Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Gen Virol ; 100(3): 431-445, 2019 03.
Article in English | MEDLINE | ID: mdl-30714896

ABSTRACT

A non-replicating form of pseudotyped influenza virus, inactivated by suppression of the haemagglutinin signal sequence (S-FLU), can act as a broadly protective vaccine. S-FLU can infect for a single round only, and induces heterotypic protection predominantly through activation of cross-reactive T cells in the lung. Unlike the licensed live attenuated virus, it cannot reassort a pandemic haemagglutinin (HA) into seasonal influenza. Here we present data on four new forms of S-FLU coated with H7 HAs from either A/Anhui/1/2013, A/Shanghai/1/2013, A/Netherlands/219/2003 or A/New York/107/2003 strains of H7 virus. We show that intranasal vaccination induced a strong local CD8 T cell response and protected against heterosubtypic X31 (H3N2) virus and highly virulent PR8 (H1N1), but not influenza B virus. Intranasal vaccination also induced a strong neutralizing antibody response to the encoded neuraminidase. If given at higher dose in the periphery with intraperitoneal administration, H7 S-FLU induced a specific neutralizing antibody response to H7 HA coating the particle. Polyvalent intraperitoneal vaccination with mixed H7 S-FLU induced a broadly neutralizing antibody response to all four H7 strains. S-FLU is a versatile vaccine candidate that could be rapidly mobilized ahead of a new pandemic threat.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza, Human/prevention & control , Neuraminidase/immunology , Animals , Cross Protection , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/virology , Male , Mice, Inbred C57BL , Neuraminidase/genetics , Vaccination
2.
J Virol ; 86(24): 13397-406, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015719

ABSTRACT

There is a need for vaccines that can protect broadly across all influenza A strains. We have produced a pseudotyped influenza virus based on suppression of the A/PR/8/34 hemagglutinin signal sequence (S-FLU) that can infect cells and express the viral core proteins and neuraminidase but cannot replicate. We show that when given by inhalation to mice, S-FLU is nonpathogenic but generates a vigorous T cell response in the lung associated with markedly reduced viral titers and weight loss after challenge with H1 and H3 influenza viruses. These properties of S-FLU suggest that it may have potential as a broadly protective A virus vaccine, particularly in the setting of a threatened pandemic before matched subunit vaccines become available.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/immunology , Administration, Inhalation , Animals , Antibodies, Viral/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique, Indirect , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutralization Tests , T-Lymphocytes/immunology
3.
J Infect Dis ; 205(1): 20-7, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22080094

ABSTRACT

BACKGROUND: Most reported human H5N1 viral infections have been severe and were detected after hospital admission. A case ascertainment bias may therefore exist, with mild cases or asymptomatic infections going undetected. We sought evidence of mild or asymptomatic H5N1 infection by examining H5N1-specific T-cell and antibody responses in a high-risk cohort in Vietnam. METHODS: Peripheral blood mononuclear cells were tested using interferon-γ enzyme-linked immunospot T assays measuring the response to peptides of influenza H5, H3, and H1 hemagglutinin (HA), N1 and N2 neuraminidase, and the internal proteins of H3N2. Horse erythrocyte hemagglutination inhibition assay was performed to detect antibodies against H5N1. RESULTS: Twenty-four of 747 individuals demonstrated H5-specific T-cell responses but little or no cross-reactivity with H3 or H1 HA peptides. H5N1 peptide-specific T-cell lines that did not cross-react with H1 or H3 influenza virus HA peptides were generated. Four individuals also had antibodies against H5N1. CONCLUSIONS: This is the first report of ex vivo H5 HA-specific T-cell responses in a healthy but H5N1-exposed population. Our results indicate that the presence of H5N1-specific T cells could be an additional diagnostic tool for asymptomatic H5N1 infection.


Subject(s)
Antibodies, Viral/blood , Asymptomatic Infections , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/diagnosis , T-Lymphocytes/physiology , Cohort Studies , Enzyme-Linked Immunospot Assay , Humans , Influenza, Human/epidemiology , Influenza, Human/immunology , Seroepidemiologic Studies , Vietnam/epidemiology
4.
Clin Infect Dis ; 55(1): 19-25, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22441650

ABSTRACT

BACKGROUND: The novel influenza vaccine MVA-NP+M1 is designed to boost cross-reactive T-cell responses to internal antigens of the influenza A virus that are conserved across all subtypes, providing protection against both influenza disease and virus shedding against all influenza A viruses. Following a phase 1 clinical study that demonstrated vaccine safety and immunogenicity, a phase 2a vaccination and influenza challenge study has been conducted in healthy adult volunteers. METHODS: Volunteers with no measurable serum antibodies to influenza A/Wisconsin/67/2005 received either a single vaccination with MVA-NP+M1 or no vaccination. T-cell responses to the vaccine antigens were measured at enrollment and again prior to virus challenge. All volunteers underwent intranasal administration of influenza A/Wisconsin/67/2005 while in a quarantine unit and were monitored for symptoms of influenza disease and virus shedding. RESULTS: Volunteers had a significantly increased T-cell response to the vaccine antigens following a single dose of the vaccine, with an increase in cytolytic effector molecules. Intranasal influenza challenge was undertaken without safety issues. Two of 11 vaccinees and 5 of 11 control subjects developed laboratory-confirmed influenza (symptoms plus virus shedding). Symptoms of influenza were less pronounced in the vaccinees and there was a significant reduction in the number of days of virus shedding in those vaccinees who developed influenza (mean, 1.09 days in controls, 0.45 days in vaccinees, P = .036). CONCLUSIONS: This study provides the first demonstration of clinical efficacy of a T-cell-based influenza vaccine and indicates that further clinical development should be undertaken. CLINICAL TRIALS REGISTRATION: NCT00993083.


Subject(s)
Antibodies, Viral/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , T-Lymphocytes/immunology , Administration, Intranasal , Adolescent , Adult , Antibodies, Viral/blood , Antigens, Viral/immunology , HLA-A2 Antigen , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Influenza, Human/virology , Interferon-gamma , Middle Aged , Nucleocapsid Proteins , Pilot Projects , RNA-Binding Proteins/immunology , Viral Core Proteins/immunology , Viral Matrix Proteins/immunology , Virus Shedding
5.
Nat Microbiol ; 4(2): 306-315, 2019 02.
Article in English | MEDLINE | ID: mdl-30478290

ABSTRACT

Little is known about the specificities and neutralization breadth of the H7-reactive antibody repertoire induced by natural H7N9 infection in humans. We have isolated and characterized 73 H7-reactive monoclonal antibodies from peripheral B cells from four donors infected in 2013 and 2014. Of these, 45 antibodies were H7-specific, and 17 of these neutralized the virus, albeit with few somatic mutations in their variable domain sequences. An additional set of 28 antibodies, isolated from younger donors born after 1968, cross-reacted between H7 and H3 haemagglutinins in binding assays, and had accumulated significantly more somatic mutations, but were predominantly non-neutralizing in vitro. Crystal structures of three neutralizing and protective antibodies in complex with the H7 haemagglutinin revealed that they recognize overlapping residues surrounding the receptor-binding site of haemagglutinin. One of the antibodies, L4A-14, bound into the sialic acid binding site and made contacts with haemagglutinin residues that were conserved in the great majority of 2016-2017 H7N9 isolates. However, only 3 of the 17 neutralizing antibodies retained activity for the Yangtze River Delta lineage viruses isolated in 2016-2017 that have undergone antigenic change, which emphasizes the need for updated H7N9 vaccines.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/metabolism , Antibodies, Viral/administration & dosage , Antibodies, Viral/metabolism , Binding Sites , Cross Reactions/immunology , Disease Models, Animal , Epitopes , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza, Human/prevention & control , Mice, Inbred BALB C , Protein Conformation
6.
JCI Insight ; 2(7): e91868, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28405622

ABSTRACT

In each influenza season, a distinct group of young, otherwise healthy individuals with no risk factors succumbs to life-threatening infection. To better understand the cause for this, we analyzed a broad range of immune responses in blood from a unique cohort of patients, comprising previously healthy individuals hospitalized with and without respiratory failure during one influenza season, and infected with one specific influenza A strain. This analysis was compared with similarly hospitalized influenza patients with known risk factors (total of n = 60 patients recruited). We found a sustained increase in a specific subset of proinflammatory monocytes, with high TNF-α expression and an M1-like phenotype (independent of viral titers), in these previously healthy patients with severe disease. The relationship between M1-like monocytes and immunopathology was strengthened using murine models of influenza, in which severe infection generated using different models (including the high-pathogenicity H5N1 strain) was also accompanied by high levels of circulating M1-like monocytes. Additionally, a raised M1/M2 macrophage ratio in the lungs was observed. These studies identify a specific subtype of monocytes as a modifiable immunological determinant of disease severity in this subgroup of severely ill, previously healthy patients, offering potential novel therapeutic avenues.


Subject(s)
Influenza, Human/immunology , Macrophages/immunology , Monocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Animals , Female , Humans , Influenza A Virus, H5N1 Subtype , Influenza, Human/pathology , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype , Viral Load , Young Adult
7.
J Clin Invest ; 125(7): 2631-45, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26011643

ABSTRACT

The selective pressure that drives antigenic changes in influenza viruses is thought to originate from the human immune response. Here, we have characterized the B cell repertoire from a previously vaccinated donor whose serum had reduced neutralizing activity against the recently evolved clade 6B H1N1pdm09 viruses. While the response was markedly polyclonal, 88% of clones failed to recognize clade 6B viruses; however, the ability to neutralize A/USSR/90/1977 influenza, to which the donor would have been exposed in childhood, was retained. In vitro selection of virus variants with representative monoclonal antibodies revealed that a single amino acid replacement at residue K163 in the Sa antigenic site, which is characteristic of the clade 6B viruses, was responsible for resistance to neutralization by multiple monoclonal antibodies and the donor serum. The K163 residue lies in a part of a conserved surface that is common to the hemagglutinins of the 1977 and 2009 H1N1 viruses. Vaccination with the 2009 hemagglutinin induced an antibody response tightly focused on this common surface that is capable of selecting current antigenic drift variants in H1N1pdm09 influenza viruses. Moreover, amino acid replacement at K163 was not highlighted by standard ferret antisera. Human monoclonal antibodies may be a useful adjunct to ferret antisera for detecting antigenic drift in influenza viruses.


Subject(s)
Antigens, Viral/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Amino Acid Substitution , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Antibody Specificity , Antigenic Variation , Antigens, Viral/chemistry , B-Lymphocytes/immunology , Cross Reactions , Ferrets , Genetic Drift , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Immunoglobulin G/metabolism , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Models, Molecular , Protein Conformation
8.
Front Immunol ; 6: 287, 2015.
Article in English | MEDLINE | ID: mdl-26082783

ABSTRACT

BACKGROUND: In a phase I clinical trial, a H5N1 pandemic live attenuated influenza virus (pLAIV) VN2004 vaccine bearing avian influenza H5N1 hemagglutinin (HA) and NA genes on the A/Ann Arbor cold-adapted vaccine backbone displayed very restricted replication. We evaluated T cell responses to H5N1 pLAIV vaccination and assessed pre-existing T cell responses to determine whether they were associated with restricted replication of the H5N1 pLAIV. METHOD: ELISPOT assays were performed using pools of overlapping peptides spanning the entire H5N1 proteome and the HA proteins of relevant seasonal H1N1 and H3N2 viruses. We tested stored peripheral blood mononuclear cells (PBMCs) from 21 study subjects who received two doses of the H5N1 pLAIV. The PBMCs were collected 1 day before and 7 days after the first and second pLAIV vaccine doses, respectively. RESULT: T cell responses to conserved internal proteins M and NP were significantly boosted by vaccination (p = 0.036). In addition, H5N1 pLAIV appeared to preferentially stimulate and boost pre-existing seasonal influenza virus HA-specific T cell responses that showed low cross-reactivity with the H5 HA. We confirmed this observation by T cell cloning and identified a novel HA-specific epitope. However, we did not find any evidence that pre-existing T cells prevented pLAIV replication and take. CONCLUSION: We found that cross-reactive T cell responses could be boosted by pLAIV regardless of the induction of antibody. The impact of the "original antigenic sin" phenomenon in a subset of volunteers, with preferential expansion of seasonal influenza-specific but not H5N1-specific T cell responses merits further investigation.

9.
Clin Vaccine Immunol ; 22(9): 1004-12, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26135973

ABSTRACT

The continued discovery and development of adjuvants for vaccine formulation are important to safely increase potency and/or reduce the antigen doses of existing vaccines and tailor the adaptive immune response to newly developed vaccines. Adjuplex is a novel adjuvant platform based on a purified lecithin and carbomer homopolymer. Here, we analyzed the adjuvant activity of Adjuplex in mice for the soluble hemagglutinin (HA) glycoprotein of influenza A virus. The titration of Adjuplex revealed an optimal dose of 1% for immunogenicity, eliciting high titers of HA-specific IgG but inducing no significant weight loss. At this dose, Adjuplex completely protected mice from an otherwise lethal influenza virus challenge and was at least as effective as the adjuvants monophosphoryl lipid A (MPL) and alum in preventing disease. Adjuplex elicited balanced Th1-/Th2-type immune responses with accompanying cytokines and triggered antigen-specific CD8(+) T-cell proliferation. The use of the peritoneal inflammation model revealed that Adjuplex recruited dendritic cells (DCs), monocytes, and neutrophils in the context of innate cytokine and chemokine secretion. Adjuplex neither triggered classical maturation of DCs nor activated a pathogen recognition receptor (PRR)-expressing NF-κB reporter cell line, suggesting a mechanism of action different from that reported for classical pathogen-associated molecular pattern (PAMP)-activated innate immunity. Taken together, these data reveal Adjuplex to be a potent and well-tolerated adjuvant with application for subunit vaccines.


Subject(s)
Adaptive Immunity , Adjuvants, Immunologic , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Acrylic Resins , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Influenza Vaccines/administration & dosage , Lecithins/immunology , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Pathogen-Associated Molecular Pattern Molecules , Th1-Th2 Balance , Vaccination
10.
Elife ; 32014 Nov 11.
Article in English | MEDLINE | ID: mdl-25385531

ABSTRACT

During infection, CD8(+) T cells initially expand then contract, leaving a small memory pool providing long lasting immunity. While it has been described that CD8(+) T cell memory formation becomes defective in old age, the cellular mechanism is largely unknown. Autophagy is a major cellular lysosomal degradation pathway of bulk material, and levels are known to fall with age. In this study, we describe a novel role for autophagy in CD8(+) T cell memory formation. Mice lacking the autophagy gene Atg7 in T cells failed to establish CD8(+) T cell memory to influenza and MCMV infection. Interestingly, autophagy levels were diminished in CD8(+) T cells from aged mice. We could rejuvenate CD8(+) T cell responses in elderly mice in an autophagy dependent manner using the compound spermidine. This study reveals a cell intrinsic explanation for poor CD8(+) T cell memory in the elderly and potentially offers novel immune modulators to improve aged immunity.


Subject(s)
Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Animals , Apoptosis/immunology , Autophagy-Related Protein 7 , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Proliferation , Cell Survival , Epitopes/immunology , Glucose Transporter Type 1/metabolism , Immunization, Secondary , Lymphocyte Count , Lymphocytic choriomeningitis virus/immunology , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Phenotype , Reactive Oxygen Species/metabolism , Viral Vaccines/immunology
11.
PLoS One ; 8(5): e62778, 2013.
Article in English | MEDLINE | ID: mdl-23658773

ABSTRACT

Current influenza vaccines stimulate neutralising antibody to the haemagglutinin antigen but as there is antigenic drift in HA it is difficult to prepare a vaccine in advance against an emergent strain. A potential strategy is to induce CD8(+) and CD4(+) T cells that recognize epitopes within internal proteins that are less subject to antigenic drift. Augmenting humoral responses to HA with T cell responses to more conserved antigens may result in a more broadly protective vaccine. In this study, we evaluate the quality of influenza specific T cell responses in a clinical trial using MVA-NP+M1 vaccination followed by influenza virus challenge. In vaccinated volunteers, the expression of Granzyme A, Perforin and CD57 on influenza HLA A*02 M158-66 antigen specific cells was higher than non-vaccinated volunteers before and after challenge despite a similar frequency of antigen specific cells. BCL2 expression was lower in vaccinated volunteers. These data indicate that antigen specific T cells are a useful additional measure for use in human vaccination or immunization studies.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Vaccination , Viral Vaccines/administration & dosage , Adolescent , Adult , Antigens, Viral/genetics , CD57 Antigens/genetics , CD57 Antigens/immunology , CD8-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte/genetics , Granzymes/genetics , Granzymes/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Hemagglutinins/genetics , Hemagglutinins/immunology , Humans , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/genetics , Influenza, Human/immunology , Middle Aged , Nucleoproteins/genetics , Nucleoproteins/immunology , Orthomyxoviridae/drug effects , Orthomyxoviridae/immunology , Perforin/genetics , Perforin/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, DNA , Vaccines, Synthetic , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
12.
J Immunol ; 178(2): 1030-8, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202366

ABSTRACT

We show in this study several novel features of T cell-based heterosubtypic immunity against the influenza A virus in mice. First, T cell-mediated heterosubtypic protection against lethal challenge can be generated by a very low priming dose. Second, it becomes effective within 5-6 days. Third, it provides protection against a very high dose challenge for >70 days. Also novel is the finding that strong, long-lasting, heterosubtypic protection can be elicited by priming with attenuated cold-adapted strains. We demonstrate that priming does not prevent infection of the lungs following challenge, but leads to earlier clearance of the virus and 100% survival after otherwise lethal challenge. Protection is dependent on CD8 T cells, and we show that CD4 and CD8 T cells reactive to conserved epitopes of the core proteins of the challenge virus are present after priming. Our results suggest that intranasal vaccination with cold-adapted, attenuated live virus has the potential to provide effective emergency protection against emerging influenza strains for several months.


Subject(s)
Adaptation, Biological , Cold Temperature , Cross-Priming/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Amino Acid Sequence , Animals , Epitopes/chemistry , Epitopes/immunology , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H3N2 Subtype/classification , Mice , Molecular Sequence Data , Nucleocapsid Proteins , Nucleoproteins/chemistry , Nucleoproteins/immunology , Orthomyxoviridae Infections/prevention & control , Phylogeny , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/immunology , Survival Rate , T-Lymphocytes/immunology , Time Factors , Viral Core Proteins/chemistry , Viral Core Proteins/immunology
13.
J Immunol ; 178(9): 5744-52, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17442958

ABSTRACT

Compared with the MHC of typical mammals, the chicken MHC (BF/BL region) of the B12 haplotype is smaller, simpler, and rearranged, with two classical class I genes of which only one is highly expressed. In this study, we describe the development of long-distance PCR to amplify some or all of each class I gene separately, allowing us to make the following points. First, six other haplotypes have the same genomic organization as B12, with a poorly expressed (minor) BF1 gene between DMB2 and TAP2 and a well-expressed (major) BF2 gene between TAP2 and C4. Second, the expression of the BF1 gene is crippled in three different ways in these haplotypes: enhancer A deletion (B12, B19), enhancer A divergence and transcription start site deletion (B2, B4, B21), and insertion/rearrangement leading to pseudogenes (B14, B15). Third, the three kinds of alterations in the BF1 gene correspond to dendrograms of the BF1 and poorly expressed class II B (BLB1) genes reflecting mostly neutral changes, while the dendrograms of the BF2 and well-expressed class II (BLB2) genes each have completely different topologies reflecting selection. The common pattern for the poorly expressed genes reflects the fact the BF/BL region undergoes little recombination and allows us to propose a pattern of descent for these chicken MHC haplotypes from a common ancestor. Taken together, these data explain how stable MHC haplotypes predominantly express a single class I molecule, which in turn leads to striking associations of the chicken MHC with resistance to infectious pathogens and response to vaccines.


Subject(s)
Chickens/genetics , Evolution, Molecular , Genes, MHC Class I , Histocompatibility Antigens Class I/classification , Histocompatibility Antigens Class I/genetics , Animals , Base Sequence , Chickens/immunology , Gene Expression , Genetic Drift , Haplotypes , Molecular Sequence Data , Mutation , Phylogeny , Promoter Regions, Genetic
14.
Clin Immunol ; 119(1): 87-94, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16386960

ABSTRACT

How influenza virus dose affects the size of the immune response has not been clearly documented. Mice were challenged with three doses of influenza virus spanning a 100-fold range. Increasing the viral input dose increased the degree of weight loss observed, the clinical score and eventual mortality. Maximum viral loads increased with viral input and lower doses peaked and declined earlier. The level of the immune response only varied 2-fold and was independent of viral dose with near maximal responses elicited by the lowest dose, as measured by influx of antigen-specific and non-specific leukocytes into the lungs and by influenza antibody titers. We conclude that a strong immune response is mounted to a small dose of virus and curbs the spread of virus early and prevents weight loss whereas larger doses of virus elicit a slightly greater response but the associated disease can overwhelm the host.


Subject(s)
Antibody Formation/immunology , Immunity, Cellular/immunology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Count , Cell Movement/immunology , Epitopes, T-Lymphocyte/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Lung/pathology , Lung/virology , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutrophils/immunology , Neutrophils/pathology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Survival Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Weight Loss
15.
Proc Natl Acad Sci U S A ; 103(5): 1434-9, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16432226

ABSTRACT

Compared with the MHC of typical mammals, the chicken MHC is smaller and simpler, with only two class I genes found in the B12 haplotype. We make five points to show that there is a single-dominantly expressed class I molecule that can have a strong effect on MHC function. First, we find only one cDNA for two MHC haplotypes (B14 and B15) and cDNAs corresponding to two genes for the other six (B2, B4, B6, B12, B19, and B21). Second, we find, for the B4, B12, and B15 haplotypes, that one cDNA is at least 10-fold more abundant than the other. Third, we use 2D gel electrophoresis of class I molecules from pulse-labeled cells to show that there is only one heavy chain spot for the B4 and B15 haplotypes, and one major spot for the B12 haplotype. Fourth, we determine the peptide motifs for B4, B12, and B15 cells in detail, including pool sequences and individual peptides, and show that the motifs are consistent with the peptides binding to models of the class I molecule encoded by the abundant cDNA. Finally, having shown for three haplotypes that there is a single dominantly expressed class I molecule at the level of RNA, protein, and antigenic peptide, we show that the motifs can explain the striking MHC-determined resistance and susceptibility to Rous sarcoma virus. These results are consistent with the concept of a "minimal essential MHC" for chickens, in strong contrast to typical mammals.


Subject(s)
Avian Sarcoma Viruses/genetics , Genes, MHC Class I , Peptides/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Antigen Presentation , Avian Sarcoma Viruses/metabolism , Chickens , DNA, Complementary/metabolism , Electrophoresis, Gel, Two-Dimensional , Flow Cytometry , Genes, Dominant , Haplotypes , Models, Molecular , Molecular Sequence Data , Poultry Diseases/virology , Sarcoma, Avian/virology , Sequence Homology, Amino Acid , Time Factors
16.
J Immunol ; 173(5): 2923-7, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15322149

ABSTRACT

Naive CD8(+) T cells are activated on encounter with Ag presented on dendritic cells and proliferate rapidly. To investigate the regulation of naive CD8(+) T cells proliferation, we adoptively transferred TCR-transgenic CD8(+) T cells into intact mice together with Ag-pulsed dendritic cells. Regardless of the number of cells initially transferred, the expansion of activated Ag-specific CD8(+) T cells was limited to a ceiling of effector cells. This limit was reached from a wide range of T cell doses, including a physiological number of precursor cells, and was not altered by changing the amount of Ag or APCs. The total Ag-specific response was composed of similar numbers of host and donor transgenic cells regardless of donor cell input, suggesting that these populations were independently regulated. Regulation of the transgenic donor cell population was TCR specific. We hypothesize that a clone-specific regulatory mechanism controls the extent of CD8(+) T cell responses to Ag.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Count , Cell Division/immunology , Cell Division/physiology , Dendritic Cells/transplantation , Mice , Receptors, Antigen, T-Cell/immunology
17.
Immunology ; 109(2): 197-208, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12757614

ABSTRACT

The relationships between different dendritic cell (DC) populations are not clearly established. In particular, it is not known how DC generated in vitro relate to those identified in vivo. Here we have characterized rat bone marrow-derived DC (BMDC) and compared them with DC isolated from spleen (SDC) and pseudo-afferent lymph (LDC). BMDC express typical DC markers and are mostly OX41 positive and CD4 negative. In contrast to ex vivo DC, some BMDC express Fc receptors. FcR+ and FcR- BMDC express similar levels of major histocompatibility complex class II molecules (MHC) and are B7 positive, but some FcR- BMDC express high levels of B7. In contrast to freshly isolated or cultured ex vivo SDC and LDC, both BMDC subpopulations can express inducible nitric oxide synthase (iNOS) and can secrete nitric oxide (NO) in amounts similar to those secreted by peritoneal macrophages. Despite expressing MHC class II and B7, FcR+ BMDC stimulate only a very weak MLR and inhibit stimulation by FcR- BMDC and ex vivo DC. Inhibition is only partially NO dependent. FcR+ BMDC are not macrophages, as judged by adherence and phagocytosis. Both subpopulations are able to present antigen to primed T cells in vitro and are able to prime naïve CD4 T cells in vivo. However, unlike SDC, BMDC are unable to stimulate cytotoxic T-lymphocyte (CTL) responses to a minor histocompatibility antigen. Thus, BMDC show marked differences to ex vivo DC and their relationship to those of in vivo DC populations, to date, is unclear.


Subject(s)
Bone Marrow Cells/metabolism , Dendritic Cells/metabolism , Nitric Oxide/metabolism , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation/immunology , Bone Marrow Cells/immunology , Cells, Cultured , Dendritic Cells/immunology , Immunophenotyping , Lymph/immunology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Rats, Inbred Strains , Receptors, Fc/metabolism , Spleen/immunology
18.
Clin Immunol ; 113(1): 89-100, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15380534

ABSTRACT

The activation, localization, phenotypic changes, and function of CFSE-labeled naive influenza-specific CD8(+) and CD4(+) T cells following influenza infection were examined. Response of adoptively transferred CD8(+) T cells was seen earliest in draining lymph node. Highly activated cells were found later in the lung, airways, and spleen, were cytolytic, and expressed IFN-gamma upon restimulation. Similar amounts of division at early time points, but higher numbers of CD8(+) T cells, were detected at 9 and 30 days postinfection after cotransfer of CD4(+) and CD8(+) T cells followed by infection. Transfer of much smaller numbers of CD4(+) and CD8(+) T cells led to more extensive expansion but the same difference in final number between the two cell types. These studies demonstrate how CD8(+) and CD4(+) T cells respond to influenza at early time points postinfection and the differential kinetics of antigen-specific CD4(+) and CD8(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Division/immunology , Interferon-gamma/metabolism , Lung/cytology , Lung/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
SELECTION OF CITATIONS
SEARCH DETAIL