1
|
Dai M, Li X, Zhu S, Chen W, Smith AL, Liao M. Chicken TCRγδ+CD8α+T cells are antigen-specific and protective in H9N2 AIV infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf083. [PMID: 40359378 DOI: 10.1093/jimmun/vkaf083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/25/2025] [Indexed: 05/15/2025]
Abstract
TCRγδ+ T cells are a major lymphocyte population of chickens, but their response or contribution to immunity against avian influenza virus (AIV) remains unknown. Here, we report an increase in the proportion and activation state of TCRγδ+CD8α+ T cells in the PBMCs of 3 chicken lines (MHC homozygous H-B2 and H-B21 lines and outbred G-WL line) with the strongest responses observed in the more resistant H-B2 chickens. H9N2 AIV infection induced mRNA upregulation of interferon (IFN)-γ and cytotoxicity-associated molecules, including, Granzyme A, Granzyme K, and perforin in sorted TCRγδ+CD8α+ T cells. Moreover, in ex vivo cultured TCRγδ+CD8α+ T cells in response to H9N2 AIV infected splenocytes, strongly indicates the activation of these cells' cytolytic potential via detection of transcription levels of cytotoxic genes with quantitative reverse transcription polymerase chain reaction (qRT-PCR), and IFN-γ protein level with ELISPOT and an intracellular cytokine staining assays. Most importantly, in vivo depletion of γδ T cells led to reduced H9N2 AIV control, which was particularly evident in the early phase of infection. Taken together, these results indicate that strong TCRγδ+CD8α+T cell response plays a critical role in protecting chicken against H9N2 AIV infection.
Collapse
Affiliation(s)
- Manman Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, China
| | - Xueqing Li
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, China
| | - Sufang Zhu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, China
| | - Weisan Chen
- T cell lab, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Adrian L Smith
- Department of Biology, Medawar Building, University of Oxford, Oxford, United Kingdom
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, China
| |
Collapse
|
2
|
Piesche R, Cazaban C, Frizzo da Silva L, Ramírez-Martínez L, Hufen H, Beer M, Harder T, Grund C. Immunogenicity and Protective Efficacy of Five Vaccines Against Highly Pathogenic Avian Influenza Virus H5N1, Clade 2.3.4.4b, in Fattening Geese. Vaccines (Basel) 2025; 13:399. [PMID: 40333332 PMCID: PMC12031072 DOI: 10.3390/vaccines13040399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025] Open
Abstract
Background/Objectives: The risk of the introduction of highly pathogenic avian influenza virus (HPAIV) in geese breeding and fattening flocks is heightened due to the necessity of free-range access to grazing grounds. This study aimed to evaluate the safety, immunogenicity, and protective efficacy of five commercial vaccines against HPAIV subtype H5N1 (clade 2.3.4.4b) in subadult fattening geese. Methods: A prime-boost vaccination trial was conducted using five commercial vaccines, including H5 expressing vaccines of novel technology (subunit, vector, RNA) and whole inactivated virus (WIV) vaccines. Based on serological results, one RNA and one WIV vaccine were selected for a homologous challenge experiment. Results: Two vaccines of novel technology (vector, RNA) required a booster dose to raise specific antibodies titers above a threshold of four log2 using a hemagglutination inhibition (HI) assay, whereas a subunit vaccine and two WIV vaccines induced seroconversion after primary vaccination. In the challenge experiment, all unvaccinated control geese succumbed to infection by day four. In contrast, all vaccinated geese that had seroconverted exhibited full clinical protection. Although sterile immunity was not achieved, viral excretion was significantly reduced in the vaccinated groups compared to controls. Conclusions: Vaccination substantially mitigated the impact of HPAIV H5N1, clade 2.3.4.4b infection in geese, greatly improving animal welfare by preventing severe disease. Additionally, there was a significant reduction in viral burden. Further studies are necessary to verify the potential of these vaccines to reduce susceptibility to infection and virus excretion in order to achieve suppression of the between-flock reproduction number to < 1 in geese flocks at high risk of infection.
Collapse
Affiliation(s)
- Ronja Piesche
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, 17493 Greifswald, Germany; (R.P.); (M.B.)
| | | | | | | | - Heike Hufen
- Boehringer Ingelheim Vetmedica GmbH, 55218 Ingelheim am Rhein, Germany;
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, 17493 Greifswald, Germany; (R.P.); (M.B.)
| | - Timm Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, 17493 Greifswald, Germany; (R.P.); (M.B.)
| | - Christian Grund
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institute, 17493 Greifswald, Germany; (R.P.); (M.B.)
| |
Collapse
|
3
|
Hassan MSH, Sharif S. Immune responses to avian influenza viruses in chickens. Virology 2025; 603:110405. [PMID: 39837219 DOI: 10.1016/j.virol.2025.110405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/28/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
Chickens are a key species in both the manifestation of avian influenza and the potential for zoonotic transmission. Avian influenza virus (AIV) infection in chickens can range from asymptomatic or mild disease with low pathogenic AIVs (LPAIVs) to systemic fatal disease with high pathogenic AIVs (HPAIVs). During AIV infection in chickens, Toll-like receptor 7 and melanoma differentiation-associated gene 5 are upregulated to detect the single-stranded ribonucleic acid genomes of AIV, triggering a signaling cascade that produces interferons (IFNs) and pro-inflammatory cytokines. These inflammatory mediators induce the expression of antiviral proteins and recruit immune system cells, such as macrophages and dendritic cells, to the infection site. AIV evades these antiviral responses primarily through its non-structural protein 1, which suppresses type I IFNs, influencing viral pathogenicity. The uncontrolled release of pro-inflammatory cytokines may contribute to the pathogenicity and high mortality associated with HPAIV infections. AIV modulates apoptosis in chicken cells to enhance its replication, with variations in apoptosis pathways influenced by viral strain and host cell type. The presentation of AIV antigens to T and B cells leads to the production of neutralizing antibodies and the targeted destruction of infected cells by CD8+ T cells, respectively, which enhances protection and establishes immunological memory. This review explores the diverse innate and adaptive immune responses in chickens to different AIVs, focusing on the dynamics of these responses relative to protection, susceptibility, and potential immunopathology. By understanding these immune mechanisms, informed strategies for controlling AIV infection and improving chicken health can be developed.
Collapse
Affiliation(s)
- Mohamed S H Hassan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada; Department of Avian and Rabbit Medicine, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
4
|
Xie Z, Chen Y, Xie J, Du S, Chen R, Zheng Y, You B, Feng M, Liao M, Dai M. Construction with recombinant epitope-expressing baculovirus enhances protective effects of inactivated H9N2 vaccine against heterologous virus. Vet Microbiol 2025; 300:110337. [PMID: 39671758 DOI: 10.1016/j.vetmic.2024.110337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Although the use of inactivated vaccines has kept avian influenza (AI) outbreaks largely under control, they fail to prevent virus shedding. To enhance the efficacy of inactivated H9N2 AIV vaccines (InV), we constructed a multi-epitope recombinant baculovirus (BV-BNT) containing two B cell epitopes and nine T cell epitopes of H9N2 AIV for combined immunization with InV. The results showed that HI titer, IgG and IgM levels, and the percentage of B cells, CD4+ T cells, CD8+ T cells, and CD4+CD8+ T cells were significantly higher in the InV+BV-BNT immunization group than the InV immunization group. Besides, the expression levels of IL-1β, IFN-γ, IFN-α, IL-4, IL-13, and CXCLi1 were significantly higher in the InV+BV-BNT group than the InV group. Moreover, four conservative peptides (NP182-190, NP455-463, NS198-106, and NP380-393) significantly stimulated splenocytes to express IFN-γ in the InV+BV-BNT group instead of InV group. After heterologous virus challenging, the percentages of CD4+ T and CD8+ T cells were significantly upregulated in the InV+BV-BNT group compared to Inv group at 3 DPI. Viral loads in oropharyngeal of the InV+BV-BNT group was significantly lower than that in the InV group at 3 days post-infection (DPI). Furthermore, compared to the InV group, the virus positivity rate of oropharyngeal and cloacal swabs in the InV+BV-BNT group was lower at 5 DPI, with none positive at 7 DPI. Hence, this study indicated that the combined immunization of InV and BV-BNT could induce stronger humoral and cellular immune responses, shorten the detoxification period and reduce viral load compared to Inv alone, which suggests BV-BNT could act as a supplementary vaccine to potentially address the protection deficiency of the H9N2 inactivated vaccine.
Collapse
Affiliation(s)
- Zimin Xie
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Yingyi Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Jun Xie
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Shanyao Du
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Rongmao Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Yuqin Zheng
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Bowen You
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Min Feng
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou 510642, PR China.
| | - Manman Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou 510642, PR China.
| |
Collapse
|
5
|
Li X, Li Z, Ma M, Yang N, Du S, Liao M, Dai M. Revealing novel and conservative CD8 +T-cell epitopes with MHC B2 restriction on ALV-J. Vet Res 2024; 55:164. [PMID: 39696681 DOI: 10.1186/s13567-024-01426-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/03/2024] [Indexed: 12/20/2024] Open
Abstract
MHC B2 haplotype chickens have been reported to induce strong immune response against various avian pathogens. However, little is known about the CD8+T-cell epitope with MHC B2-restricted on subgroup J avian leukosis virus (ALV-J). In this study, we explored the ALV-J-induced cellular immune response in B2 haplotype chickens in vivo. We found that ALV-J infection significantly increased the proportion of CD8+T cells in chickens and up-regulated the expression of cytotoxic genes like Granzyme A and antiviral genes like IFIT5 at 14 days post-infection (dpi). We selected 32 candidate peptides based on the peptide-binding motif and further identified three MHC B2-restricted CD8+T epitopes on ALV-J, including Pol652-660, Gag374-382, and Gag403-411 which induced significant levels of chicken IFN-γ production in splenocytes from ALV-J infected chickens using the ELISpot assay. In addition, we also verified that the three identified epitopes stimulated memory splenocytes elevating TNF-α and IL-2 protein expression. Importantly, we found that the three positive peptides were highly conserved among ALV-A, ALV-B, ALV-E, ALV-J, and ALV-K. Taken together, we identified three MHC B2-restricted CD8+T cell epitopes on ALV-J, providing a foundation for developing effective T cell epitope vaccines targeting conserved internal viral proteins.
Collapse
Affiliation(s)
- Xueqing Li
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ziwei Li
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Mulin Ma
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Na Yang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Shanyao Du
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, 510642, China.
| | - Manman Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- UK-China Centre of Excellence for Research on Avian Diseases, Guangzhou, 510642, China.
| |
Collapse
|
6
|
Jia Y, Wu Q, Li Y, Ma M, Song W, Chen R, Yao Y, Nair V, Zhang N, Liao M, Dai M. Revealing novel and conservative T-cell epitopes with MHC B2 restriction on H9N2 avian influenza virus (AIV). J Biol Chem 2024; 300:107395. [PMID: 38768812 PMCID: PMC11223079 DOI: 10.1016/j.jbc.2024.107395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
B2 haplotype major histocompatibility complex (MHC) has been extensively reported to confer resistance to various avian diseases. But its peptide-binding motif is unknown, and the presenting peptide is rarely identified. Here, we identified its peptide-binding motif (X-A/V/I/L/P/S/G-X-X-X-X-X-X-V/I/L) in vitro using Random Peptide Library-based MHC I LC-MS/MS analysis. To further clarify the structure basis of motif, we determined the crystal structure of the BF2∗02:01-PB2552-560 complex at 1.9 Å resolution. We found that BF2∗02:01 had a relatively wide antigen-binding groove, and the structural characterization of pockets was consistent with the characterization of peptide-binding motif. The wider features of the peptide-binding motif and increased number of peptides bound by BF2∗02:01 than BF2∗04:01 might resolve the puzzles for the presence of potential H9N2 resistance in B2 chickens. Afterward, we explored the H9N2 avian influenza virus (AIV)-induced cellular immune response in B2 haplotype chickens in vivo. We found that ratio of CD8+ T cell and kinetic expression of cytotoxicity genes including Granzyme K, interferon-γ, NK lysin, and poly-(ADP-ribose) polymerase in peripheral blood mononuclear cells were significantly increased in defending against H9N2 AIV infection. Especially, we selected 425 epitopes as candidate epitopes based on the peptide-binding motif and further identified four CD8+ T-cell epitopes on H9N2 AIV including NS198-106, PB2552-560, NP182-190, and NP455-463 via ELI-spot interferon-γ detections after stimulating memory lymphocytes with peptides. More importantly, these epitopes were found to be conserved in H7N9 AIV and H9N2 AIV. These findings provide direction for developing effective T cell epitope vaccines using well-conserved internal viral antigens in chickens.
Collapse
Affiliation(s)
- Yusheng Jia
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Qingxin Wu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Yilin Li
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Mulin Ma
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Wei Song
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Rongmao Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China
| | - Yongxiu Yao
- Viral Oncogenesis Group, The Pirbright Institute and UK-China Centre of Excellence for Research on Avian Diseases, Surrey, United Kingdom
| | - Venugopal Nair
- Viral Oncogenesis Group, The Pirbright Institute and UK-China Centre of Excellence for Research on Avian Diseases, Surrey, United Kingdom; Department of Zoology, Oxford University, Oxford, United Kingdom
| | - Nianzhi Zhang
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China.
| | - Manman Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
7
|
Luczo JM, Spackman E. Epitopes in the HA and NA of H5 and H7 avian influenza viruses that are important for antigenic drift. FEMS Microbiol Rev 2024; 48:fuae014. [PMID: 38734891 PMCID: PMC11149724 DOI: 10.1093/femsre/fuae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/13/2024] Open
Abstract
Avian influenza viruses evolve antigenically to evade host immunity. Two influenza A virus surface glycoproteins, the haemagglutinin and neuraminidase, are the major targets of host immunity and undergo antigenic drift in response to host pre-existing humoral and cellular immune responses. Specific sites have been identified as important epitopes in prominent subtypes such as H5 and H7, which are of animal and public health significance due to their panzootic and pandemic potential. The haemagglutinin is the immunodominant immunogen, it has been extensively studied, and the antigenic reactivity is closely monitored to ensure candidate vaccine viruses are protective. More recently, the neuraminidase has received increasing attention for its role as a protective immunogen. The neuraminidase is expressed at a lower abundance than the haemagglutinin on the virus surface but does elicit a robust antibody response. This review aims to compile the current information on haemagglutinin and neuraminidase epitopes and immune escape mutants of H5 and H7 highly pathogenic avian influenza viruses. Understanding the evolution of immune escape mutants and the location of epitopes is critical for identification of vaccine strains and development of broadly reactive vaccines that can be utilized in humans and animals.
Collapse
Affiliation(s)
- Jasmina M Luczo
- Australian Animal Health Laboratory, Australian Centre for Disease Preparedness, Commonwealth Scientific and Industrial Research Organisation, East Geelong, Victoria 3219, Australia
| | - Erica Spackman
- Exotic & Emerging Avian Viral Diseases Research, Southeast Poultry Research Laboratory, United States National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture, Athens, GA 30605, United States
| |
Collapse
|
8
|
Martins de Camargo M, Caetano AR, Ferreira de Miranda Santos IK. Evolutionary pressures rendered by animal husbandry practices for avian influenza viruses to adapt to humans. iScience 2022; 25:104005. [PMID: 35313691 PMCID: PMC8933668 DOI: 10.1016/j.isci.2022.104005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Commercial poultry operations produce and crowd billions of birds every year, which is a source of inexpensive animal protein. Commercial poultry is intensely bred for desirable production traits, and currently presents very low variability at the major histocompatibility complex. This situation dampens the advantages conferred by the MHC’s high genetic variability, and crowding generates immunosuppressive stress. We address the proteins of influenza A viruses directly and indirectly involved in host specificities. We discuss how mutants with increased virulence and/or altered host specificity may arise if few class I alleles are the sole selective pressure on avian viruses circulating in immunocompromised poultry. This hypothesis is testable with peptidomics of MHC ligands. Breeding strategies for commercial poultry can easily and inexpensively include high variability of MHC as a trait of interest, to help save billions of dollars as a disease burden caused by influenza and decrease the risk of selecting highly virulent strains.
Collapse
|
9
|
Hao X, Zhang F, Yang Y, Shang S. The Evaluation of Cellular Immunity to Avian Viral Diseases: Methods, Applications, and Challenges. Front Microbiol 2021; 12:794514. [PMID: 34950125 PMCID: PMC8689181 DOI: 10.3389/fmicb.2021.794514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022] Open
Abstract
Cellular immune responses play critical roles in the control of viral infection. However, the immune protection against avian viral diseases (AVDs), a major challenge to poultry industry, is yet mainly evaluated by measuring humoral immune response though antibody-independent immune protection was increasingly evident in the development of vaccines against some of these diseases. The evaluation of cellular immune response to avian viral infection has long been neglected due to limited reagents and methods. Recently, with the availability of more immunological reagents and validated approaches, the evaluation of cellular immunity has become feasible and necessary for AVD. Herein, we reviewed the methods used for evaluating T cell immunity in chickens following infection or vaccination, which are involved in the definition of different cellular subset, the analysis of T cell activation, proliferation and cytokine secretion, and in vitro culture of antigen-presenting cells (APC) and T cells. The pros and cons of each method were discussed, and potential future directions to enhance the studies of avian cellular immunity were suggested. The methodological improvement and standardization in analyzing cellular immune response in birds after viral infection or vaccination would facilitate the dissection of mechanism of immune protection and the development of novel vaccines and therapeutics against AVD.
Collapse
Affiliation(s)
- Xiaoli Hao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Fan Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yi Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Shaobin Shang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- International Corporation Laboratory of Agriculture and Agricultural Products Safety, Yangzhou University, Yangzhou, China
| |
Collapse
|
10
|
Tregaskes CA, Kaufman J. Chickens as a simple system for scientific discovery: The example of the MHC. Mol Immunol 2021; 135:12-20. [PMID: 33845329 PMCID: PMC7611830 DOI: 10.1016/j.molimm.2021.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 01/07/2023]
Abstract
Chickens have played many roles in human societies over thousands of years, most recently as an important model species for scientific discovery, particularly for embryology, virology and immunology. In the last few decades, biomedical models like mice have become the most important model organism for understanding the mechanisms of disease, but for the study of outbred populations, they have many limitations. Research on humans directly addresses many questions about disease, but frank experiments into mechanisms are limited by practicality and ethics. For research into all levels of disease simultaneously, chickens combine many of the advantages of humans and of mice, and could provide an independent, integrated and overarching system to validate and/or challenge the dogmas that have arisen from current biomedical research. Moreover, some important systems are simpler in chickens than in typical mammals. An example is the major histocompatibility complex (MHC) that encodes the classical MHC molecules, which play crucial roles in the innate and adaptive immune systems. Compared to the large and complex MHCs of typical mammals, the chicken MHC is compact and simple, with single dominantly-expressed MHC molecules that can determine the response to infectious pathogens. As a result, some fundamental principles have been easier to discover in chickens, with the importance of generalist and specialist MHC alleles being the latest example.
Collapse
Affiliation(s)
- Clive A Tregaskes
- University of Cambridge, Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP, United Kingdom
| | - Jim Kaufman
- University of Cambridge, Department of Pathology, Tennis Court Road, Cambridge, CB2 1QP, United Kingdom; University of Edinburgh, Institute for Immunology and Infection Research, Ashworth Laboratories, Kings Buildings, Edinburgh, EH9 3FL, United Kingdom.
| |
Collapse
|
11
|
Reemers S, Verstegen I, Basten S, Hubers W, van de Zande S. A broad spectrum HVT-H5 avian influenza vector vaccine which induces a rapid onset of immunity. Vaccine 2021; 39:1072-1079. [PMID: 33483211 DOI: 10.1016/j.vaccine.2021.01.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/15/2020] [Accepted: 01/04/2021] [Indexed: 11/25/2022]
Abstract
Current methods to combat highly pathogenic avian influenza (HPAI) outbreaks in poultry rely on stamping out and preventive culling, which can lead to high economic losses and invoke ethical resistance. Emergency vaccination could be an alternative as vaccination is one of the most efficient and cost-effective measures to protect poultry from HPAI infection, preventing spreading to other poultry and greatly reducing the potential transmission to humans. Current conventional inactivated AI vaccines may be useful for combating AI outbreaks, but do not fulfil all targets of an ideal AI vaccine, including mass applicability and rapid onset of immunity. We aimed to further investigate the potential of Herpesvirus of Turkeys (HVT) as a vector containing a recombinant H5 hemagglutinin of HPAI H5N1. This HVT-H5 vector was analysed in vitro, tested for onset of immunity against AI challenge, breadth of protection, reduction of virus shedding, and induction of both antibody and cellular responses in SPF layers or broiler chicks containing maternal derived antibodies (MDA+). In SPF layers HVT-H5 provided full protection to lethal challenges with 4 antigenically diverse HPAI H5N1 strains from 2 weeks post vaccination (w.p.v.), while in MDA+ birds full protection was provided from 3 w.p.v. to homologous challenge. Also shedding of challenge virus was reduced in both SPF and MDA+ birds. HVT-H5 induced a protective HI titre (≥4) to 11 HPAI H5N1 strains at 3 w.p.v. in 3-week-old SPF layers and to HPAI H5N8 A/ch/Neth/14015531/2014. Besides inducing a protective antibody response HVT-H5 also induced an influenza-specific T cell response. This data demonstrates that HVT-H5 vaccine appears to fulfil many of the criteria for an ideal AI vaccine including early onset of immunity, a broad protection, reduced virus shedding, protection in presence of AI-MDA and could be a useful tool in the combat of AI outbreaks worldwide.
Collapse
Affiliation(s)
- Sylvia Reemers
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands.
| | - Iwan Verstegen
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands
| | - Stephanie Basten
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands
| | - Willem Hubers
- MSD Animal Health, Wim de Körverstraat 35, 5831 AN Boxmeer, the Netherlands
| | | |
Collapse
|
12
|
Improvement of PR8-Derived Recombinant Clade 2.3.4.4c H5N6 Vaccine Strains by Optimization of Internal Genes and H103Y Mutation of Hemagglutinin. Vaccines (Basel) 2020; 8:vaccines8040781. [PMID: 33419331 PMCID: PMC7766170 DOI: 10.3390/vaccines8040781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/04/2022] Open
Abstract
Clade 2.3.4.4c H5N6 avian influenza A viruses (AIVs) may have originally adapted to infect chickens and have caused highly pathogenic avian influenza (HPAI) in poultry and human fatalities. Although A/Puerto Rico/8/1934 (H1N1) (PR8)-derived recombinant clade 2.3.4.4c H5N6 vaccine strains have been effective in embryonated chicken eggs-based vaccine production system, they need to be improved in terms of immunogenicity and potential mammalian pathogenicity. We replaced the PB2 gene alone or the PB2 (polymerase basic protein 2), NP (nucleoprotein), M (matrix protein) and NS (non-structural protein) genes together in the PR8 strain with corresponding genes from AIVs with low pathogenicity to remove mammalian pathogenicity and to match CD8+ T cell epitopes with contemporary HPAI viruses, respectively, without loss of viral fitness. Additionally, we tested the effect of the H103Y mutation of hemagglutinin (HA) on antigen productivity, mammalian pathogenicity and heat/acid stability. The replacement of PB2 genes and the H103Y mutation reduced the mammalian pathogenicity but increased the antigen productivity of the recombinant vaccine strains. The H103Y mutation increased heat stability but unexpectedly decreased acid stability, probably resulting in increased activation pH for HA. Interestingly, vaccination with inactivated recombinant virus with replaced NP, M and NS genes halted challenge virus shedding earlier than the recombinant vaccine without internal genes replacement. In conclusion, we successfully generated recombinant clade 2.3.4.4c H5N6 vaccine strains that were less pathogenic to mammals and more productive and heat stable than conventional PR8-derived recombinant strains by optimization of internal genes and the H103Y mutation of HA.
Collapse
|
13
|
Yang Y, Dong M, Hao X, Qin A, Shang S. Revisiting cellular immune response to oncogenic Marek's disease virus: the rising of avian T-cell immunity. Cell Mol Life Sci 2020; 77:3103-3116. [PMID: 32080753 PMCID: PMC7391395 DOI: 10.1007/s00018-020-03477-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/21/2022]
Abstract
Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that causes deadly T-cell lymphomas and serves as a natural virus-induced tumor model in chickens. Although Marek's disease (MD) is well controlled by current vaccines, the evolution of MDV field viruses towards increasing virulence is concerning as a better vaccine to combat very virulent plus MDV is still lacking. Our understanding of molecular and cellular immunity to MDV and its immunopathogenesis has significantly improved, but those findings about cellular immunity to MDV are largely out-of-date, hampering the development of more effective vaccines against MD. T-cell-mediated cellular immunity was thought to be of paramount importance against MDV. However, MDV also infects macrophages, B cells and T cells, leading to immunosuppression and T-cell lymphoma. Additionally, there is limited information about how uninfected immune cells respond to MDV infection or vaccination, specifically, the mechanisms by which T cells are activated and recognize MDV antigens and how the function and properties of activated T cells correlate with immune protection against MDV or MD tumor. The current review revisits the roles of each immune cell subset and its effector mechanisms in the host immune response to MDV infection or vaccination from the point of view of comparative immunology. We particularly emphasize areas of research requiring further investigation and provide useful information for rational design and development of novel MDV vaccines.
Collapse
Affiliation(s)
- Yi Yang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Maoli Dong
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoli Hao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Aijian Qin
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
- International Corporation Laboratory of Agriculture and Agricultural Products Safety, Yangzhou University, Yangzhou, 225009, China.
- Ministry of Education Key Laboratory for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
- Key Laboratory of Jiangsu Preventive Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
| | - Shaobin Shang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
- International Corporation Laboratory of Agriculture and Agricultural Products Safety, Yangzhou University, Yangzhou, 225009, China.
- Ministry of Education Key Laboratory for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
14
|
Li X, Zhang L, Liu Y, Ma L, Zhang N, Xia C. Structures of the MHC-I molecule BF2*1501 disclose the preferred presentation of an H5N1 virus-derived epitope. J Biol Chem 2020; 295:5292-5306. [PMID: 32152225 PMCID: PMC7170506 DOI: 10.1074/jbc.ra120.012713] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/06/2020] [Indexed: 01/05/2023] Open
Abstract
Lethal infections by strains of the highly-pathogenic avian influenza virus (HPAIV) H5N1 pose serious threats to both the poultry industry and public health worldwide. A lack of confirmed HPAIV epitopes recognized by cytotoxic T lymphocytes (CTLs) has hindered the utilization of CD8+ T-cell-mediated immunity and has precluded the development of effectively diversified epitope-based vaccination approaches. In particular, an HPAIV H5N1 CTL-recognized epitope based on the peptide MHC-I-β2m (pMHC-I) complex has not yet been designed. Here, screening a collection of selected peptides of several HPAIV strains against a specific pathogen-free pMHC-I (pBF2*1501), we identified a highly-conserved HPAIV H5N1 CTL epitope, named HPAIV-PA123-130 We determined the structure of the BF2*1501-PA123-130 complex at 2.1 Å resolution to elucidate the molecular mechanisms of a preferential presentation of the highly-conserved PA123-130 epitope in the chicken B15 lineage. Conformational characteristics of the PA123-130 epitope with a protruding Tyr-7 residue indicated that this epitope has great potential to be recognized by specific TCRs. Moreover, significantly increased numbers of CD8+ T cells specific for the HPAIV-PA123-130 epitope in peptide-immunized chickens indicated that a repertoire of CD8+ T cells can specifically respond to this epitope. We anticipate that the identification and structural characterization of the PA123-130 epitope reported here could enable further studies of CTL immunity against HPAIV H5N1. Such studies may aid in the development of vaccine development strategies using well-conserved internal viral antigens in chickens.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Lijie Zhang
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Yanjie Liu
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China; Key Laboratory for Insect-Pollinator Biology of the Ministry of Agriculture, Institute of Apiculture, Chinese Academy of Agricultural Sciences, Beijing 100093, People's Republic of China
| | - Lizhen Ma
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Nianzhi Zhang
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China
| | - Chun Xia
- Department of Microbiology and Immunology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, People's Republic of China; Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, China Agricultural University, Beijing 100094, People's Republic of China.
| |
Collapse
|
15
|
Dai M, Xu C, Chen W, Liao M. Progress on chicken T cell immunity to viruses. Cell Mol Life Sci 2019; 76:2779-2788. [PMID: 31101935 PMCID: PMC11105491 DOI: 10.1007/s00018-019-03117-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/14/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022]
Abstract
Avian virus infection remains one of the most important threats to the poultry industry. Pathogens such as avian influenza virus (AIV), avian infectious bronchitis virus (IBV), and infectious bursal disease virus (IBDV) are normally controlled by antibodies specific for surface proteins and cellular immune responses. However, standard vaccines aimed at inducing neutralizing antibodies must be administered annually and can be rendered ineffective because immune-selective pressure results in the continuous mutation of viral surface proteins of different strains circulating from year to year. Chicken T cells have been shown to play a crucial role in fighting virus infection, offering lasting and cross-strain protection, and offer the potential for developing universal vaccines. This review provides an overview of our current knowledge of chicken T cell immunity to viruses. More importantly, we point out the limitations and barriers of current research and a potential direction for future studies.
Collapse
Affiliation(s)
- Manman Dai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
| | - Chenggang Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Weisan Chen
- T Cell Lab, Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, Australia.
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China.
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture, Guangzhou, People's Republic of China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China.
| |
Collapse
|
16
|
Xiao J, Xiang W, Zhang Y, Peng W, Zhao M, Niu L, Chai Y, Qi J, Wang F, Qi P, Pan C, Han L, Wang M, Kaufman J, Gao GF, Liu WJ. An Invariant Arginine in Common with MHC Class II Allows Extension at the C-Terminal End of Peptides Bound to Chicken MHC Class I. THE JOURNAL OF IMMUNOLOGY 2018; 201:3084-3095. [PMID: 30341185 DOI: 10.4049/jimmunol.1800611] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/11/2018] [Indexed: 12/30/2022]
Abstract
MHC molecules are found in all jawed vertebrates and are known to present peptides to T lymphocytes. In mammals, peptides can hang out either end of the peptide-binding groove of classical class II molecules, whereas the N and C termini of peptides are typically tightly bound to specific pockets in classical class I molecules. The chicken MHC, like many nonmammalian vertebrates, has a single dominantly expressed classical class I molecule encoded by the BF2 locus. We determined the structures of BF2*1201 bound to two peptides and found that the C terminus of one peptide hangs outside of the groove with a conformation much like the peptides bound to class II molecules. We found that BF2*1201 binds many peptides that hang out of the groove at the C terminus, and the sequences and structures of this MHC class I allele were determined to investigate the basis for this phenomenon. The classical class I molecules of mammals have a nearly invariant Tyr (Tyr84 in humans) that coordinates the peptide C terminus, but all classical class I molecules outside of mammals have an Arg in that position in common with mammalian class II molecules. We find that this invariant Arg residue switches conformation to allow peptides to hang out of the groove of BF2*1201, suggesting that this phenomenon is common in chickens and other nonmammalian vertebrates, perhaps allowing the single dominantly expressed class I molecule to bind a larger repertoire of peptides.
Collapse
Affiliation(s)
- Jin Xiao
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Engineering and Technology Research Center for Beijing Veterinary Peptide Vaccine Design and Preparation, Zhongmu Institutes of China Animal Husbandry Industry Co. Ltd., Beijing 100095, China.,College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Wangzhen Xiang
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Engineering and Technology Research Center for Beijing Veterinary Peptide Vaccine Design and Preparation, Zhongmu Institutes of China Animal Husbandry Industry Co. Ltd., Beijing 100095, China.,College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yongli Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Weiyu Peng
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.,College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Min Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ling Niu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Wang
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Engineering and Technology Research Center for Beijing Veterinary Peptide Vaccine Design and Preparation, Zhongmu Institutes of China Animal Husbandry Industry Co. Ltd., Beijing 100095, China
| | - Peng Qi
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Engineering and Technology Research Center for Beijing Veterinary Peptide Vaccine Design and Preparation, Zhongmu Institutes of China Animal Husbandry Industry Co. Ltd., Beijing 100095, China
| | - Chungang Pan
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Engineering and Technology Research Center for Beijing Veterinary Peptide Vaccine Design and Preparation, Zhongmu Institutes of China Animal Husbandry Industry Co. Ltd., Beijing 100095, China
| | - Lingxia Han
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Ming Wang
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Engineering and Technology Research Center for Beijing Veterinary Peptide Vaccine Design and Preparation, Zhongmu Institutes of China Animal Husbandry Industry Co. Ltd., Beijing 100095, China.,College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jim Kaufman
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, United Kingdom; .,Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 1QP, United Kingdom; and
| | - George F Gao
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; .,NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,China Research Network of Immunity and Health, Beijing Institutes of Life Science Chinese Academy of Sciences, Beijing 100101, China
| | - William J Liu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China; .,NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
17
|
Kazemi H, Najafi M, Ghasemian E, Rahimi-Mianji G, Pirsaraei ZA. Polymorphism detection of promoter region of IFN-
$$\gamma $$
γ
and IL-2 genes and their association with productive traits in Mazandaran native breeder fowls. J Genet 2018. [DOI: 10.1007/s12041-018-0981-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
18
|
Kazemi H, Najafi M, Ghasemian E, Rahimi-Mianji G, Ansari Pirsaraei Z. Polymorphism detection of promoter region of IFN-γ and IL-2 genes and their association with productive traits in Mazandaran native breeder fowls. J Genet 2018; 97:843-851. [PMID: 30262696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To identify polymorphism in interferon gamma (IFN-γ) and interleukin-2 (IL-2) genes, blood samples were collected from 380 breeder hens of the Mazandaran native fowls breeding station. DNA extraction was performed through a modified saltingout method and fragments of 670 and 659 bp from the promoter regions of IFN-γ and IL-2 genes were amplified by using specific primers, respectively. Following genotyping in the IFN-γ gene using the Tsp509I restriction enzyme, two alleles of A and G with the frequencies of 0.55 and 0.45 and three genotypes of AA, AG and GG were observed with the frequencies of 0.32, 0.46 and 0.22, respectively. For the IL-2 gene, two alleles of A and G were also detected using the MnlI restriction enzyme with the frequencies of 0.58 and 0.42 and three genotypes of AA, AG and GG with the frequencies of 0.33, 0.50 and 0.17, respectively. Statistical analysis revealed significant associations between IL-2 gene single-nucleotide polymorphism and productive traits including the average egg weight (EW) at 345-375 days of age, egg number (EN) at 345-375 days of age and body weight (BW) at 8weeks of age traits (P<0.05). Further, in a mean comparison analysis, there were also significant differences between different genotypes of the IL-2 gene in average EWat 28 and 30weeks of age, in which AG genotypes showed higher performance. Additionally, for the IFN-γ gene, a significant difference was found between the genotypes in average EW at 28 weeks of age trait. Therefore, it can be concluded that the above-mentioned polymorphisms could be considered as the pivotal geneticmakers to improveMazandaran native fowl breeding programmes to achieve the optimum performance in productive traits more efficiently.
Collapse
Affiliation(s)
- Hamed Kazemi
- Animal Breeding and Genetics, SANRU, Sari 4818168984, Iran.
| | | | | | | | | |
Collapse
|
19
|
Quantification and phenotypic characterisation of peripheral IFN-γ producing leucocytes in chickens vaccinated against Newcastle disease. Vet Immunol Immunopathol 2017; 193-194:18-28. [PMID: 29129224 PMCID: PMC5697524 DOI: 10.1016/j.vetimm.2017.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 09/28/2017] [Accepted: 10/06/2017] [Indexed: 12/04/2022]
Abstract
An avian ICS assay for detection of chIFN-γ was established. Commercially available chIFN-γ antibodies were evaluated using tranfected CHO cells. Functional T cell responses were addressed in NDV vaccination study. Circulating T cells producing IFN-γ were quantified and phenotyped by flow cytometry.
The aim of this study was to optimise and evaluate an intracellular cytokine staining (ICS) assay for assessment of T cell IFN-γ responses in chickens vaccinated against Newcastle disease (ND). We aimed to validate currently available antibodies to chicken IFN-γ using transfected CHO cells. Moreover, this ICS assay was evaluated for use to detect mitogen and antigen induced IFN-γ production in chicken peripheral blood leucocytes. Chickens from an inbred white leghorn line containing two MHC haplotypes, B19 and B21, were divided into three experimental groups; one group was kept as naive controls, one group was vaccinated intramuscularly twice with a commercial inactivated ND virus (NDV) vaccine, and the last group was vaccinated orally twice with a commercial live attenuated NDV vaccine. PBMC were ex vivo stimulated with ConA or with NDV antigen. The ICS assay was used to determine the phenotype and frequency of IFN-γ positive cells. ConA stimulation induced extensive IFN-γ production in both CD3+TCRγδ+ (γδ T cells) cells and CD3+TCRγδ− cells (αβ T cells), but no significant differences were observed between the experimental groups. Furthermore, a large proportion of the IFN-γ producing cells were CD3− indicating that other cells than classic T cells, secreted this cytokine. NDV antigen stimulation induced IFN-γ production but to a lower extent than ConA and with a large variation between individuals. The CD3+TCR1γδ−CD8α+ (CTL) population produced the highest NDV specific IFN-γ responses, with significantly elevated levels of IFN-γ producing cells in the B19 chickens vaccinated orally with live attenuated NDV vaccine. This was not the case in the B21 animals, indicating a haplotype restricted variation. In contrast, the CD3+TCR1γδ−CD4+ (Th) population did not show a significant increase in IFN-γ production in NDV stimulated samples which was in part due to a high number of IFN-γ producing cells after incubation with medium alone. In conclusion, an ICS assay for phenotyping of IFN-γ producing chicken leukocytes was set up that proved useful in identifying cytokine producing cells upon either mitogen or antigen-specific stimulation.
Collapse
|
20
|
Mucksová J, Plachý J, Staněk O, Hejnar J, Kalina J, Benešová B, Trefil P. Cytokine response to the RSV antigen delivered by dendritic cell-directed vaccination in congenic chicken lines. Vet Res 2017; 48:18. [PMID: 28381295 PMCID: PMC5382389 DOI: 10.1186/s13567-017-0423-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 01/12/2017] [Indexed: 01/05/2023] Open
Abstract
Systems of antigen delivery into antigen-presenting cells represent an important novel strategy in chicken vaccine development. In this study, we verified the ability of Rous sarcoma virus (RSV) antigens fused with streptavidin to be targeted by specific biotinylated monoclonal antibody (anti-CD205) into dendritic cells and induce virus-specific protective immunity. The method was tested in four congenic lines of chickens that are either resistant or susceptible to the progressive growth of RSV-induced tumors. Our analyses confirmed that the biot-anti-CD205-SA-FITC complex was internalized by chicken splenocytes. In the cytokine expression profile, several significant differences were evident between RSV-challenged progressor and regressor chicken lines. A significant up-regulation of IL-2, IL-12, IL-15, and IL-18 expression was detected in immunized chickens of both regressor and progressor groups. Of these cytokines, IL-2 and IL-12 were most up-regulated 14 days post-challenge (dpc), while IL-15 and IL-18 were most up-regulated at 28 dpc. On the contrary, IL-10 expression was significantly down-regulated in all immunized groups of progressor chickens at 14 dpc. We detected significant up-regulation of IL-17 in the group of immunized progressors. LITAF down-regulation with iNOS up-regulation was especially observed in the progressor group of immunized chickens that developed large tumors. Based on the increased expression of cytokines specific for activated dendritic cells, we conclude that our system is able to induce partial stimulation of specific cell types involved in cell-mediated immunity.
Collapse
Affiliation(s)
- Jitka Mucksová
- BIOPHARM, Research Institute of Biopharmacy and Veterinary Drugs, Jílové U Prahy, Czech Republic
| | - Jiří Plachý
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Ondřej Staněk
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jiří Hejnar
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jiří Kalina
- BIOPHARM, Research Institute of Biopharmacy and Veterinary Drugs, Jílové U Prahy, Czech Republic
| | - Barbora Benešová
- BIOPHARM, Research Institute of Biopharmacy and Veterinary Drugs, Jílové U Prahy, Czech Republic
| | - Pavel Trefil
- BIOPHARM, Research Institute of Biopharmacy and Veterinary Drugs, Jílové U Prahy, Czech Republic.
| |
Collapse
|
21
|
Abstract
Live attenuated viral vaccines are widely used in commercial poultry production, but the development of new effective inactivated/subunit vaccines is needed. Studies of avian antigen-specific T cells are primarily based on analyses ex vivo after activating the cells with recall antigen. There is a particular interest in developing robust high-throughput assays as chicken vaccine trials usually comprise many individuals. In many respects, the avian immune system differs from the mammalian, and T cell assessment protocols must be adjusted accordingly to account for, e.g., differences in leukocyte subsets.The carboxyfluorescein succinimidyl ester (CFSE) method described in this chapter has been adapted to chicken cells. In this test, cells of interest are stained with CFSE. The succinimidyl ester group covalently binds to cellular amines forming fluorescent conjugates that are retained in the cells even throughout division. This leads to daughter cells containing half the fluorescence of their parents. When lymphocytes are loaded with CFSE prior to ex vivo stimulation with specific antigen, the measurement of serial halving of its fluorescence by flow cytometry identifies the cells responding to the stimulation. This method has been successfully applied to studies of chicken antigen-specific T cells.
Collapse
|
22
|
Truong AD, Ban J, Park B, Hong YH, Lillehoj HS. Characterization and functional analyses of a novel chicken CD8α variant X1 (CD8α1)1,2. J Anim Sci 2016; 94:2737-51. [DOI: 10.2527/jas.2015-0133] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
23
|
Miller MM, Taylor RL. Brief review of the chicken Major Histocompatibility Complex: the genes, their distribution on chromosome 16, and their contributions to disease resistance. Poult Sci 2016; 95:375-92. [PMID: 26740135 PMCID: PMC4988538 DOI: 10.3382/ps/pev379] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/11/2015] [Indexed: 12/25/2022] Open
Abstract
Nearly all genes presently mapped to chicken chromosome 16 (GGA 16) have either a demonstrated role in immune responses or are considered to serve in immunity by reason of sequence homology with immune system genes defined in other species. The genes are best described in regional units. Among these, the best known is the polymorphic major histocompatibility complex-B (MHC-B) region containing genes for classical peptide antigen presentation. Nearby MHC-B is a small region containing two CD1 genes, which encode molecules known to bind lipid antigens and which will likely be found in chickens to present lipids to specialized T cells, as occurs with CD1 molecules in other species. Another region is the MHC-Y region, separated from MHC-B by an intervening region of tandem repeats. Like MHC-B, MHC-Y is polymorphic. It contains specialized class I and class II genes and c-type lectin-like genes. Yet another region, separated from MHC-Y by the single nucleolar organizing region (NOR) in the chicken genome, contains olfactory receptor genes and scavenger receptor genes, which are also thought to contribute to immunity. The structure, distribution, linkages and patterns of polymorphism in these regions, suggest GGA 16 evolves as a microchromosome devoted to immune defense. Many GGA 16 genes are polymorphic and polygenic. At the moment most disease associations are at the haplotype level. Roles of individual MHC genes in disease resistance are documented in only a very few instances. Provided suitable experimental stocks persist, the availability of increasingly detailed maps of GGA 16 genes combined with new means for detecting genetic variability will lead to investigations defining the contributions of individual loci and more applications for immunogenetics in breeding healthy poultry.
Collapse
Affiliation(s)
- Marcia M Miller
- Beckman Research Institute, City of Hope, Department of Molecular and Cellular Biology, Duarte, CA 91010
| | - Robert L Taylor
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV 26506
| |
Collapse
|
24
|
Tan L, Liao Y, Fan J, Zhang Y, Mao X, Sun Y, Song C, Qiu X, Meng C, Ding C. Prediction and identification of novel IBV S1 protein derived CTL epitopes in chicken. Vaccine 2015; 34:380-6. [PMID: 26620841 DOI: 10.1016/j.vaccine.2015.11.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/30/2015] [Accepted: 11/12/2015] [Indexed: 01/22/2023]
Abstract
Infectious bronchitis virus (IBV) is a major pathogen common in the poultry industry. Broad cytotoxic T lymphocyte (CTL) response against IBV is one of the crucial factors that help to control viral replication. Spike glycoproteins on the surface of the IBV virion harbor major T cell epitopes. In this study, based on the peptide-binding motifs of chicken MHC I molecules for the BF2*4, BF2*12, BF2*15, and BF2*19 haplotypes, potential CTL epitopes were predicted using S1 proteins from different IBV strains. Twenty-one peptides were predicted to be potential CTL epitopes; they were manually synthesized and the CTL responses to them tested in vitro. Spleen lymphocytes were collected from specific-pathogen free (SPF) chicken that had been immunized with the S1 protein expression plasmid, pV-S1, and were stimulated by the synthesized peptides. IFN-γ secretion and CD8(+) T cell proliferation in chickens were tested by ELISpot array and flow cytometry, respectively. Four epitopes (P8SRIQTATDP, P9SRNATGSQP, P18GAYAVVNV, and P19SRIQTATQP) were identified to stimulate CD8(+) T cell proliferation and IFN-γ secretion, indicating their efficacy as CTL epitopes in chicken. Poly-CTL-epitope DNA vaccine (pV-S1T) was constructed by inserting nucleotide sequences encoding the P8, P9, P18, and P19 CTL epitopes into the pVAX1 vector. Chickens were vaccinated with either pV-S1, pV-S1T, or pVAX1 and the protection efficacy was analyzed, revealing that ninety percent of chickens immunized with pV-S1T were protected after challenge with 10(6) ELD50 of IBV, demonstrating that these novel CTL epitopes were effective against IBV challenge. This study provides a new method to screen virus CTL epitopes in chicken and to develop poly-CTL-epitope DNA vaccines.
Collapse
Affiliation(s)
- Lei Tan
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Ying Liao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Jin Fan
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Yuqiang Zhang
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Xiang Mao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Yingjie Sun
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Cuiping Song
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Xusheng Qiu
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Chunchun Meng
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Chan Ding
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China.
| |
Collapse
|
25
|
Ji B, Sun TT, Ma ZL, Lu QF, Hu WL, Jian ZH, Jiang Y, Chen PF. Possible Association of IFN-γ Gene -316A/G SNP with Humoral Immune Response to Killed H5N1 HPAI Vaccine in a Red Junglefowl Population. J Interferon Cytokine Res 2015; 35:844-9. [PMID: 26154533 DOI: 10.1089/jir.2015.0018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To understand the role of interferon (IFN)-γ gene (IFNG) single-nucleotide polymorphisms (SNPs) in the resistance to H5N1 highly pathogenic avian influenza (HPAI), we determined the SNPs, the log2-transformed specific antibody titers, and ex vivo recall antigen-specific IFN-γ production by peripheral blood mononuclear cells (PBMCs) in 62 red junglefowls (Gallus gallus) immunized twice with inactivated H5N1 HPAI vaccine. Consequently, 52 SNPs were detected in the amplified 1137-bp length covering the promoter region and all exons, with +2133A/T SNP in the coding DNA sequence that caused a missense mutation and was identified in G. gallus for the first time. For -316A/G SNP genotypes, the chi-squared test showed that the bird sample was in the Hardy-Weinberg equilibrium (P=0.369>0.05), and the least squares analysis revealed an increasing tendency in the antibody titers with G to A substitution so that a significant difference occurred between the AA genotype (8.031±0.247) and the GG genotype (6.571±0.528) (P=0.015<0.05, 95% confidence interval [CI]: 0.0004-0.0866), as basically consistent with the antigen-induced IFN-γ protein expression, which indicated possible association of the -316A/G SNP with a secondary humoral immune response to the HPAI vaccine in the bird population. These findings may help to improve genetic resistance using cross-breeding and enhance HPAI vaccine-induced immunity in chicken production.
Collapse
Affiliation(s)
- Bin Ji
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| | - Ting-Ting Sun
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| | - Zhi-Liang Ma
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| | - Qiong-Fen Lu
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| | - Wen-Li Hu
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| | - Zong-Hui Jian
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| | - Yu Jiang
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| | - Pei-Fu Chen
- Key Lab of Veterinary Public Health of Yunnan Higher Education Institutes, Division of Veterinary Medicine, College of Animal Science and Technology, Yunnan Agricultural University , Kunming, People's Republic of China
| |
Collapse
|
26
|
Neulen ML, Viertlboeck BC, Straub C, Göbel TW. Identification of novel chicken CD4⁺ CD3⁻ blood population with NK cell like features. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 49:72-78. [PMID: 25445913 DOI: 10.1016/j.dci.2014.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/10/2014] [Accepted: 11/12/2014] [Indexed: 06/04/2023]
Abstract
Chicken NK cells have been defined in embryonic spleen and intestinal epithelium as CD8(+) lymphoid cells that lack BCR and TCR, whereas blood NK cells have not been phenotypically defined. Here we employed the mab, 8D12 directed against CHIR-AB1, a chicken Fc receptor, to define a previously uncharacterized lymphoid cell population in the blood. Although CHIR-AB1 expression was found on several cell populations, cells with extraordinary high CHIR-AB1 levels ranged between 0.4 and 2.8% in five different chicken lines. The widespread applicability of the CHIR-AB1 mab was unexpected, since CHIR-AB1-like genes form a polygenic and polymorphic subfamily. Surprisingly the CHIR-AB1 high cells coexpressed low MHCII, low CD4 and CD5, while other T cell markers CD3 and CD8, the B cell marker Bu1, the macrophage marker KUL01 were absent. Moreover, they stained with the mab 28-4, 20E5 and 1G7, which define chicken NK cells and they also expressed CD25, CD57, CD244 and the vitronectin receptor (αVβ3 integrin). In functional assays, PMA stimulation led to high levels of IFNγ release, while spontaneous cytotoxicity was not detectable. The expression of typical NK cell markers in the absence of characteristic B- or T-cell markers, and their IFNγ release is suggestive of a yet unidentified NK like population.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- CD3 Complex/immunology
- CD3 Complex/metabolism
- CD4 Antigens/immunology
- CD4 Antigens/metabolism
- CD5 Antigens/immunology
- CD5 Antigens/metabolism
- CD57 Antigens/immunology
- CD57 Antigens/metabolism
- Cells, Cultured
- Chickens/blood
- Chickens/immunology
- Enzyme-Linked Immunospot Assay
- Flow Cytometry
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-2 Receptor alpha Subunit/immunology
- Interleukin-2 Receptor alpha Subunit/metabolism
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Vitronectin/immunology
- Receptors, Vitronectin/metabolism
- Signaling Lymphocytic Activation Molecule Family
- Tetradecanoylphorbol Acetate/pharmacology
Collapse
Affiliation(s)
- Marie-Luise Neulen
- Institute for Animal Physiology, Department of Veterinary Sciences, University of Munich, Veterinärstrasse 13, 80539 Munich, Germany
| | - Birgit C Viertlboeck
- Institute for Animal Physiology, Department of Veterinary Sciences, University of Munich, Veterinärstrasse 13, 80539 Munich, Germany
| | - Christian Straub
- Institute for Animal Physiology, Department of Veterinary Sciences, University of Munich, Veterinärstrasse 13, 80539 Munich, Germany
| | - Thomas W Göbel
- Institute for Animal Physiology, Department of Veterinary Sciences, University of Munich, Veterinärstrasse 13, 80539 Munich, Germany.
| |
Collapse
|
27
|
An infected chicken kidney cell co-culture ELISpot for enhanced detection of T cell responses to avian influenza and vaccination. J Immunol Methods 2014; 416:40-8. [PMID: 25450002 PMCID: PMC4334094 DOI: 10.1016/j.jim.2014.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 09/25/2014] [Accepted: 10/24/2014] [Indexed: 11/23/2022]
Abstract
A better understanding of the immune responses of chickens to the influenza virus is essential for the development of new strategies of vaccination and control. We have developed a method incorporating infected chicken kidney cells (CKC) in culture with splenocytes in an IFNγ ELISpot assay to enumerate ex vivo responses against influenza virus antigens. Splenocytes from birds challenged with influenza showed specific responses to the influenza virus, with responding cells being mainly CD8 positive. The utility of the assay was also demonstrated in the detection of an antigen specific enhancement of IFNγ producing cells from birds vaccinated with recombinant Fowlpox vectored influenza nucleoprotein and matrix protein. Chickens infected with avian influenza developed IFNγ responses. The use of infected CKC in ELISpot overcomes limitations at detection of responses. This methods allows the quantification of influenza specific CD8 T cells. The use of recombinant virus to infect CKC can further define antigen specificity.
Collapse
|
28
|
Chen X, Yang Z, Lu Y, Xu Q, Wang Q, Chen L. Clinical features and factors associated with outcomes of patients infected with a Novel Influenza A (H7N9) virus: a preliminary study. PLoS One 2013; 8:e73362. [PMID: 24069191 PMCID: PMC3775774 DOI: 10.1371/journal.pone.0073362] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/18/2013] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The present study aimed to analyze clinical features and factors associated with treatment outcomes of H7N9 influenza A virus infection. METHODS The clinical progress in 18 H7N9-infected patients was monitored and recorded. The clinical features of H7N9 infection were noted and factors associated with treatment outcomes were analyzed by univariate analyses. RESULTS The average ages of patients in recovered and critical conditions were 67.0±10.83 years and 72.75±12.0 years, respectively. Renal insufficiency developed more frequently in critically ill patients (P = 0.023). The duration of traditional Chinese medicine (TCM) therapy was longer in recovered patients than in critically ill patients (P = 0.01). Laboratory tests showed that levels of C-reactive protein, serum creatinine, and myoglobin were significantly higher in critically ill patients than in recovered patients (P = 0.011, 0.04, and 0.016, respectively). Meanwhile, levels of all T cell subsets examined including total CD3(+), CD4(+), CD8(+), and CD45(+) T cells were lower in critically ill patients than in recovered patients (P = 0.033, 0.059, 0.015, and 0.039, respectively). Logistic regression analysis demonstrated that C-reactive protein level, myoglobin level and TCM therapy duration were likely associated with treatment outcomes of H7N9 infection (P = 0.032, 0.041 and 0.017, respectively). CONCLUSION Elderly people may have increased risk for H7N9 virus infection. T cell-mediated responses play an important role in defense against the H7N9 virus. C-reactive protein level, myoglobin level and TCM duration may be associated with treatment outcomes of H7N9 infection.
Collapse
Affiliation(s)
- Xiaorong Chen
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center Affiliated to Fudan University, Shanghai, China
- Key Laboratory of Infectious Diseases of State Administration of Traditional Chinese Medicine (Clinical base), Shanghai, China
| | - Zongguo Yang
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center Affiliated to Fudan University, Shanghai, China
- Key Laboratory of Infectious Diseases of State Administration of Traditional Chinese Medicine (Clinical base), Shanghai, China
| | - Yunfei Lu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center Affiliated to Fudan University, Shanghai, China
- Key Laboratory of Infectious Diseases of State Administration of Traditional Chinese Medicine (Clinical base), Shanghai, China
| | - Qingnian Xu
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center Affiliated to Fudan University, Shanghai, China
- Key Laboratory of Infectious Diseases of State Administration of Traditional Chinese Medicine (Clinical base), Shanghai, China
| | - Qiang Wang
- Department of Traditional Chinese Medicine, Shanghai Public Health Clinical Center Affiliated to Fudan University, Shanghai, China
| | - Liang Chen
- Department of Hepatitis, Shanghai Public Health Clinical Center Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
29
|
Szurgot I, Szolajska E, Laurin D, Lambrecht B, Chaperot L, Schoehn G, Chroboczek J. Self-adjuvanting influenza candidate vaccine presenting epitopes for cell-mediated immunity on a proteinaceous multivalent nanoplatform. Vaccine 2013; 31:4338-46. [PMID: 23880363 DOI: 10.1016/j.vaccine.2013.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/03/2013] [Accepted: 07/10/2013] [Indexed: 11/17/2022]
Abstract
We exploit the features of a virus-like particle, adenoviral dodecahedron (Ad Dd), for engineering a multivalent vaccination platform carrying influenza epitopes for cell-mediated immunity. The delivery platform, Ad Dd, is a proteinaceous, polyvalent, and biodegradable nanoparticle endowed with remarkable endocytosis activity that can be engineered to carry 60 copies of a peptide. Influenza M1 is the most abundant influenza internal protein with the conserved primary structure. Two different M1 immunodominant epitopes were separately inserted in Dd external positions without destroying the particles' dodecahedric structure. Both kinds of DdFluM1 obtained through expression in baculovirus system were properly presented by human dendritic cells triggering efficient activation of antigen-specific T cells responses. Importantly, the candidate vaccine was able to induce cellular immunity in vivo in chickens. These results warrant further investigation of Dd as a platform for candidate vaccine, able to stimulate cellular immune responses.
Collapse
Affiliation(s)
- Inga Szurgot
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02106 Warsaw, Poland.
| | | | | | | | | | | | | |
Collapse
|
30
|
Butter C, Staines K, van Hateren A, Davison TF, Kaufman J. The peptide motif of the single dominantly expressed class I molecule of the chicken MHC can explain the response to a molecular defined vaccine of infectious bursal disease virus (IBDV). Immunogenetics 2013; 65:609-18. [PMID: 23644721 PMCID: PMC3710569 DOI: 10.1007/s00251-013-0705-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 04/17/2013] [Indexed: 12/22/2022]
Abstract
In contrast to typical mammals, the chicken MHC (the BF-BL region of the B locus) has strong genetic associations with resistance and susceptibility to infectious pathogens as well as responses to vaccines. We have shown that the chicken MHC encodes a single dominantly expressed class I molecule whose peptide-binding motifs can determine resistance to viral pathogens, such as Rous sarcoma virus and Marek’s disease virus. In this report, we examine the response to a molecular defined vaccine, fp-IBD1, which consists of a fowlpox virus vector carrying the VP2 gene of infectious bursal disease virus (IBDV) fused with β-galactosidase. We vaccinated parental lines and two backcross families with fp-IBD1, challenged with the virulent IBDV strain F52/70, and measured damage to the bursa. We found that the MHC haplotype B15 from line 15I confers no protection, whereas B2 from line 61 and B12 from line C determine protection, although another locus from line 61 was also important. Using our peptide motifs, we found that many more peptides from VP2 were predicted to bind to the dominantly expressed class I molecule BF2*1201 than BF2*1501. Moreover, most of the peptides predicted to bind BF2*1201 did in fact bind, while none bound BF2*1501. Using peptide vaccination, we identified one B12 peptide that conferred protection to challenge, as assessed by bursal damage and viremia. Thus, we show the strong genetic association of the chicken MHC to a T cell vaccine can be explained by peptide presentation by the single dominantly expressed class I molecule.
Collapse
Affiliation(s)
- Colin Butter
- Institute for Animal Health, Compton, Reading, Berkshire RG20 7NN UK
- The Pirbright Institute, Compton Laboratory, Compton, RG20 7NN UK
| | - Karen Staines
- Institute for Animal Health, Compton, Reading, Berkshire RG20 7NN UK
- The Pirbright Institute, Compton Laboratory, Compton, RG20 7NN UK
| | - Andrew van Hateren
- Institute for Animal Health, Compton, Reading, Berkshire RG20 7NN UK
- Institute for Life Science, University of Southampton, Building 85, M55, Southampton, SO17 1BJ UK
| | - T. Fred Davison
- Institute for Animal Health, Compton, Reading, Berkshire RG20 7NN UK
| | - Jim Kaufman
- Institute for Animal Health, Compton, Reading, Berkshire RG20 7NN UK
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP UK
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES UK
| |
Collapse
|
31
|
Towards a universal vaccine for avian influenza: protective efficacy of modified Vaccinia virus Ankara and Adenovirus vaccines expressing conserved influenza antigens in chickens challenged with low pathogenic avian influenza virus. Vaccine 2012. [PMID: 23200938 PMCID: PMC3605591 DOI: 10.1016/j.vaccine.2012.11.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Current vaccines targeting surface proteins can drive antigenic variation resulting either in the emergence of more highly pathogenic viruses or of antigenically distinct viruses that escape control by vaccination and thereby persist in the host population. Influenza vaccines typically target the highly mutable surface proteins and do not provide protection against heterologous challenge. Vaccines which induce immune responses against conserved influenza epitopes may confer protection against heterologous challenge. We report here the results of vaccination with recombinant modified Vaccinia virus Ankara (MVA) and Adenovirus (Ad) expressing a fusion construct of nucleoprotein and matrix protein (NP + M1). Prime and boost vaccination regimes were trialled in different ages of chicken and were found to be safe and immunogenic. Interferon-γ (IFN-γ) ELISpot was used to assess the cellular immune response post secondary vaccination. In ovo Ad prime followed by a 4 week post hatch MVA boost was identified as the most immunogenic regime in one outbred and two inbred lines of chicken. Following vaccination, one inbred line (C15I) was challenged with low pathogenic avian influenza (LPAI) H7N7 (A/Turkey/England/1977). Birds receiving a primary vaccination with Ad-NP + M1 and a secondary vaccination with MVA-NP + M1 exhibited reduced cloacal shedding as measured by plaque assay at 7 days post infection compared with birds vaccinated with recombinant viruses containing irrelevant antigen. This preliminary indication of efficacy demonstrates proof of concept in birds; induction of T cell responses in chickens by viral vectors containing internal influenza antigens may be a productive strategy for the development of vaccines to induce heterologous protection against influenza in poultry.
Collapse
|
32
|
Geus EDD, Rebel JM, Vervelde L. Induction of respiratory immune responses in the chicken; implications for development of mucosal avian influenza virus vaccines. Vet Q 2012; 32:75-86. [DOI: 10.1080/01652176.2012.711956] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|