single-chain Fv fragment of 2F4 antibody as another potential biopharmaceutical to target measles. The stable 2F4 scFv was successfully prepared by the refolding method and shown to interact with MV-H at the µm level. Like 2F4 Fab, scFv inhibited receptor binding and viral entry. This indicates that 2F4 mAb uses the receptor-binding site and/or a neighboring region as an epitope with high affinity. These results provide insight into the neutralizing activity and potential therapeutic use of antibody fragments for MV infection.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunoglobulin Fab Fragments/immunology , Measles virus/immunology , Measles/immunology , Single-Chain Antibodies/immunology , Viral Proteins/immunology , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/genetics , Antibodies, Viral/biosynthesis , Antibodies, Viral/genetics , Epitopes/immunology , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/genetics , Measles/virology , Nectins/antagonists & inhibitors , Nectins/immunology , Nectins/metabolism , Protein Binding , Signaling Lymphocytic Activation Molecule Family Member 1/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family Member 1/immunology , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Virus Internalization
20.
J Virol ; 82(9): 4630-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18287234

ABSTRACT

Measles is one of the most contagious human infectious diseases and remains a major cause of childhood morbidity and mortality worldwide. The signaling lymphocyte activation molecule (SLAM), also called CD150, is a cellular receptor for measles virus (MV), presumably accounting for its tropism for immune cells and its immunosuppressive properties. On the other hand, pathological studies have shown that MV also infects epithelial cells at a later stage of infection, although its mechanism has so far been unknown. In this study, we show that wild-type MV can infect and produce syncytia in human polarized epithelial cell lines independently of SLAM and CD46 (a receptor for the vaccine strains of MV). Progeny viral particles are released exclusively from the apical surface of these polarized epithelial cell lines. We have also identified amino acid residues on the MV attachment protein that are likely to interact with a putative receptor on epithelial cells. All of these residues have aromatic side chains and may form a receptor-binding pocket located in a different position from the putative SLAM- and CD46-binding sites on the MV attachment protein. Thus, our results indicate that MV has an intrinsic ability to infect both polarized epithelial and immune cells by using distinctive receptor-binding sites on the attachment protein corresponding to each of their respective receptors. The ability of MV to infect polarized epithelial cells and its exclusive release from the apical surface may facilitate its efficient transmission via aerosol droplets, resulting in its highly contagious nature.


Subject(s)
Epithelial Cells/virology , Hemagglutinins/metabolism , Lymphocytes/virology , Measles virus/pathogenicity , Animals , Antigens, CD , Binding Sites , Cell Line , Cell Polarity , Humans , Immune System/cytology , Immune System/virology , Membrane Cofactor Protein , Receptors, Cell Surface , Receptors, Virus/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Virus Attachment
SELECTION OF CITATIONS
SEARCH DETAIL