Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Virology ; 540: 57-65, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31739185

ABSTRACT

Among the Herpesviridae, human cytomegalovirus (HCMV) owns the largest genome and displays a huge coding potential. Here, we characterized the UL5 gene product (pUL5) of the clinical isolate TR strain. The protein was predicted as a 166-amino-acid membrane protein with a theoretical mass of 19 kDa. Recombinant virus expressing pUL5 with a tag allowed the identification of two pUL5 non-glycosylated species of approximately 19 and 9 kDa, expressed with early and late kinetic respectively. Experiments in infection confirmed that the lower molecular weight species was translated from an internal ATG in the UL5 open reading frame. Confocal microscopy analysis showed that pUL5 localized within the assembly compartment, but is not incorporated in the virion, as shown by Western blot on purified viral particles. Finally, pull-down experiments coupled with mass spectrometry analysis identified IQGAP1 as a pUL5 interactor, giving new hints on possible roles of pUL5 during HCMV infection.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Host-Pathogen Interactions , Viral Proteins/metabolism , ras GTPase-Activating Proteins/metabolism , Amino Acid Sequence , Cell Line , Cells, Cultured , Cytomegalovirus/isolation & purification , Cytomegalovirus/ultrastructure , Gene Expression Regulation, Viral , Humans , Open Reading Frames , Protein Binding , Protein Transport , RNA, Viral , Transcription, Genetic
2.
Zoolog Sci ; 34(2): 112-121, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28397602

ABSTRACT

To study the effects of post-glacial isolation by islands on population genetic diversity and differentiation of the large Japanese field mouse, Apodemus speciosus, we examined partial nucleotide sequences of the mitochondrial Dloop region (ca. 300 bp) in 231 individuals collected from islands in the Seto Inland Sea and adjacent regions on Honshu and Shikoku Islands in the western part of the Japanese archipelago. Molecular phylogenetic and network analyses showed that haplotypes in each island tended to form monophyletic groups, while those in Honshu and Shikoku (the major Japanese islands) showed scattered relationships and were connected with island haplotypes. These observations suggest that a set of Honshu and Shikoku haplotypes became the ancestral lineages of the island population. No gene flow was detected among island populations, indicating that independent evolution occurred on each island, without the influence of human activities, since the establishment of the islands in the Holocene. Population genetic diversities on each island were lower than those on Honshu and Shikoku. Comparison between genetic diversity and island area size showed positive correlations and supported the suggestion that genetic drift is a major factor that shaped the current haplotype constitution of the islands in the Seto Inland Sea.


Subject(s)
Animal Distribution/physiology , DNA, Mitochondrial/genetics , Genetic Variation , Islands , Murinae/genetics , Animals , Base Sequence , Haplotypes , Japan , Phenelzine , Phylogeny , Protein Binding
3.
Immunol Cell Biol ; 94(9): 849-860, 2016 10.
Article in English | MEDLINE | ID: mdl-27192938

ABSTRACT

Human cytomegalovirus (HCMV) is known to exert suppressive effects on the host immune system through expression of various viral genes, thus directly and indirectly affecting antiviral immunity of the infected individuals. We report here that HCMV UL10 encodes a protein (pUL10) with immunosuppressive properties. UL10 has been classified as a member of the HCMV RL11 gene family. Although pUL10 is known to be dispensable for viral replication in cultured cells, its amino-acid sequence is well conserved among different HCMV isolates, suggesting that the protein has a crucial role in viral survival in the host environment. We show that pUL10 is cleaved from the cell surface of fibroblasts as well as epithelial cells and interacts with a cellular receptor ubiquitously expressed on the surface of human leukocytes, demonstrated by ex vivo cell-based assays and flow cytometric analyses on both lymphoid cell lines and primary blood cells. Furthermore, preincubation of peripheral blood mononuclear cells with purified pUL10 ectodomain results in significantly impaired proliferation and substantially reduced pro-inflammatory cytokine production, in particular in CD4+ T cells upon in vitro T-cell stimulation. The inhibitory effect of pUL10 is also observed on antigen receptor-mediated intracellular tyrosine phosphorylation in a T-cell line. Based on these observations, we suggest that pUL10 is a newly identified immunomodulatory protein encoded by HCMV. Further elucidation of interactions between pUL10 and the host immune system during HCMV may contribute to finding ways towards new therapies for HCMV infection.


Subject(s)
Capsid Proteins/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Amino Acid Sequence , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Capsid Proteins/chemistry , Cell Line , Cell Membrane/metabolism , Cell Proliferation , Cytokines/biosynthesis , Glycosylation , HEK293 Cells , Humans , Membrane Proteins/metabolism , Recombinant Proteins/metabolism , Signal Transduction
4.
J Virol ; 90(10): 4926-38, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26937030

ABSTRACT

UNLABELLED: Human cytomegalovirus (HCMV) is a major cause of morbidity and mortality in transplant patients and is the leading viral cause of birth defects after congenital infection. HCMV infection relies on the recognition of cell-specific receptors by one of the viral envelope glycoprotein complexes. Either the gH/gL/gO or the gH/gL/UL128/UL130/UL131A (Pentamer) complex has been found to fulfill this role, accounting for HCMV entry into almost all cell types. We have studied the UL116 gene product, a putative open reading frame identified by in silico analysis and predicted to code for a secreted protein. Virus infection experiments in mammalian cells demonstrated that UL116 is expressed late in the HCMV replication cycle and is a heavily glycosylated protein that first localizes to the cellular site of virus assembly and then inserts into the virion envelope. Transient-transfection studies revealed that UL116 is efficiently transported to the plasma membrane when coexpressed with gH and that gL competes with UL116 for gH binding. Further evidence for gH/UL116 complex formation was obtained by coimmunoprecipitation experiments on both transfected and infected cells and biochemical characterization of the purified complex. In summary, our results show that the product of the UL116 gene is an HCMV envelope glycoprotein that forms a novel gH-based complex alternative to gH/gL. Remarkably, the gH/UL116 complex is the first herpesvirus gH-based gL-less complex. IMPORTANCE: HCMV infection can cause severe disease in immunocompromised adults and infants infected in utero The dissection of the HCMV entry machinery is important to understand the mechanism of viral infection and to identify new vaccine antigens. The gH/gL/gO and gH/gL/UL128/UL130/UL131 (Pentamer) complexes play a key role in HCMV cell entry and tropism. Both complexes are formed by an invariant gH/gL scaffold on which the other subunits assemble. Here, we show that the UL116 gene product is expressed in infected cells and forms a heterodimer with gH. The gH/UL116 complex is carried on the infectious virions, although in smaller amounts than gH/gL complexes. No gH/UL116/gL ternary complex formed in transfected cells, suggesting that the gH/UL116 complex is independent from gL. This new gH-based gL-free complex represents a potential target for a protective HCMV vaccine and opens new perspectives on the comprehension of the HCMV cell entry mechanism and tropism.


Subject(s)
Cytomegalovirus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Animals , Cell Line , Cytomegalovirus/chemistry , Genome, Viral , Humans , Microscopy, Electron , Mutation , Protein Multimerization , Transfection , Viral Envelope Proteins/chemistry , Virus Assembly , Virus Internalization
5.
PLoS One ; 10(8): e0135474, 2015.
Article in English | MEDLINE | ID: mdl-26280677

ABSTRACT

Developing a universal influenza vaccine that induces broad spectrum and longer-term immunity has become an important potentially achievable target in influenza vaccine research and development. Hemagglutinin (HA) and neuraminidase (NA) are the two major influenza virus antigens. Although antibody responses against influenza virus are mainly directed toward HA, NA is reported to be more genetically stable; hence NA-based vaccines have the potential to be effective for longer time periods. NA-specific immunity has been shown to limit the spread of influenza virus, thus reducing disease symptoms and providing cross-protection against heterosubtypic viruses in mouse challenge experiments. The production of large quantities of highly pure and stable NA could be beneficial for the development of new antivirals, subunit-based vaccines, and novel diagnostic tools. In this study, recombinant NA (rNA) was produced in mammalian cells at high levels from both swine A/California/07/2009 (H1N1) and avian A/turkey/Turkey/01/2005 (H5N1) influenza viruses. Biochemical, structural, and immunological characterizations revealed that the soluble rNAs produced are tetrameric, enzymatically active and immunogenic, and finally they represent good alternatives to conventionally used sources of NA in the Enzyme-Linked Lectin Assay (ELLA).


Subject(s)
Antigens, Viral/immunology , Lectins/immunology , Neuraminidase/immunology , Recombinant Proteins/immunology , Viral Proteins/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Birds , Cell Line , Cross Protection/immunology , Cross Reactions/immunology , Enzyme-Linked Immunospot Assay/methods , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza in Birds/immunology , Mice , Orthomyxoviridae Infections/immunology , Swine
6.
Proc Natl Acad Sci U S A ; 112(19): 6056-61, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25918416

ABSTRACT

Varicella-zoster virus (VZV), of the family Alphaherpesvirinae, causes varicella in children and young adults, potentially leading to herpes zoster later in life on reactivation from latency. The conserved herpesvirus glycoprotein gB and the heterodimer gHgL mediate virion envelope fusion with cell membranes during virus entry. Naturally occurring neutralizing antibodies against herpesviruses target these entry proteins. To determine the molecular basis for VZV neutralization, crystal structures of gHgL were determined in complex with fragments of antigen binding (Fabs) from two human monoclonal antibodies, IgG-94 and IgG-RC, isolated from seropositive subjects. These structures reveal that the antibodies target the same site, composed of residues from both gH and gL, distinct from two other neutralizing epitopes identified by negative-stain electron microscopy and mutational analysis. Inhibition of gB/gHgL-mediated membrane fusion and structural comparisons with herpesvirus homologs suggest that the IgG-RC/94 epitope is in proximity to the site on VZV gHgL that activates gB. Immunization studies proved that the anti-gHgL IgG-RC/94 epitope is a critical target for antibodies that neutralize VZV. Thus, the gHgL/Fab structures delineate a site of herpesvirus vulnerability targeted by natural immunity.


Subject(s)
Antibodies, Neutralizing/chemistry , Glycoproteins/chemistry , Herpesvirus 3, Human/immunology , Viral Envelope Proteins/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Viral/immunology , Crystallography, X-Ray , Epitopes/chemistry , Humans , Immunoglobulin Fragments/chemistry , Mice , Models, Molecular , Neutralization Tests , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Surface Plasmon Resonance
7.
Adv Genet ; 89: 179-233, 2015.
Article in English | MEDLINE | ID: mdl-25620012

ABSTRACT

This chapter provides a brief introduction to nucleic acid-based vaccines and recent research in developing self-amplifying mRNA vaccines. These vaccines promise the flexibility of plasmid DNA vaccines with enhanced immunogenicity and safety. The key to realizing the full potential of these vaccines is efficient delivery of nucleic acid to the cytoplasm of a cell, where it can amplify and express the encoded antigenic protein. The hydrophilicity and strong net negative charge of RNA impedes cellular uptake. To overcome this limitation, electrostatic complexation with cationic lipids or polymers and physical delivery using electroporation or ballistic particles to improve cellular uptake has been evaluated. This chapter highlights the rapid progress made in using nonviral delivery systems for RNA-based vaccines. Initial preclinical testing of self-amplifying mRNA vaccines has shown nonviral delivery to be capable of producing potent and robust innate and adaptive immune responses in small animals and nonhuman primates. Historically, the prospect of developing mRNA vaccines was uncertain due to concerns of mRNA instability and the feasibility of large-scale manufacturing. Today, these issues are no longer perceived as barriers in the widespread implementation of the technology. Currently, nonamplifying mRNA vaccines are under investigation in human clinical trials and can be produced at a sufficient quantity and quality to meet regulatory requirements. If the encouraging preclinical data with self-amplifying mRNA vaccines are matched by equivalently positive immunogenicity, potency, and tolerability in human trials, this platform could establish nucleic acid vaccines as a versatile new tool for human immunization.


Subject(s)
RNA, Messenger/administration & dosage , Vaccines/administration & dosage , Animals , Antigens/genetics , Electroporation , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA, Messenger/adverse effects , RNA, Messenger/genetics , Vaccines/adverse effects , Viral Vaccines
8.
Clin Vaccine Immunol ; 19(7): 991-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22623651

ABSTRACT

Antivector immunity has been recognized as a potential caveat of using virus-based vaccines. In the present study, an alphavirus-based replicon particle vaccine platform, which has demonstrated robust immunogenicity in animal models, was tested for effects of antivector immunity on immunogenicity against hemagglutinin of influenza virus as a target antigen and efficacy for protection against lethal challenge with the virus. Chimeric alphavirus-based replicon particles, comprising Venezuelan equine encephalitis virus nonstructural and Sindbis virus structural components, induced efficient protective antibody responses, which were not adversely influenced after multiple immunizations with the same vector expressing various antigens.


Subject(s)
Alphavirus/immunology , Genetic Vectors/immunology , Influenza Vaccines/immunology , Vaccination/methods , Alphavirus/genetics , Animals , Antibodies, Viral/blood , Drug Carriers , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/mortality , Survival Analysis
9.
Clin Vaccine Immunol ; 16(4): 471-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19193829

ABSTRACT

Seasonal influenza virus infections cause considerable morbidity and mortality in the world, and there is a serious threat of a pandemic influenza with the potential to cause millions of deaths. Therefore, practical influenza vaccines and vaccination strategies that can confer protection against intranasal infection with influenza viruses are needed. In this study, we demonstrate that using LTK63, a nontoxic mutant of the heat-labile toxin from Escherichia coli, as an adjuvant for both mucosal and systemic immunizations, systemic (intramuscular) immunization or combinations of mucosal (intranasal) and intramuscular immunizations protected mice against intranasal challenge with a lethal dose of live influenza virus at 3.5 months after the second immunization.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Toxins/administration & dosage , Enterotoxins/administration & dosage , Escherichia coli Proteins/administration & dosage , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Bacterial Toxins/pharmacology , Body Weight , Cytokines/blood , Enterotoxins/pharmacology , Enzyme-Linked Immunosorbent Assay , Escherichia coli Proteins/pharmacology , Female , Hemagglutination Inhibition Tests , Immunization/methods , Immunoglobulin A/blood , Immunoglobulin G/blood , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Survival Analysis
10.
Proc Natl Acad Sci U S A ; 104(10): 3984-9, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17360464

ABSTRACT

Invariant natural killer T (iNKT) cells are innate-like lymphocytes recognizing CD1d-restricted glycolipid antigens, such as alpha-galactosylceramide (alphaGC). We assessed whether iNKT cells help B lymphocyte responses and found that mice immunized with proteins and alphaGC develop antibody titers 1-2 logs higher than those induced by proteins alone. Activation of iNKT cells enhances protection against infections such as influenza and elicits higher frequencies of memory B cells and higher antibody responses to booster immunizations. Protein vaccination with alphaGC, but not with conventional adjuvants, elicits IgG responses in mice lacking MHC class II molecules, demonstrating that iNKT cells can substitute for CD4(+) T cell help to B cells. Interestingly, the decay of circulating antibodies is faster in mice lacking iNKT cells. These findings point to a homeostatic role for iNKT cells on critical features of the antibody response such as immunity and B cell memory.


Subject(s)
B-Lymphocytes/metabolism , Immunologic Memory , Killer Cells, Natural/metabolism , Animals , Antigens, CD1/metabolism , Antigens, CD1d , B-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Galactosylceramides/metabolism , Immune System , Killer Cells, Natural/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Time Factors
11.
Vaccine ; 25(3): 481-9, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17052811

ABSTRACT

Parainfluenza virus type 3 (PIV3) infections continue to be a significant health risk for infants, young children, and immunocompromised adults. We describe a gene-based vaccine strategy against PIV3 using replication-defective alphavirus vectors. These RNA replicon vectors, delivered as virus-like particles and expressing the PIV3 hemagglutinin-neuraminidase glycoprotein, were shown to be highly immunogenic in mice and hamsters, inducing PIV3-specific neutralizing antibody responses. Importantly, the replicon particle-based vaccine administered intramuscularly or intranasally protected against mucosal PIV3 challenge in hamsters, preventing virus replication in both nasal turbinates and lungs. These data suggest that the alphavirus replicon platform can be useful for a PIV3 vaccine and possibly other respiratory viruses.


Subject(s)
Alphavirus/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , RNA, Viral/genetics , RNA, Viral/immunology , Replicon/genetics , Replicon/immunology , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Cricetinae , Encephalitis Virus, Venezuelan Equine/immunology , Enzyme-Linked Immunosorbent Assay , Female , Hemagglutination Inhibition Tests , Mesocricetus , Mice , Mice, Inbred BALB C , Neutralization Tests , Parainfluenza Virus 3, Human/growth & development , Sindbis Virus/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
12.
J Virol ; 78(19): 10328-35, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367599

ABSTRACT

We have expressed and characterized the severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein in cDNA-transfected mammalian cells. The full-length spike protein (S) was newly synthesized as an endoglycosidase H (endo H)-sensitive glycoprotein (gp170) that is further modified into an endo H-resistant glycoprotein (gp180) in the Golgi apparatus. No substantial proteolytic cleavage of S was observed, suggesting that S is not processed into head (S1) and stalk (S2) domains as observed for certain other coronaviruses. While the expressed full-length S glycoprotein was exclusively cell associated, a truncation of S by excluding the C-terminal transmembrane and cytoplasmic tail domains resulted in the expression of an endoplasmic reticulum-localized glycoprotein (gp160) as well as a Golgi-specific form (gp170) which was ultimately secreted into the cell culture medium. Chemical cross-linking, thermal denaturation, and size fractionation analyses suggested that the full-length S glycoprotein of SARS-CoV forms a higher order structure of approximately 500 kDa, which is consistent with it being an S homotrimer. The latter was also observed in purified virions. The intracellular form of the C-terminally truncated S protein (but not the secreted form) also forms trimers, but with much less efficiency than full-length S. Deglycosylation of the full-length homotrimer with peptide N-glycosidase-F under native conditions abolished recognition of the protein by virus-neutralizing antisera raised against purified virions, suggesting the importance of the carbohydrate in the correct folding of the S protein. These data should aid in the design of recombinant vaccine antigens to prevent the spread of this emerging pathogen.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Animals , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Culture Media/chemistry , DNA, Complementary , DNA, Viral/genetics , DNA, Viral/metabolism , Endoplasmic Reticulum/chemistry , Glycoside Hydrolases/metabolism , Golgi Apparatus/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Molecular Weight , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Protein Folding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Subunits/analysis , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
13.
Nat Med ; 10(8): 871-5, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15247913

ABSTRACT

Passive serotherapy can confer immediate protection against microbial infection, but methods to rapidly generate human neutralizing monoclonal antibodies are not yet available. We have developed an improved method for Epstein-Barr virus transformation of human B cells. We used this method to analyze the memory repertoire of a patient who recovered from severe acute respiratory syndrome coronavirus (SARS-CoV) infection and to isolate monoclonal antibodies specific for different viral proteins, including 35 antibodies with in vitro neutralizing activity ranging from 10(-8)M to 10(-11)M. One such antibody confers protection in vivo in a mouse model of SARS-CoV infection. These results show that it is possible to interrogate the memory repertoire of immune donors to rapidly and efficiently isolate neutralizing antibodies that have been selected in the course of natural infection.


Subject(s)
Antibodies, Monoclonal/isolation & purification , B-Lymphocytes/immunology , Immunization, Passive/methods , Immunologic Memory/immunology , Severe Acute Respiratory Syndrome/immunology , Adult , Animals , Chlorocebus aethiops , DNA Primers , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Herpesvirus 4, Human/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Microscopy, Immunoelectron , Neutralization Tests , Polymerase Chain Reaction , Spike Glycoprotein, Coronavirus , Transformation, Genetic , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , alpha-Macroglobulins/metabolism
14.
Virology ; 304(2): 187-96, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12504561

ABSTRACT

We previously demonstrated that hepatitis C virus (HCV) binds to human CD81 through the E2 glycoprotein. Therefore, expression of the human CD81 molecule in transgenic mice was expected to provide a new tool to study HCV infection in vivo, as the chimpanzee is the only species currently available as a laboratory animal model that can be infected with HCV. We produced transgenic mice expressing the human CD81 protein in a wide variety of tissues. We confirmed binding of recombinant E2 glycoprotein to the liver tissue as well as to thymocytes and splenic lymphocytes in the transgenic mice. We inoculated chimpanzee plasma infected with HCV into these animals. None of these transgenic animals showed evidence of viral replication. Furthermore, human CD81 transgenic mice that lack expression of endogenous mouse CD81 were also resistant to HCV infection. We conclude that expression of human CD81 alone is insufficient to confer susceptibility to HCV infection in the mouse. The presence of additional possible factors for HCV infection is discussed.


Subject(s)
Antigens, CD/physiology , Hepatitis C/etiology , Membrane Proteins/physiology , Animals , Disease Susceptibility , Humans , Mice , Mice, Transgenic , Tetraspanin 28 , Viral Envelope Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...