Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
1.
Anticancer Drugs ; 29(3): 197-207, 2018 03.
Article in English | MEDLINE | ID: mdl-29438228

ABSTRACT

Newcastle disease virus (NDV) can specifically kill cancer cells and has less toxicity to normal cells. The hemagglutinin-neuraminidase (HN) protein is an important structural protein in NDV pathogenesis and has been postulated as a promising candidate for antitumor therapy. The aim of this study was to investigate the anticancer potential of recombinant adenovirus Ad-HN-PEG3p-E1a. An MTS assay was performed to determine viral proliferation after viral infection, the data showed that the proliferation ability of osteosarcoma cells decreased, whereas there was no significant change in normal hepatic cells. DAPI and Annexin V experiments showed that osteosarcoma cells were killed because of apoptosis, active oxygen content, and augmented mitochondrial membrane potential loss. Caspase Activity Assay Kits were used to detect the caspase-3 activities of the treated OS-732 for increased expression. Western blot analysis showed that cytochrome C increased significantly and apoptosis of the virus was confirmed in tumor cells. In-vivo experiments show that NDV has an inhibitory effect on tumor growth. The recombinant adenovirus, which is composed of a HN protein and progressive increment promoter PEG3p, could inhibit the growth of OS-732 and promote the apoptosis of tumor cells. However, there was no clear relationship with normal cell (L02) apoptosis.


Subject(s)
Bone Neoplasms/therapy , Hemagglutinins, Viral/genetics , Neuraminidase/genetics , Newcastle disease virus/genetics , Oncolytic Virotherapy/methods , Osteosarcoma/therapy , Animals , Apoptosis/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/virology , Cell Line, Tumor , Female , HEK293 Cells , Hemagglutinins, Viral/biosynthesis , Humans , Mice , Neuraminidase/biosynthesis , Newcastle disease virus/enzymology , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/virology , Random Allocation
2.
PLoS One ; 7(7): e41332, 2012.
Article in English | MEDLINE | ID: mdl-22859976

ABSTRACT

The evolution of the H5N1 highly pathogenic avian influenza virus (HPAIV) has resulted in high sequence variations and diverse antigenic properties in circulating viral isolates. We investigated immune responses induced by HA DNA vaccines of two contemporary H5N1 HPAIV isolates, A/bar-headed goose/Qinghai/3/2005 (QH) and A/chicken/Shanxi/2/2006 (SX) respectively, against the homologous as well as the heterologous virus isolate for comparison. Characterization of antibody responses induced by immunization with QH-HA and SX-HA DNA vaccines showed that the two isolates are antigenically distinctive. Interestingly, after immunization with the QH-HA DNA vaccine, subsequent boosting with the SX-HA DNA vaccine significantly augmented antibody responses against the QH isolate but only induced low levels of antibody responses against the SX isolate. Conversely, after immunization with the SX-HA DNA vaccine, subsequent boosting with the QH-HA DNA vaccine significantly augmented antibody responses against the SX isolate but only induced low levels of antibody responses against the QH isolate. In contrast to the antibody responses, cross-reactive T cell responses are readily detected between these two isolates at similar levels. These results indicate the existence of original antigenic sin (OAS) between concurrently circulating H5N1 HPAIV strains, which may need to be taken into consideration in vaccine development against the potential H5N1 HPAIV pandemic.


Subject(s)
Adaptive Immunity , Antigens, Viral/immunology , Hemagglutinins, Viral/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/genetics , Influenza in Birds/prevention & control , Vaccination , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Chickens , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Female , Geese , HeLa Cells , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Humans , Immunization, Secondary , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza in Birds/immunology , Influenza in Birds/virology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology
3.
J Virol ; 85(3): 1370-83, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21106745

ABSTRACT

Increasing levels of plasmid vector-mediated activation of innate immune signaling pathways is an approach to improve DNA vaccine-induced adaptive immunity for infectious disease and cancer applications. Retinoic acid-inducible gene I (RIG-I) is a critical cytoplasmic double-stranded RNA (dsRNA) pattern receptor required for innate immune activation in response to viral infection. Activation of RIG-I leads to type I interferon (IFN) and inflammatory cytokine production through interferon promoter stimulator 1 (IPS-1)-mediated activation of interferon regulatory factor 3 (IRF3) and NF-κB signaling. DNA vaccines coexpressing antigen and an expressed RNA (eRNA) RIG-I agonist were made, and the effect of RIG-I activation on antigen-specific immune responses to the encoded antigen was determined. Plasmid vector backbones expressing various RIG-I ligands from RNA polymerase III promoters were screened in a cell culture assay for RIG-I agonist activity, and optimized, potent RIG-I ligands were developed. One of these, eRNA41H, combines (i) eRNA11a, an immunostimulatory dsRNA expressed by convergent transcription, with (ii) adenovirus VA RNAI. eRNA41H was integrated into the backbone of DNA vaccine vectors expressing H5N1 influenza virus hemagglutinin (HA). The resultant eRNA vectors potently induced type 1 IFN production in cell culture through RIG-I activation and combined high-level HA antigen expression with RNA-mediated type I IFN activation in a single plasmid vector. The eRNA vectors induced increased HA-specific serum antibody binding avidity after naked DNA intramuscular prime and boost delivery in mice. This demonstrates that DNA vaccine potency may be augmented by the incorporation of RIG-I-activating immunostimulatory RNA into the vector backbone.


Subject(s)
Antibodies, Viral/blood , DEAD-box RNA Helicases/immunology , Influenza Vaccines/immunology , RNA, Double-Stranded/immunology , Vaccines, DNA/immunology , Adenoviridae/genetics , Animals , DEAD Box Protein 58 , Hemagglutinins, Viral/biosynthesis , Immunity, Humoral , Immunization, Secondary/methods , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Injections, Intramuscular , Interferon Type I/biosynthesis , Mice , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Double-Stranded/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
4.
Sheng Wu Gong Cheng Xue Bao ; 25(5): 773-8, 2009 May.
Article in Chinese | MEDLINE | ID: mdl-19670649

ABSTRACT

We produced high pathogenic avian influenza H5N1 haemagglutinin protein HA1 in recombinant Pichia pastoris in a 10 L fermentor, to establish a high-density cell fermentation method. We studied the effects of different factors such as culture temperature, induced temperature, methanol feeding methods, trace elements on the growth of Pichia pastoris, the yield and the biologic activity of recombinant HA1 protein. The culture temperature in pre-induced and induced stage were optimized at 25 degrees C to adapt cell growth and recombinant protein expression, and induced temperature at 25 degrees C also resulted in higher biologic activity of rHA1 than at 30 degrees C. The binding activity of rHA1 against a wide-spectrum neutralizing antibody was susceptible to the presence of any trace elements, although trace elements would essentially benefit for the cell fermentation. As a conclusion, the expression level of rHA1 produced with optimized fermentation process reached 120 mg/L, which was 10.5 times higher than the one produced in regular shaking flask. The resultant high-density cell fermentation can likely produce rHA1 of H5N1 in large scale.


Subject(s)
Fermentation , Hemagglutinins, Viral/biosynthesis , Influenza A Virus, H5N1 Subtype/genetics , Pichia/metabolism , Recombinant Proteins/biosynthesis , Hemagglutinins, Viral/genetics , Influenza A Virus, H5N1 Subtype/metabolism , Pichia/genetics
5.
J Am Chem Soc ; 130(38): 12568-9, 2008 Sep 24.
Article in English | MEDLINE | ID: mdl-18759389

ABSTRACT

Inclusion bodies are insoluble aggregates that are formed by bacteria to store excess recombinant protein produced during expression. The structure of the protein in inclusion bodies is poorly understood but it has been hypothesized that the protein may form misfolded beta sheet aggregates. This paper presents an isotopic labeling and solid-state nuclear magnetic resonance approach to determine the secondary structure of individual residues within a recombinant influenza virus "FHA2" protein in inclusion bodies. The inclusion bodies were studied either in the context of the unlysed hydrated E. coli cells or in the hydrated pellet formed from centrifugation of the material insoluble in the cell lysate. The native structure of FHA2 is predominantly helical and native helical structure was also observed for several specific residues in the inclusion body FHA2. This approach will be applicable to structural analysis of many inclusion body proteins and should provide useful information for optimizing solubilization and purification protocols of these proteins.


Subject(s)
Hemagglutinins, Viral/chemistry , Inclusion Bodies/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Carbon Isotopes , Escherichia coli/genetics , Escherichia coli/metabolism , Hemagglutinins, Viral/analysis , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Inclusion Bodies/metabolism , Protein Structure, Secondary , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
6.
Protein Expr Purif ; 62(2): 206-15, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18799134

ABSTRACT

This study presents the heterologous production and purification of a soluble and functional form of the hemagglutinin esterase (HE) of the infectious salmon anemia virus (ISAV) isolate 4 (Glesvaer/2/90). The HE possesses receptor binding and receptor destroying enzyme (RDE) activity and is probably involved in the infection process. The recombinant HE protein (recHE 4) was expressed in insect cells (Sf9) using the baculovirus expression vector system. Both the transmembrane region and the cytoplasmic tail were deleted, and a C-terminal His(6)-tag was attached to facilitate identification and purification of the recHE 4 protein. As determined by Western analysis the recHE 4 was secreted at 20 degrees C and not at 28 degrees C. By testing three HE constructs differing in their promoter and secretion signal sequences it was clear that the HE's own secretion signal sequence is more important than the promoter with respect to the amount of secreted recHE 4 obtained under the conditions used. A one-step purification by nickel-affinity chromatography resulted in a highly purified recHE 4, identified by matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS) analysis. Also, the recHE 4 is glycosylated and contains disulfide bridges within the molecule. Functional studies including the verification of the receptor destroying enzyme (RDE) activity as well as the binding to Atlantic salmon erythrocytes (hemagglutination) indicate that the recHE 4 has similar functions as its native counterpart. In conclusion, insect cells secrete a functional form of the ISAV 4 HE. This is suitable for further analyses on its function and immunogenicity.


Subject(s)
Baculoviridae/metabolism , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/isolation & purification , Isavirus/enzymology , Viral Fusion Proteins/biosynthesis , Viral Fusion Proteins/isolation & purification , Acetylesterase/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Enzyme Stability , Erythrocytes/cytology , Glycosylation , Hemagglutination , Hemagglutinins, Viral/chemistry , Insecta , Molecular Sequence Data , Protein Denaturation , Salmo salar/virology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism
7.
J Immunol ; 180(4): 2149-57, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250421

ABSTRACT

We have used TCR transgenic mice directed to different MHC class II-restricted determinants from the influenza virus hemagglutinin (HA) to analyze how specificity for self-peptides can shape CD4+CD25+ regulatory T (Treg) cell formation. We show that substantial increases in the number of CD4+CD25+ Treg cells can occur when an autoreactive TCR directed to a major I-E(d)-restricted determinant from HA develops in mice expressing HA as a self-Ag, and that the efficiency of this process is largely unaffected by the ability to coexpress additional TCR alpha-chains. This increased formation of CD4+CD25+ Treg cells in the presence of the self-peptide argues against models that postulate selective survival rather than induced formation as mechanisms of CD4+CD25+ Treg cell formation. In contrast, T cells bearing a TCR directed to a major I-A(d)-restricted determinant from HA underwent little or no selection to become CD4+CD25+ Treg cells in mice expressing HA as a self-Ag, correlating with inefficient processing and presentation of the peptide from the neo-self-HA polypeptide. These findings show that interactions with a self-peptide can induce thymocytes to differentiate along a pathway to become CD4+CD25+ Treg cells, and that peptide editing by DM molecules may help bias the CD4+CD25+ Treg cell repertoire away from self-peptides that associate weakly with MHC class II molecules.


Subject(s)
Antigen Presentation/immunology , Autoantigens/immunology , Autoantigens/metabolism , Cell Differentiation/immunology , Peptides/immunology , Peptides/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antigen Presentation/genetics , Autoantigens/biosynthesis , Autoantigens/genetics , Cell Differentiation/genetics , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Hemagglutinins, Viral/metabolism , Influenza A virus/immunology , L Cells , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory/cytology
8.
Viral Immunol ; 20(3): 441-52, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17931114

ABSTRACT

We have prepared a virus-like particle (VLP) vaccine bearing the surface glycoproteins HA and NA of the 1918 influenza A virus by infecting Sf9 cells with a quadruple recombinant baculovirus that expresses the four influenza proteins (HA, NA, M1, and M2) required for the assembly and budding of the VLPs. The presence of HA and M1 in the purified VLPs was confirmed by Western blot, and that of NA by a neuraminidase enzymatic assay. For in vivo studies, the 1918 VLP vaccine was formulated with or without an oligonucleotide containing two CpG motifs and administered in two doses 2 wk apart via the intranasal route. The antibody titers in mice immunized with VLP vaccines were higher than in mice vaccinated with an inactivated swine virus (H1N1) control, when CHO cells expressing 1918 HA were used as antigen. The opposite result was obtained when disrupted swine virus was the antigen for the ELISA test. Vaccine efficacy was evaluated by challenging immunized mice with the 1918 antigenically related influenza virus A/swine/Iowa/15/30 (H1N1) and measuring viral titers in the upper and lower respiratory tract. Mice immunized with VLP vaccine plus CpG demonstrated significantly lower viral titers in the nose and lungs than did the control on days 2 and 4 postchallenge and completely cleared the virus by day 6. Furthermore, they did not show symptoms of disease although there was a minor decrease in body weight. Mice vaccinated with VLP alone also demonstrated significantly lower viral titers in the nose and lungs than did the placebo group as well as the inactivated virus group on days 4 and 6 postchallenge. These results suggest that it is feasible to make a safe and immunogenic vaccine to protect against the extremely virulent 1918 virus, using a novel and safe cell-based technology.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Vaccines, Virosome/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/blood , Body Weight , Female , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Humans , Influenza, Human/immunology , Influenza, Human/virology , Lung/virology , Mice , Mice, Inbred BALB C , Neuraminidase/biosynthesis , Neuraminidase/genetics , Nose/virology , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Placebos , Viral Matrix Proteins/biosynthesis , Viral Matrix Proteins/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics , Viral Vaccines/immunology
9.
Antibiot Khimioter ; 52(11-12): 18-20, 2007.
Article in Russian | MEDLINE | ID: mdl-19275052

ABSTRACT

Analysis of triazavirin efficacy with respect to influenza A virus (H5N1) in sensitive cell culture MDSK vs. effective antigrippe drugs, such as tamiflu, remantadin and arbidol showed that triazavirin in a wide range of the concentrations was efficient in inhibition of the virus cytopathic activity and formation of the specific hemagglutinin.


Subject(s)
Antiviral Agents/pharmacology , Azoles/pharmacology , Influenza A Virus, H5N1 Subtype/drug effects , Triazines/pharmacology , Animals , Cell Line , Dogs , Hemagglutinins, Viral/biosynthesis , Triazoles
10.
Biochemistry ; 43(19): 5902-11, 2004 May 18.
Article in English | MEDLINE | ID: mdl-15134464

ABSTRACT

The influenza virus uses hemagglutinin (HA) to fuse the viral and cellular membranes. As part of an effort to study the membrane-interacting elements of HA, the fusion peptide, and the C-terminal transmembrane anchor, we have expressed in Escherichia coli the full-length HA(2) chain with maltose-binding protein fused at its N-terminus. The chimeric protein can be refolded in vitro in the presence of specific detergents to yield stable, homogeneous trimers, as determined by analytical ultracentrifugation. The trimers have the so-called "low pH" conformation-the rearranged HA(2) conformation obtained when intact HA(1)/HA(2) is induced to refold by exposure to low pH-as detected by electron microscopy and monoclonalantibody reactivity. These results provide further evidence for the notion that the neutral-pH structure of intact HA is metastable and that binding of protons lowers the kinetic barriers that prevent rearrangement to the minimum-free-energy conformation. The refolded chimeric protein described here is a suitable species for undertaking studies of how the fusion peptide inserts into membranes and assessing the nature of possible intermediates in the fusion process.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinins, Viral/chemistry , Protein Folding , Amino Acid Sequence , Carrier Proteins/chemical synthesis , Carrier Proteins/genetics , Carrier Proteins/ultrastructure , Detergents/chemistry , Enzyme-Linked Immunosorbent Assay , Escherichia coli Proteins/chemical synthesis , Escherichia coli Proteins/genetics , Escherichia coli Proteins/ultrastructure , Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/ultrastructure , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/ultrastructure , Hydrogen-Ion Concentration , Maltose-Binding Proteins , Molecular Sequence Data , Protein Conformation , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/ultrastructure , Solubility , Structure-Activity Relationship , Ultracentrifugation
11.
Virology ; 313(2): 502-13, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12954217

ABSTRACT

To enhance mucosal immune responses using simian/human immunodeficiency virus-like particles (SHIV VLPs), we have produced novel phenotypically mixed chimeric influenza HA/SHIV VLPs and used them to immunize C57BL/6J mice intranasally. Antibody and cytotoxic T-cell (CTL) responses as well as cytokine production in both systemic and mucosal sites were compared after immunization with SHIV VLPs or chimeric HA/SHIV VLPs. By using enzyme-linked immunosorbent assay (ELISA), the levels of serum IgG and mucosal IgA to the HIV envelope protein (Env) were found to be highest in the group immunized with chimeric HA/SHIV VLPs. Furthermore, the highest titer of serum neutralizing antibody against HIV Env was found with the group immunized with chimeric HA/SHIV VLPs. Analysis of the IgG1/IgG2a ratio indicated that a T(H)1-oriented immune response resulted from these VLP immunizations. HA/SHIV VLP-immunized mice also showed significantly higher CTL responses than those observed in SHIV VLP-immunized mice. Moreover, a MHC class I restricted T-cell activation ELISPOT assay showed a mixed type of T(H)1/T(H)2 cytokines in the HA/SHIV VLP-immunized mice, indicating that the chimeric VLPs can enhance both humoral and cellular immune responses to the HIV Env protein at multiple mucosal and systemic sites. The results indicate that incorporation of influenza HA into heterotypic VLPs may be highly effective for targeting vaccines to mucosal surfaces.


Subject(s)
Antibodies, Viral/analysis , HIV/immunology , Hemagglutinins, Viral/immunology , Influenza, Human/immunology , Reassortant Viruses , Simian Immunodeficiency Virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line , Cells, Cultured , Cytokines/analysis , Dose-Response Relationship, Immunologic , Female , Gene Products, gag/biosynthesis , Gene Products, gag/immunology , HIV/chemistry , Hemagglutinins, Viral/biosynthesis , Humans , Immunity, Mucosal , Immunization , Immunoglobulin G/blood , Influenza, Human/blood , Mice , Mice, Inbred C57BL , Neutralization Tests , Reassortant Viruses/immunology , Recombinant Proteins/biosynthesis , Simian Immunodeficiency Virus/chemistry , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/immunology
12.
J Immunol ; 169(7): 3622-9, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12244153

ABSTRACT

It has long been established that exposure of naive T cells to specific Ag in the absence of adjuvant leads to tolerization. Nonetheless, the potential of effector CD4 cells to be tolerized has been less well characterized. To address this issue, we have used an adoptive transfer system in which naive TCR transgenic hemagglutinin (HA)-specific CD4(+) T cells are initially primed to express effector function upon exposure to an immunogenic recombinant vaccinia virus expressing HA, and then exposed to forms of HA that are tolerogenic for naive CD4 cells. HA-specific effector CD4 cells residing in both the spleen as well as in two separate nonlymphoid tissues were tolerized upon exposure to high doses of exogenous soluble HA peptide. Additionally, tolerance could also be induced by bone marrow-derived APCs that cross-present parenchymally derived self-HA. Thus, effector CD4 cells are susceptible to similar tolerogenic stimuli as are naive CD4 cells.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Immune Tolerance/genetics , Interphase/immunology , Liver/cytology , Liver/immunology , Liver/virology , Lung/cytology , Lung/immunology , Lung/virology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/administration & dosage , Oligopeptides/immunology , Solubility , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Vaccinia virus/genetics , Vaccinia virus/immunology
13.
Biochim Biophys Acta ; 1565(1): 81-9, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12225855

ABSTRACT

X-ray studies show that influenza hemagglutinin (HA) forms an elongated structure connecting the influenza virus at one end to cell-surface receptors at the other. At neutral pH, the 20 N-terminal residues of HA2-referred to as the fusion peptide-are buried in a hydrophobic pocket, about 100 A away from the receptor-binding site, and thus seem unlikely to affect HA binding to the receptor. To test this assumption, we mutated residues in the fusion peptide, heterologically expressed the mutated proteins in COS7 cells, and examined their ability to bind fluorescently labeled red blood cells (RBCs). Surprisingly, a significantly reduced binding was recorded for some of the mutants. Ample experimental data indicate that HA has at least two forms: the native structure at neutral pH (N) that is metastable and the fusogenic form (F), observed at low pH, which is stable. Thus, a simple interpretation of our data is that HA can bind to its receptors at the RBC surface in the N form much more effectively than in the F (or in any other stable) form and that the altered binding properties are due to destabilizing effects of the mutations on the N form. That is, some of the mutations involve reduction in the free energy barrier between the N and F forms. This, in turn, leads to reduction in the population of the N form, which is the only form capable of binding to the cell-surface receptors. To explore this possibility, we estimated the stability free energy difference between HA wild-type (wt) and mutants in the N form using an empirical surface tension coefficient. The calculated stability differences correlated well with the measured binding, supporting the above interpretation. Our results are examined taking into account the available experimental data on the affinity of different soluble and membrane-attached forms of HA to its receptors.


Subject(s)
Hemagglutinins, Viral/immunology , Orthomyxoviridae/immunology , Receptors, Cell Surface/immunology , Animals , COS Cells/metabolism , Erythrocyte Membrane/immunology , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Hydrogen-Ion Concentration , Mutation , Orthomyxoviridae/metabolism , Protein Binding , Protein Conformation , Protein Isoforms/immunology , Thermodynamics , Time Factors , Transfection
14.
Microbes Infect ; 4(10): 1035-44, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12191653

ABSTRACT

A procedure described here allows the efficient and rapid purification of histidine-tagged measles virus haemagglutinin that is synthesized under the control of powerful promoters (PSFJ1-10 and PSFJ2-16) of the highly attenuated vaccinia virus (VV) strain LC16mO. A single affinity chromatography step purifies recombinant haemagglutinin proteins from the lysates of cells infected with the recombinant VVs. The recovery and purity are both very high (a yield of 0.5-2.8 mg/10(8) cells and purity of >94-98%), indicating that this procedure is approximately 400 times more efficient than the conventional methods used to prepare haemagglutinin. The haemagglutinins are correctly transported to the cell surface and have haemadsorption activity. Moreover, the recombinant haemagglutinin proteins cooperate with the measles virus fusion protein to elicit cell fusion activity. In addition, the antibody titres against measles virus, as measured by enzyme-linked immunosorbent assay using the purified haemagglutinin as the capture antigen, correlated closely with neutralization test titres (R(2) = 0.84, p < 0.05), indicating the preservation of immunologically relevant antigenicity. Such recombinant haemagglutinin preparations will be useful in diagnostic tests that measure functional anti-measles immunity and investigate the biological functions and structure of the haemagglutinin.


Subject(s)
Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/isolation & purification , Measles virus/genetics , Vaccinia virus/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Cricetinae , Enzyme-Linked Immunosorbent Assay , Gene Expression , Genetic Vectors/genetics , Hemagglutination Tests , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/chemistry , Humans , Molecular Sequence Data , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Viral Fusion Proteins/biosynthesis , Viral Fusion Proteins/genetics
15.
J Biochem ; 132(1): 83-91, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12097164

ABSTRACT

CD46, a complement regulatory protein widely expressed on human cells, serves as an entry receptor for measles virus (MV). We have previously shown that the expression of human CD46 in mouse macrophages restricts MV replication in these cells and enhances the production of nitric oxide (NO) in the presence of gamma interferon (IFN-gamma). In this study, we show that crosslinking human CD46 expressed on the mouse macrophage-like cell line RAW264.7 with purified C3b multimer but not monomer enhances NO production. The enhanced production of NO in response to IFN-gamma was observed again with C3b multimer but not monomer. The augmentation of NO production is human CD46-dependent with a CYT1>CYT2 profile. Thus, the reported MV-mediated NO production, irrespective of whether it is IFN-gamma-dependent or -independent, should be largely attributable to CD46 signaling but not to MV replication. Similar CYT1-dependent augmentation of NO production was reproducible with two CD46 ligating reagents, CD46-specific monoclonal antibodies (mAb) or their F(ab')(2) and MV hemagglutinin (H) and fusion (F) glycoproteins. Co-cultivation of mouse macrophages bearing human CD46 with Chinese hamster ovary (CHO) cells expressing MV H and F enhanced IFN-gamma-induced NO production. Yet, the NO levels induced by F(ab')(2) against CD46 or MV H/F on CHO cells were much lower than those induced by CD46-crosslinking mAb with Fc or MV infection. Removing the cytoplasmic tails of CD46 abrogated the augmentation of NO production triggered by all three stimulators. Thus, the CD46 CYT1 and CYT2 isoforms functionally diverge to elicit innate immune responses, which can be modulated by purified C3b multimer or anti-CD46 mAbs.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, CD/metabolism , Complement C3b/metabolism , Immunoglobulin Fab Fragments/metabolism , Interferon-gamma/pharmacology , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Nitric Oxide/biosynthesis , Animals , Antigens, CD/immunology , CHO Cells/metabolism , Cell Line , Complement C3b/immunology , Cricetinae , Hemagglutinins, Viral/biosynthesis , Humans , Immunoglobulin Fab Fragments/immunology , Macrophages/immunology , Measles virus/metabolism , Membrane Cofactor Protein , Membrane Glycoproteins/immunology , Mice , Protein Isoforms , Transfection
16.
EMBO J ; 21(5): 885-97, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11867517

ABSTRACT

Cell attachment and membrane penetration are functions of the rotavirus outer capsid spike protein, VP4. An activating tryptic cleavage of VP4 produces the N-terminal fragment, VP8*, which is the viral hemagglutinin and an important target of neutralizing antibodies. We have determined, by X-ray crystallography, the atomic structure of the VP8* core bound to sialic acid and, by NMR spectroscopy, the structure of the unliganded VP8* core. The domain has the beta-sandwich fold of the galectins, a family of sugar binding proteins. The surface corresponding to the galectin carbohydrate binding site is blocked, and rotavirus VP8* instead binds sialic acid in a shallow groove between its two beta-sheets. There appears to be a small induced fit on binding. The residues that contact sialic acid are conserved in sialic acid-dependent rotavirus strains. Neutralization escape mutations are widely distributed over the VP8* surface and cluster in four epitopes. From the fit of the VP8* core into the virion spikes, we propose that VP4 arose from the insertion of a host carbohydrate binding domain into a viral membrane interaction protein.


Subject(s)
Capsid Proteins , Capsid/chemistry , Carbohydrate Metabolism , N-Acetylneuraminic Acid/metabolism , Rotavirus/chemistry , Amino Acid Sequence , Binding Sites , Capsid/physiology , Consensus Sequence , Crystallography, X-Ray , Epitopes/chemistry , Evolution, Molecular , Galectins , Hemagglutinins/chemistry , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/chemistry , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Rotavirus/physiology , Structure-Activity Relationship , Substrate Specificity , Viral Nonstructural Proteins/biosynthesis , Viral Nonstructural Proteins/chemistry , Virion/chemistry , Virion/ultrastructure
17.
J Virol ; 76(3): 1505-9, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11773423

ABSTRACT

Measles has a host range restricted to humans and monkeys in captivity. Fresh measles virus (MV) isolates replicate readily in several human and simian B-cell lines but need a period of adaptation to other types of cells. The identification of CD46 and CD150 (SLAM) as cellular receptors for MV has helped to clarify certain aspects of the immunobiology of MV infections. We have examined the properties of an MV wild-type strain grown in the epithelial cell line Vero. After adaptation, this virus expressed high levels of both the viral glycoproteins (hemagglutinin and fusion protein) but did not induce fusion (syncytia). No changes in the amino acid sequence were found in either of the viral glycoproteins. Using several approaches, the Vero-adapted virus could not be shown to interact with CD46 either in the initiation or during the course of infection. The presence of human SLAM expressed in the Vero cells rapidly gave rise to fusion and lower yields of infectious virus.


Subject(s)
Adaptation, Physiological , Measles virus/physiology , Animals , Antigens, CD/metabolism , Cell Line , Chlorocebus aethiops , Glycoproteins/metabolism , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Humans , Immunoglobulins/metabolism , Measles virus/metabolism , Membrane Cofactor Protein , Membrane Glycoproteins/metabolism , Receptors, Cell Surface , Receptors, Virus/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Vero Cells , Viral Fusion Proteins/biosynthesis , Viral Fusion Proteins/genetics
18.
J Virol ; 76(2): 484-91, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752138

ABSTRACT

Rinderpest is an acute and highly contagious viral disease of ruminants, often resulting in greater than 90% mortality. We have constructed a recombinant vaccinia virus vaccine (v2RVFH) that expresses both the fusion (F) and hemagglutinin (H) genes of rinderpest virus (RPV) under strong synthetic vaccinia virus promoters. v2RVFH-infected cells express high levels of the F and H glycoproteins and show extensive syncytium formation. Cattle vaccinated intramuscularly with as little as 10(3) PFU of v2RVFH and challenged 1 month later with a lethal dose of RPV were completely protected from clinical disease; the 50% protective dose was determined to be 10(2) PFU. Animals vaccinated with v2RVFH did not develop pock lesions and did not transmit the recombinant vaccinia virus to contact animals. Intramuscular vaccination of cattle with 10(8) PFU of v2RVFH provided long-term sterilizing immunity against rinderpest. In addition to being highly safe and efficacious, v2RVFH is a heat-stable, inexpensive, and easily administered vaccine that allows the serological differentiation between vaccinated and naturally infected animals. Consequently, mass vaccination of cattle with v2RVFH could eradicate rinderpest.


Subject(s)
Cattle Diseases/immunology , Hemagglutinins, Viral/immunology , Rinderpest/immunology , Rinderpest/prevention & control , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Viral Vaccines/immunology , Animals , Cattle , Cattle Diseases/prevention & control , Cattle Diseases/virology , Cells, Cultured , Chlorocebus aethiops , Giant Cells/pathology , Giant Cells/virology , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Injections, Intramuscular , Morbillivirus/genetics , Morbillivirus/immunology , Neutralization Tests , Precipitin Tests , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Rinderpest/virology , Survival Rate , Time Factors , Vaccination , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vero Cells , Viral Proteins/biosynthesis , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/adverse effects , Viral Vaccines/genetics
19.
Intervirology ; 44(4): 209-14, 2001.
Article in English | MEDLINE | ID: mdl-11509881

ABSTRACT

The authors studied the effects of immunosuppressive peptide cyclosporin A (CsA) on cell fusion efficiency in cells persistently infected with measles virus (448-PI-Vero cells). Treatment of 448-PI-Vero cells with 5 microM CsA enhanced the infusion. In addition, the expression of measles virus antigen on cell surface was increased by treatment with CsA. The addition of phenothiazine, an anti-calmodulin drug, enhanced the fusion of 448-PI-Vero cells in the presence of CsA, although treatment with phenothiazine alone did not affect polykaryocyte formation. The enhancement of fusion efficiency in 448-PI-Vero cells by CsA was suppressed by oligopeptide Z-D-Phe-Phe-Gly, a synthetic oligopeptide that inhibits fusion induced by measles virus. Since the cell content of major virus-specific polypeptides, such as hemagglutinin, nucleoprotein or matrix protein is the same as in untreated controls, this fusion enhancement may be related to transport and accumulation of measles virus glycoproteins.


Subject(s)
Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Measles virus/drug effects , Vero Cells/drug effects , Animals , Antigens, Viral/analysis , Antiviral Agents/pharmacology , Cell Survival , Chlorocebus aethiops , Drug Synergism , Giant Cells/drug effects , Giant Cells/virology , Hemagglutinins, Viral/analysis , Hemagglutinins, Viral/biosynthesis , Oligopeptides/pharmacology , Phenothiazines/pharmacology , Virus Replication/drug effects
20.
J Gen Virol ; 82(Pt 7): 1757-1765, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11413388

ABSTRACT

Infectious salmon anaemia virus (ISAV) is an orthomyxo-like virus that causes serious disease in Atlantic salmon (Salmo salar). Like the orthomyxoviruses, ISAV has been shown to possess haemagglutinin (HA) activity. This study presents the cloning, expression and identification of the ISAV HA gene, which was isolated from a cDNA library by immunoscreening. The HA gene contained an ISAV-specific conserved nucleotide motif in the 5' region and a 1167 bp open reading frame encoding a protein with a predicted molecular mass of 42.4 kDa. The HA gene was expressed in a baculovirus system. A monoclonal antibody (MAb) shown previously to be directed against the ISAV HA reacted with insect cells infected with recombinant baculovirus. Salmon erythrocytes also adsorbed to these cells and adsorption was inhibited by the addition of either the ISAV-specific MAb or a polyclonal rabbit serum prepared against purified virus, confirming the virus specificity of the reaction. Immunoblot analyses indicated that ISAV HA, in contrast to influenza virus HA, is not posttranslationally cleaved. Sequence comparisons of the HA gene from five Norwegian, one Scottish and one Canadian isolate revealed a highly polymorphic region that may be useful in epidemiological studies.


Subject(s)
Anemia/veterinary , Fish Diseases/virology , Genes, Viral , Hemagglutinins, Viral/genetics , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae/genetics , Salmo salar , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Fishes , Fluorescent Antibody Technique , Hemagglutinins, Viral/biosynthesis , Molecular Sequence Data , Orthomyxoviridae/immunology , Recombinant Proteins/biosynthesis , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...