Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Infect Dis ; 215(5): 818-823, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28011910

ABSTRACT

Antibodies that mediate antibody-dependent cellular cytotoxicity (ADCC) against avian influenza virus subtypes, including H7N9 and H5N1, have been detected in human sera. Using NK cell activation and NK cytotoxicity assays, we compared ADCC-mediating antibodies (ADCC-Abs) in sera collected from healthy infants, children and adults against H7N9 virus-infected cells and recombinant hemagglutinin (HA), neuraminidase (NA), and nucleoprotein (NP) proteins. High titers of ADCC-Abs against H7N9 virus-infected cells were detected in sera from adults and children but not infants. ADCC-Abs titers directed against H7N9 HA or NA proteins. Further analysis showed that ADCC-Abs titers were significantly higher toward H7N9 NP, as compared with H7N9 HA or NA proteins, and correlated strongly with ADCC-Abs titers against H7N9 virus-infected cells. Indeed, ADCC-Abs to NPs of seasonal H1N1 and H3N2 viruses correlated strongly with ADCC-Abs to H7N9 NP, suggesting that seasonal influenza infections and vaccinations may induce these cross-reactive antibodies. Targeting ADCC-Abs to internal proteins may be a potential mechanism of universal vaccine design.


Subject(s)
Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity , RNA-Binding Proteins/immunology , Viral Core Proteins/immunology , Adolescent , Adult , Antibodies, Viral/blood , Child , Child, Preschool , Cross Reactions , Hemagglutinins/blood , Hemagglutinins/immunology , Humans , Infant , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza A Virus, H5N1 Subtype , Influenza A Virus, H7N9 Subtype , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Influenza, Human/prevention & control , Killer Cells, Natural/immunology , Middle Aged , Neuraminidase/blood , Neuraminidase/immunology , Nucleocapsid Proteins , RNA-Binding Proteins/blood , Recombinant Proteins/blood , Recombinant Proteins/immunology , Viral Core Proteins/blood , Young Adult
2.
J Infect Dis ; 212(7): 1052-60, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25795791

ABSTRACT

Human influenza is a highly contagious acute respiratory illness that is responsible for significant morbidity and excess mortality worldwide. In addition to neutralizing antibodies, there are antibodies that bind to influenza virus-infected cells and mediate lysis of the infected cells by natural killer (NK) cells (antibody-dependent cellular cytotoxicity [ADCC]) or complement (complement-dependent lysis [CDL]). We analyzed sera obtained from 16 healthy adults (18-63 years of age), 52 children (2-17 years of age), and 10 infants (0.75-1 year of age) in the United States, who were unlikely to have been exposed to the avian H7N9 subtype of influenza A virus, by ADCC and CDL assays. As expected, none of these sera had detectable levels of hemagglutination-inhibiting antibodies against the H7N9 virus, but we unexpectedly found high titers of ADCC antibodies to the H7N9 subtype virus in all sera from adults and children aged ≥8 years.


Subject(s)
Antibodies, Viral/blood , Antibody-Dependent Cell Cytotoxicity/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/immunology , Killer Cells, Natural/immunology , Adolescent , Adult , Child , Child, Preschool , Complement System Proteins/immunology , Hemagglutination Tests , Humans , Infant , Influenza, Human/prevention & control , Middle Aged , Young Adult
3.
J Virol ; 86(17): 9233-43, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22718815

ABSTRACT

The hemagglutinin protein (HA) of the influenza virus family is a major antigen for protective immunity. Thus, it is a relevant target for developing vaccines. Here, we describe a human CD4(+) T cell epitope in the influenza virus HA that lies in the fusion peptide of the HA. This epitope is well conserved in all 16 subtypes of the HA protein of influenza A virus and the HA protein of influenza B virus. By stimulating peripheral blood mononuclear cells (PBMCs) from a healthy adult donor with peptides covering the entire HA protein based on the sequence of A/Japan/305/1957 (H2N2), we generated a T cell line specific to this epitope. This CD4(+) T cell line recognizes target cells infected with influenza A virus seasonal H1N1 and H3N2 strains, a reassortant H2N1 strain, the 2009 pandemic H1N1 strain, and influenza B virus in cytotoxicity assays and intracellular-cytokine-staining assays. It also lysed target cells infected with avian H5N1 virus. We screened healthy adult PBMCs for T cell responses specific to this epitope and found individuals who had ex vivo gamma interferon (IFN-γ) responses to the peptide epitope in enzyme-linked immunospot (ELISPOT) assays. Almost all donors who responded to the epitope had the HLA-DRB1*09 allele, a relatively common HLA allele. Although natural infection or standard vaccination may not induce strong T and B cell responses to this highly conserved epitope in the fusion peptide, it may be possible to develop a vaccination strategy to induce these CD4(+) T cells, which are cross-reactive to both influenza A and B viruses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza B virus/immunology , Influenza, Human/immunology , Antibodies, Viral/immunology , Cells, Cultured , Cross Reactions , Epitopes, T-Lymphocyte/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H2N2 Subtype/genetics , Influenza A Virus, H2N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A virus/genetics , Influenza B virus/genetics , Influenza, Human/genetics , Influenza, Human/virology , Leukocytes, Mononuclear/immunology
4.
Virol J ; 10: 244, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23886073

ABSTRACT

Influenza A and B viruses form different genera, which were originally distinguished by antigenic differences in their nucleoproteins and matrix 1 proteins. Cross-protection between these two genera has not been observed in animal experiments, which is consistent with the low homology in viral proteins common to both viruses except for one of three polymerase proteins, polymerase basic 1 (PB1). Recently, however, antibody and CD4+ T cell epitopes conserved between the two genera were identified in humans. A protective antibody epitope was located in the stalk region of the surface glycoprotein, hemagglutinin, and a CD4+ T cell epitope was located in the fusion peptide of the hemagglutinin. The fusion peptide was also found to contain antibody epitopes in humans and animals. A short stretch of well-conserved peptide was also identified in the other surface glycoprotein, neuraminidase, and antibodies binding to this peptide were generated by peptide immunization in rabbits. Although PB1, the only protein which has relatively high overall sequence homology between influenza A and B viruses, is not considered an immunodominant protein in the T cell responses to influenza A virus infection, amino acid sequence comparisons show that a considerable number of previously identified T cell epitopes in the PB1 of influenza A viruses are conserved in the PB1 of influenza B viruses. These data indicate that B and T cell cross-reactivity exists between influenza A and B viruses, which may have modulatory effects on the disease process and recovery. Although the antibody titers and the specific T cell frequencies induced by natural infection or standard vaccination may not be high enough to provide cross protection in humans, it might be possible to develop immunization strategies to induce these cross-reactive responses more efficiently.


Subject(s)
Cross Reactions , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Influenza A virus/immunology , Influenza B virus/immunology , Viral Proteins/immunology , Humans
5.
Virus Genes ; 46(1): 20-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23001690

ABSTRACT

Vaccinia virus (VACV), a member of the Poxviridae family of large double-stranded DNA viruses, is being used as a smallpox vaccine as well as an expression vector for immunization against other infectious diseases and cancer. The host range of wild type VACV is very broad among mammalian cells. C7L is a host range gene identified in VACV and is well conserved in mammalian poxviruses except for parapoxviruses and molluscum contagiosum virus. The molecular mechanisms by which the C7L gene exerts host range function are not well understood. The C7L protein does not have any known conserved domains or show sequence similarity to cellular proteins or viral proteins other than the C7L homologs in mammalian poxviruses. We generated recombinant vaccinia viruses carrying deletion mutants of the C7L gene using NYVAC as a parental strain and found that the N-terminus is essential for host range function of C7L, which is consistent with a previous report that showed that homology among C7L homologs are greater near the N-terminus than the C-terminus.


Subject(s)
Host Specificity , Vaccinia virus/physiology , Viral Proteins/metabolism , DNA Mutational Analysis , Recombination, Genetic , Vaccinia virus/genetics , Viral Proteins/genetics
6.
J Virol ; 85(24): 13463-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21994454

ABSTRACT

We characterized human monoclonal antibodies (MAbs) cloned from influenza virus-infected patients and from influenza vaccine recipients by complement-dependent lysis (CDL) assay. Most MAbs active in CDL were neutralizing, but not all neutralizing MAbs can mediate CDL. Two of the three stalk-specific neutralizing MAbs tested were able to mediate CDL and were more cross-reactive to temporally distant H1N1 strains than the conventional hemagglutination-inhibiting and neutralizing MAbs. One of the stalk-specific MAbs was subtype cross-reactive to H1 and H2 hemagglutinins, suggesting a role for stalk-specific antibodies in protection against influenza illness, especially by a novel viral subtype which can cause pandemics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Complement System Proteins/immunology , Influenza A Virus, H1N1 Subtype/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Cell Death , Cross Reactions , Humans , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Molecular Sequence Data , Sequence Analysis, DNA
7.
J Infect Dis ; 203(4): 513-22, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21216868

ABSTRACT

ChimeriVax-WN02 is a novel live-attenuated West Nile virus (WNV) vaccine containing modified WNV premembrane (prM) and envelope (E) sequences inserted into the yellow fever 17D vaccine genome. We investigated the induction and evolution of CD8(+) T cell responses to a WNV envelope epitope, which is a dominant target in naturally infected HLA-A*02-positive individuals. WNV epitope-specific CD8(+) T cells were detected by HLA tetramer staining in 22 of 23 donors tested, with peak frequencies occurring between days 14 and 28. WNV epitope-specific T cells evolved from an effector phenotype to a long-lived memory phenotype. In the majority of donors, CD8(+) T cells were able to lyse targets expressing WNV envelope protein and produced macrophage inflammatory protein 1ß, interferon γ, and/or tumor necrosis factor α following envelope peptide stimulation. WNV E-specific CD8(+) T cell responses were detected for up to 1 year after vaccination. The evolution of this WNV-specific T cell response is similar to that observed in established, highly immunogenic vaccines.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Chemokine CCL4/metabolism , Epitopes, T-Lymphocyte/immunology , Human Experimentation , Humans , Immunologic Memory , Interferon-gamma/metabolism , Placebos/administration & dosage , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/metabolism , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Envelope Proteins/immunology , West Nile Virus Vaccines/administration & dosage
8.
J Exp Med ; 197(7): 927-32, 2003 Apr 07.
Article in English | MEDLINE | ID: mdl-12668642

ABSTRACT

Immunization with vaccinia virus resulted in long-lasting protection against smallpox and was the approach used to eliminate natural smallpox infections worldwide. Due to the concern about the potential use of smallpox virus as a bioweapon, smallpox vaccination is currently being reintroduced. Severe complications from vaccination were associated with congenital or acquired T cell deficiencies, but not with congenital agammaglobulinemia, suggesting the importance of T cell immunity in recovery from infection. In this report, we identified two CD8+ T cell epitopes restricted by the most common human major histocompatibility complex (MHC) class I allele, HLA-A*0201. Both epitopes are highly conserved in vaccinia and variola viruses. The frequency of vaccinia-specific CD8+ T cell responses to these epitopes measured by interferon (IFN)-gamma enzyme-linked immunospot (ELISPOT) assay and HLA/peptide tetramer staining peaked 2 wk after primary immunization and then declined, but were still detectable 1 to 3 yr after primary immunization. 2 wk after immunization, IFN-gamma-producing cells specific to these two epitopes were 14% of total vaccinia virus-specific IFN-gamma-producing cells in one donor, 35% in the second donor, and 6% in the third donor. This information will be useful for studies of human T cell memory and for the design and analyses of the immunogenicity of experimental vaccinia vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte , HLA-A Antigens/immunology , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Cell Line , Humans , Immunologic Memory , T-Lymphocytes, Cytotoxic/immunology
9.
J Exp Med ; 196(5): 579-88, 2002 Sep 02.
Article in English | MEDLINE | ID: mdl-12208874

ABSTRACT

Puumala virus (PUUV) is a hantavirus that causes hemorrhagic fever with renal syndrome (HFRS), which is an important public health problem in large parts of Europe. We examined the memory cytolytic T lymphocyte (CTL) responses in 13 Finnish individuals who had HFRS between 1984 and 1995. In seven of these donors, we detected virus-specific CTL responses against the PUUV nucleocapsid (N) protein after in vitro stimulation with PUUV. Six novel CD8(+) CTL epitopes were defined on the N protein and were found to be restricted by various HLA alleles including A2, A28, B7, and B8. This is the first demonstration of PUUV-specific CTL responses in humans, and the first identification of CTL epitopes on PUUV. In addition, this study provides one of the few characterizations of a human antiviral memory T cell response, without the complicating issues of virus persistence or reinfection. Interferon (IFN)-gamma ELISPOT analysis showed that memory CTL specific for these epitopes were present at high frequency in PUUV-immune individuals many years after acute infection in the absence of detectable viral RNA. The frequencies of PUUV-specific CTL were comparable to or exceeded those found in other viral systems including influenza, EBV and HIV, in which CTL responses may be boosted by periodic reinfection or virus persistence.


Subject(s)
Hemorrhagic Fever with Renal Syndrome/immunology , Immunologic Memory , Puumala virus , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigens, Viral/genetics , Epitopes/genetics , Finland , Hemorrhagic Fever with Renal Syndrome/virology , Humans , In Vitro Techniques , Molecular Sequence Data , Nucleocapsid/genetics , Nucleocapsid/immunology , Nucleocapsid Proteins , Puumala virus/genetics , Puumala virus/immunology , Time Factors
10.
Cell Immunol ; 264(1): 71-7, 2010.
Article in English | MEDLINE | ID: mdl-20483407

ABSTRACT

The major gammadelta T cell subset in the human peripheral blood expresses the Vgamma9delta2 TCR and recognizes non-peptidic prenyl pyrophosphate antigens such as isopentylpyrophosphate (IPP). Upon activation the gammadelta T cells rapidly secrete antiviral cytokines similar to classical memory alphabeta T cells. Here we have investigated the ability of gammadelta T lymphocytes from human PBMC to become activated by influenza A virus infection. Vgamma9Vdelta2 T lymphocytes rapidly upregulate expression of CD25 and CD69 and produce IFN-gamma following influenza infection of PBMC. Moreover, the recognition is cross-reactive between various subtypes of influenza, but not with vaccinia virus. Vgamma9Vdelta2 T cell responses are potently reduced by the HMG-CoA reductase inhibitor mevastatin, which inhibits the mevalonate pathway and IPP synthesis. Our results indicate that influenza virus infection induces the rapid activation and function of Vgamma9Vdelta2 T lymphocytes in the peripheral blood via a mechanism that depends on the mevalonate pathway.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Influenza A virus/immunology , Influenza, Human/immunology , Lovastatin/analogs & derivatives , T-Lymphocytes/drug effects , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , Cross Reactions , Humans , Indoles/immunology , Indoles/metabolism , Influenza A virus/pathogenicity , Influenza, Human/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Lovastatin/pharmacology , Lymphocyte Activation/drug effects , Mevalonic Acid/metabolism , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Species Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology
11.
J Virol ; 82(18): 9283-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18614638

ABSTRACT

Among 17 HLA-A2-positive healthy adults, CD8+ T-cell responses against an HLA-A2-restricted matrix protein 1 (M1) epitope increased after immunization with trivalent inactivated influenza vaccine (TIV) in two individuals. The presence of M1 in TIV was confirmed by Western blotting. T-cell cytotoxicity assays showed that TIV is processed and the epitope is presented by antigen-presenting cells to an M1 epitope-specific CD8+ T-cell line for specific lysis. These data show that TIV, which is formulated to contain surface glycoproteins to induce serotype-specific antibody responses, also contains M1, capable of inducing subtype cross-reactive CD8+ T-cell responses in some vaccinees.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Vaccines, Inactivated/immunology , Viral Matrix Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/metabolism , Humans , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Vaccines, Inactivated/administration & dosage
13.
Hum Immunol ; 69(12): 815-25, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18955096

ABSTRACT

Immunization with vaccinia virus (VACV) resulted in long-lasting protection against smallpox and successful global eradication of the disease. VACV elicits strong cellular and humoral immune responses. Although neutralizing antibody is essential for protection, cellular immunity seems to be more important for recovery from infection in humans. We analyzed the immunodominance hierarchy of 73 previously identified VACV human CD8(+) T-cell epitopes restricted by HLA-A1, -A2, -A3, -A24, -B7, or -B44 alleles or the alleles belonging to one of these supertypes in 56 donors after primary VACV immunization. With the exception of the responses to HLA-A24 supertype-restricted epitopes, there were no consistent patterns of epitope immunodominance among donors sharing the same HLA alleles or supertypes, which is in sharp contrast with the mouse studies. However, we identified 12 epitopes that were recognized by >or=20% of donors sharing the same HLA allele; 6 of these epitopes contributed >or=20% of the total VACV-specific T-cell response in at least one individual. VACV-specific CD8(+) T-cell responses targeted a group of epitopes, "relatively dominant" epitopes, without a strong immunodominance hierarchy in humans, which may be advantageous to humans to prevent the emergence of T-cell escape mutants.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunodominant Epitopes , Interferon-gamma/metabolism , Vaccinia virus/immunology , Vaccinia/immunology , Antigen Presentation , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Epitope Mapping , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , HLA-B Antigens/immunology , HLA-B Antigens/metabolism , Humans , Peptides/immunology , Smallpox Vaccine , Species Specificity , T-Cell Antigen Receptor Specificity , Vaccination
14.
Virus Res ; 123(2): 120-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16979772

ABSTRACT

Hantavirus infection causes two human diseases, hemorrhagic fever with renal syndrome and hantavirus pulmonary syndrome. The typical feature of these diseases is increased permeability in microvascular beds in the kidneys and the lungs, respectively. The mechanism of capillary leakage, however, is not understood. Some evidence suggests that hantavirus disease pathogenesis is immunologically mediated by cytotoxic T lymphocytes and other immune cells in target organs producing inflammatory cytokines. In this study we examined the roles of virus-specific cytotoxic T lymphocytes in increased permeability of human endothelial cells infected with hantavirus. We used a human CD8(+) hantavirus-specific cytotoxic T lymphocyte line, 1A-E2, specific for the HLA-A24-restricted epitope in Sin Nombre and Puumala virus G2 protein, and the human endothelial cell line, EA.hy926 that expresses HLA-A24 molecule. The cytotoxic T lymphocyte line recognized and lysed target cells infected with Sin Nombre virus, and in transwell permeability assays increased permeability of EA.hy926 cell monolayer infected with Sin Nombre virus or recombinant adenovirus expressing the Sin Nombre virus G2 protein. These results suggest that cytotoxic T lymphocyte activity contribute to capillary leakage observed in patients with hantavirus pulmonary syndrome or hemorrhagic fever with renal syndrome.


Subject(s)
Hantavirus Pulmonary Syndrome/physiopathology , Sin Nombre virus/immunology , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Cytotoxic/physiology , Cell Line , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/virology , HLA-A Antigens , Humans , Permeability , Species Specificity
15.
Immunol Lett ; 113(2): 117-20, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17897725

ABSTRACT

There are many viruses known to cause viral hemorrhagic fevers in humans. The mechanisms causing hemorrhage are likely to vary among viruses. Some viruses, such as Marburg virus, are directly cytopathic to infected endothelial cells, suggesting infection of endothelial cells alone can cause hemorrhage. On the other hand, there are viruses which infect endothelial cells without causing any cytopathic effects, suggesting the involvement of host immune responses in developing hemorrhage. Typical examples of these include viruses of the hantavirus species. We hypothesize that impairment of endothelial cell's defense mechanisms against cytotoxic CD8+ T cells is the mechanism of capillary leakage in hantavirus pulmonary syndrome and hemorrhagic fever with renal syndrome, which may be common to other viral hemorrhagic fevers. CD8+ T cells may be a potential target for therapy of some viral hemorrhagic fevers.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Capillary Permeability , Hantavirus Pulmonary Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/immunology , Animals , Endothelial Cells/immunology , Endothelial Cells/metabolism , Hantavirus Pulmonary Syndrome/physiopathology , Hemorrhagic Fever with Renal Syndrome/physiopathology , Humans , Mice
16.
Virol J ; 4: 22, 2007 Feb 27.
Article in English | MEDLINE | ID: mdl-17326843

ABSTRACT

BACKGROUND: Respiratory infection with the neurovirulent vaccinia virus (VV) strain Western Reserve (WR) results in an acute infection of the lung followed by dissemination of the virus to other organs and causes lethality in mice. The mechanisms of lethality are not well-understood. In this study, we analyzed virus replication and host immune responses after intranasal infection with lethal and non-lethal doses of VV using the WR strain and the less virulent Wyeth strain. RESULTS: The WR strain replicated more vigorously in the lung and in the brain than the Wyeth strain. There were, however, no differences between the virus titers in the brains of mice infected with the higher lethal dose and the lower non-lethal dose of WR strain, suggesting that the amount of virus replication in the brain is unlikely to be the sole determining factor of lethality. The WR strain grew better in primary mouse lung cells than the Wyeth strain. Lethal infection with WR strain was associated with a reduced number of lymphocytes and an altered phenotype of the T cells in the lung compared to non-lethal infections with the WR or Wyeth strains. Severe thymus atrophy with a reduction of CD4 and CD8 double positive T cells was also observed in the lethal infection. CONCLUSION: These results suggest that the lethality induced by intranasal infection with a high dose of the WR strain is caused by the higher replication of virus in lung cells and immune suppression during the early phase of the infection, resulting in uncontrolled virus replication in the lung.


Subject(s)
Lung/virology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Vaccinia virus/pathogenicity , Vaccinia/immunology , Vaccinia/virology , Animals , Atrophy , Brain/virology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Female , Immune Tolerance , Lung/immunology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Nose/virology , Respiratory Tract Infections/pathology , Thymus Gland/pathology , Vaccinia/pathology , Vaccinia virus/immunology , Vaccinia virus/physiology , Virulence , Virus Replication
17.
Hum Immunol ; 67(7): 512-20, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16829305

ABSTRACT

Immunization with vaccinia virus results in long-lasting protection against smallpox and is an approach that has been successfully used to eliminate natural smallpox infections worldwide. Today, vaccinia virus is very important not only as a vaccine virus to protect human against smallpox, but also as an expression vector for immunization against other infectious diseases, such as HIV and cancer. In this article, we identify three new vaccinia human CD8+ T-cell epitopes conserved among vaccinia and variola viruses restricted by HLA-A2, HLA-B7, or HLA-B*3502, which belongs to the HLA-B7 supertype. Identification of these CD8+ T-cell epitopes restricted by common HLA alleles will help to quantitate human CD8+ T-cell responses to licensed and experimental smallpox vaccines and to vaccinia virus vectors. CD8+ T-cell responses specific to these epitopes can also be used to quantitate cellular immune responses, especially with new smallpox vaccines that do not induce a "take," such as the modified vaccinia virus Ankara strain. Combined with previous reports by us and others, these results show that there are some vaccinia viral proteins containing multiple epitopes restricted by different MHC molecules of humans and mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , HLA-B7 Antigen/immunology , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Variola virus/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay/methods , HLA-B35 Antigen/immunology , Humans , Interferon-gamma/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
18.
Cell Host Microbe ; 19(6): 800-13, 2016 Jun 08.
Article in English | MEDLINE | ID: mdl-27281570

ABSTRACT

Pathogenic H7N9 avian influenza viruses continue to represent a public health concern, and several candidate vaccines are currently being developed. It is vital to assess if protective antibodies are induced following vaccination and to characterize the diversity of epitopes targeted. Here we characterized the binding and functional properties of twelve H7-reactive human antibodies induced by a candidate A/Anhui/1/2013 (H7N9) vaccine. Both neutralizing and non-neutralizing antibodies protected mice in vivo during passive transfer challenge experiments. Mapping the H7 hemagglutinin antigenic sites by generating escape mutant variants against the neutralizing antibodies identified unique epitopes on the head and stalk domains. Further, the broadly cross-reactive non-neutralizing antibodies generated in this study were protective through Fc-mediated effector cell recruitment. These findings reveal important properties of vaccine-induced antibodies and provide a better understanding of the human monoclonal antibody response to influenza in the context of vaccines.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/immunology , Antigens, Viral/immunology , Disease Models, Animal , Dogs , Female , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
19.
Virus Res ; 114(1-2): 104-12, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16039000

ABSTRACT

The K1L protein of vaccinia virus is required for its growth in certain cell lines (RK-13 and human). The cowpox host-range protein CP77 has been shown to complement K1L function in RK-13 cells, despite a lack of homology between the two proteins except for ankyrin repeats. We investigated the role of ankyrin repeats of K1L protein in RK-13 cells. The growth of a recombinant vaccinia virus, with K1L gene mutated in the most conserved ankyrin repeat, was severely impaired. Infection with the mutant virus caused shutdown of cellular and viral protein synthesis early in infection. We also investigated the interaction of K1L protein with cellular proteins and found that K1L interacts with the rabbit homologue of human ACAP2, a GTPase-activating protein with ankyrin repeats. Our result suggests the importance of ankyrin repeat for host-range function of K1L in RK-13 cells and identifies ACAP2 as a cellular protein, which may be interacting with K1L.


Subject(s)
Ankyrin Repeat , GTPase-Activating Proteins/metabolism , Vaccinia virus/pathogenicity , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Humans , Immunoprecipitation , Kidney/cytology , Kidney/virology , Molecular Sequence Data , Mutation , Rabbits , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Proteins/genetics
20.
Viral Immunol ; 18(4): 722-9, 2005.
Article in English | MEDLINE | ID: mdl-16359238

ABSTRACT

Interferon-gamma (IFN-gamma) has antiviral activity against poxviruses as well as many other viruses, bacteria and a parasite, Toxoplasma gondii. Inducible nitric oxide synthase (NOS2) has been shown to mediate the antiviral activity of IFN-gamma in both in vivo and in vitro experiments. In macrophages, inhibition of replication of poxviruses by IFN-gamma is NOS2-dependent. In this report we tested nonmacrophage cell lines and found that indoleamine 2,3-dioxygenase (IDO) also mediated the antiviral activity of IFN-gamma against vaccinia virus. L-tryptophan, an inhibitor of IDO, completely blocked the antiviral activity of IFN-gamma against vaccinia virus in 143B cells, an human osteosarcoma cell line, whereas N(G)-methyl-L-arginine, a NOS2 inhibitor, did not. IDO may account for the NOS2-independent antiviral mechanism induced by IFN-gamma. IFN-gamma may use different antiviral mechanisms in different cell types.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Interferon-gamma/pharmacology , Vaccinia virus/growth & development , Antiviral Agents/pharmacology , Blotting, Western , Cell Division/drug effects , Cell Line, Transformed , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/analysis , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Tryptophan/pharmacology , Vaccinia virus/drug effects , Viral Plaque Assay , omega-N-Methylarginine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL