1
|
Bliss CM, Nachbagauer R, Mariottini C, Cuevas F, Feser J, Naficy A, Bernstein DI, Guptill J, Walter EB, Berlanda-Scorza F, Innis BL, García-Sastre A, Palese P, Krammer F, Coughlan L. A chimeric haemagglutinin-based universal influenza virus vaccine boosts human cellular immune responses directed towards the conserved haemagglutinin stalk domain and the viral nucleoprotein. EBioMedicine 2024; 104:105153. [PMID: 38805853 PMCID: PMC11154122 DOI: 10.1016/j.ebiom.2024.105153] [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: 10/25/2023] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The development of a universal influenza virus vaccine, to protect against both seasonal and pandemic influenza A viruses, is a long-standing public health goal. The conserved stalk domain of haemagglutinin (HA) is a promising vaccine target. However, the stalk is immunosubdominant. As such, innovative approaches are required to elicit robust immunity against this domain. In a previously reported observer-blind, randomised placebo-controlled phase I trial (NCT03300050), immunisation regimens using chimeric HA (cHA)-based immunogens formulated as inactivated influenza vaccines (IIV) -/+ AS03 adjuvant, or live attenuated influenza vaccines (LAIV), elicited durable HA stalk-specific antibodies with broad reactivity. In this study, we sought to determine if these vaccines could also boost T cell responses against HA stalk, and nucleoprotein (NP). METHODS We measured interferon-γ (IFN-γ) responses by Enzyme-Linked ImmunoSpot (ELISpot) assay at baseline, seven days post-prime, pre-boost and seven days post-boost following heterologous prime:boost regimens of LAIV and/or adjuvanted/unadjuvanted IIV-cHA vaccines. FINDINGS Our findings demonstrate that immunisation with adjuvanted cHA-based IIVs boost HA stalk-specific and NP-specific T cell responses in humans. To date, it has been unclear if HA stalk-specific T cells can be boosted in humans by HA-stalk focused universal vaccines. Therefore, our study will provide valuable insights for the design of future studies to determine the precise role of HA stalk-specific T cells in broad protection. INTERPRETATION Considering that cHA-based vaccines also elicit stalk-specific antibodies, these data support the further clinical advancement of cHA-based universal influenza vaccine candidates. FUNDING This study was funded in part by the Bill and Melinda Gates Foundation (BMGF).
Collapse
Affiliation(s)
- Carly M Bliss
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Cancer & Genetics and Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Mariottini
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frans Cuevas
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jodi Feser
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Abdi Naficy
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - David I Bernstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffrey Guptill
- Duke Early Phase Clinical Research Unit, Duke Clinical Research Institute, Durham, NC, USA
| | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Bruce L Innis
- Center for Vaccine Innovation and Access, PATH, Seattle, WA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; University of Maryland School of Medicine, Department of Microbiology and Immunology, Baltimore, MD 21201, USA; University of Maryland School of Medicine, Center for Vaccine Development and Global Health (CVD), Baltimore, MD 21201, USA.
| |
Collapse
|
2
|
Pascha MN, Ballegeer M, Roelofs MC, Meuris L, Albulescu IC, van Kuppeveld FJM, Hurdiss DL, Bosch BJ, Zeev-Ben-Mordehai T, Saelens X, de Haan CAM. Nanoparticle display of neuraminidase elicits enhanced antibody responses and protection against influenza A virus challenge. NPJ Vaccines 2024; 9:97. [PMID: 38821988 PMCID: PMC11143307 DOI: 10.1038/s41541-024-00891-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 05/20/2024] [Indexed: 06/02/2024] Open
Abstract
Current Influenza virus vaccines primarily induce antibody responses against variable epitopes in hemagglutinin (HA), necessitating frequent updates. However, antibodies against neuraminidase (NA) can also confer protection against influenza, making NA an attractive target for the development of novel vaccines. In this study, we aimed to enhance the immunogenicity of recombinant NA antigens by presenting them multivalently on a nanoparticle carrier. Soluble tetrameric NA antigens of the N1 and N2 subtypes, confirmed to be correctly folded by cryo-electron microscopy structural analysis, were conjugated to Mi3 self-assembling protein nanoparticles using the SpyTag-SpyCatcher system. Immunization of mice with NA-Mi3 nanoparticles induced higher titers of NA-binding and -inhibiting antibodies and improved protection against a lethal challenge compared to unconjugated NA. Additionally, we explored the co-presentation of N1 and N2 antigens on the same Mi3 particles to create a mosaic vaccine candidate. These mosaic nanoparticles elicited antibody titers that were similar or superior to the homotypic nanoparticles and effectively protected against H1N1 and H3N2 challenge viruses. The NA-Mi3 nanoparticles represent a promising vaccine candidate that could complement HA-directed approaches for enhanced potency and broadened protection against influenza A virus.
Collapse
Affiliation(s)
- M N Pascha
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - M Ballegeer
- VIB Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium
| | - M C Roelofs
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - L Meuris
- VIB Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium
| | - I C Albulescu
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - F J M van Kuppeveld
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - D L Hurdiss
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - B J Bosch
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - T Zeev-Ben-Mordehai
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - X Saelens
- VIB Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium.
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium.
| | - C A M de Haan
- Section of Virology, Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Decker CH, Rapier-Sharman N, Pickett BE. Mutation in Hemagglutinin Antigenic Sites in Influenza A pH1N1 Viruses from 2015-2019 in the United States Mountain West, Europe, and the Northern Hemisphere. Genes (Basel) 2022; 13:909. [PMID: 35627294 PMCID: PMC9141826 DOI: 10.3390/genes13050909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
H1N1 influenza A virus is a respiratory pathogen that undergoes antigenic shift and antigenic drift to improve viral fitness. Tracking the evolutionary trends of H1N1 aids with the current detection and the future response to new viral strains as they emerge. Here, we characterize antigenic drift events observed in the hemagglutinin (HA) sequence of the pandemic H1N1 lineage from 2015-2019. We observed the substitutions S200P, K147N, and P154S, together with other mutations in structural, functional, and/or epitope regions in 2015-2019 HA protein sequences from the Mountain West region of the United States, the larger United States, Europe, and other Northern Hemisphere countries. We reconstructed multiple phylogenetic trees to track the relationships and spread of these mutations and tested for evidence of selection pressure on HA. We found that the prevalence of amino acid substitutions at positions 147, 154, 159, 200, and 233 significantly changed throughout the studied geographical regions between 2015 and 2019. We also found evidence of coevolution among a subset of these amino acid substitutions. The results from this study could be relevant for future epidemiological tracking and vaccine prediction efforts. Similar analyses in the future could identify additional sequence changes that could affect the pathogenicity and/or infectivity of this virus in its human host.
Collapse
Affiliation(s)
| | | | - Brett E. Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA; (C.H.D.); (N.R.-S.)
| |
Collapse
|
4
|
Noisumdaeng P, Roytrakul T, Prasertsopon J, Pooruk P, Lerdsamran H, Assanasen S, Kitphati R, Auewarakul P, Puthavathana P. T cell mediated immunity against influenza H5N1 nucleoprotein, matrix and hemagglutinin derived epitopes in H5N1 survivors and non-H5N1 subjects. PeerJ 2021; 9:e11021. [PMID: 33854839 PMCID: PMC7955671 DOI: 10.7717/peerj.11021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
Background Protection against the influenza virus by a specific antibody is relatively strain specific; meanwhile broader immunity may be conferred by cell-mediated immune response to the conserved epitopes across influenza virus subtypes. A universal broad-spectrum influenza vaccine which confronts not only seasonal influenza virus, but also avian influenza H5N1 virus is promising. Methods This study determined the specific and cross-reactive T cell responses against the highly pathogenic avian influenza A (H5N1) virus in four survivors and 33 non-H5N1 subjects including 10 H3N2 patients and 23 healthy individuals. Ex vivo IFN-γ ELISpot assay using overlapping peptides spanning the entire nucleoprotein (NP), matrix (M) and hemagglutinin (HA) derived from A/Thailand/1(KAN-1)/2004 (H5N1) virus was employed in adjunct with flow cytometry for determining T cell functions. Microneutralization (microNT) assay was performed to determine the status of previous H5N1 virus infection. Results IFN-γ ELISpot assay demonstrated that survivors nos. 1 and 2 had markedly higher T cell responses against H5N1 NP, M and HA epitopes than survivors nos. 3 and 4; and the magnitude of T cell responses against NP were higher than that of M and HA. Durability of the immunoreactivity persisted for as long as four years after disease onset. Upon stimulation by NP in IFN-γ ELISpot assay, 60% of H3N2 patients and 39% of healthy subjects exhibited a cross-reactive T cell response. The higher frequency and magnitude of responses in H3N2 patients may be due to blood collection at the convalescent phase of the patients. In H5N1 survivors, the effector peptide-specific T cells generated from bulk culture PBMCs by in vitro stimulation displayed a polyfunction by simultaneously producing IFN-γ and TNF-α, together with upregulation of CD107a in recognition of the target cells pulsed with peptide or infected with rVac-NP virus as investigated by flow cytometry. Conclusions This study provides an insight into the better understanding on the homosubtypic and heterosubtypic T cell-mediated immune responses in H5N1 survivors and non-H5N1 subjects. NP is an immunodominant target of cross-recognition owing to its high conservancy. Therefore, the development of vaccine targeting the conserved NP may be a novel strategy for influenza vaccine design.
Collapse
Affiliation(s)
- Pirom Noisumdaeng
- Faculty of Public Health, Thammasat University, Khlong Luang, Pathum Thani, Thailand.,Thammasat University Research Unit in Modern Microbiology and Public Health Genomics, Thammasat University, Khlong Luang, Pathum Thani, Thailand.,Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand
| | - Thaneeya Roytrakul
- National Center for Genetic Engineering and Biotechnology, Khlong Luang, Pathum Thani, Thailand
| | - Jarunee Prasertsopon
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Phisanu Pooruk
- The Government Pharmaceutical Organization, Biological Product Vaccine Production Plant, Kaengkhoi, Saraburi, Thailand
| | - Hatairat Lerdsamran
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Susan Assanasen
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand
| | | | - Prasert Auewarakul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand
| | - Pilaipan Puthavathana
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-noi, Bangkok, Thailand.,Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
5
|
Topham DJ, DeDiego ML, Nogales A, Sangster MY, Sant A. Immunity to Influenza Infection in Humans. Cold Spring Harb Perspect Med 2021; 11:a038729. [PMID: 31871226 PMCID: PMC7919402 DOI: 10.1101/cshperspect.a038729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review discusses the human immune responses to influenza infection with some insights from studies using animal models, such as experimental infection of mice. Recent technological advances in the study of human immune responses have greatly added to our knowledge of the infection and immune responses, and therefore much of the focus is on recent studies that have moved the field forward. We consider the complexity of the adaptive response generated by many sequential encounters through infection and vaccination.
Collapse
Affiliation(s)
- David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Marta L DeDiego
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Cientificas, 28049 Madrid, Spain
| | - Aitor Nogales
- Instituto Nacional de Investigación y Tecnologia Agraria y Ailmentaria, 28040 Madrid, Spain
| | - Mark Y Sangster
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Andrea Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
6
|
T Cell Immunity against Influenza: The Long Way from Animal Models Towards a Real-Life Universal Flu Vaccine. Viruses 2021; 13:v13020199. [PMID: 33525620 PMCID: PMC7911237 DOI: 10.3390/v13020199] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Current flu vaccines rely on the induction of strain-specific neutralizing antibodies, which leaves the population vulnerable to drifted seasonal or newly emerged pandemic strains. Therefore, universal flu vaccine approaches that induce broad immunity against conserved parts of influenza have top priority in research. Cross-reactive T cell responses, especially tissue-resident memory T cells in the respiratory tract, provide efficient heterologous immunity, and must therefore be a key component of universal flu vaccines. Here, we review recent findings about T cell-based flu immunity, with an emphasis on tissue-resident memory T cells in the respiratory tract of humans and different animal models. Furthermore, we provide an update on preclinical and clinical studies evaluating T cell-evoking flu vaccines, and discuss the implementation of T cell immunity in real-life vaccine policies.
Collapse
|
7
|
Wang Q, Sun Z, Li J, Qin T, Ma H, Chen S, Peng D, Liu X. Identification of a universal antigen epitope of influenza A virus using peptide microarray. BMC Vet Res 2021; 17:22. [PMID: 33413356 PMCID: PMC7792037 DOI: 10.1186/s12917-020-02725-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/15/2020] [Indexed: 02/08/2023] Open
Abstract
Background Hemagglutinin is a major surface protein in influenza A virus (IAV), and HA2 is relative conserved among different IAVs. It will be meaningful to identify broad-spectrum epitopes based on the HA2 protein. Results Overlapping peptides of the HA2 protein of the H5N1 IAV A/Mallard/Huadong/S/2005 were synthesized and loaded on modified silica gel film to form a microarray, and antisera against different subtypes of IAVs were used to screen universal epitopes. The selected epitope was further confirmed by western blotting using anti-peptide immune serum and viruses rescued with amino acid substitution. The results showed that 485-FYHKCDNECME-495 of the H5 14th peptide in HA2 had broad-spectrum binding activity with antisera against H1, H3, H4, H5, H6, H7, H8, H9, and H10 subtype IAV. Substitution of amino acids (K or D) in rescued viruses resulted in decreased serum binding, indicating that they were critical residues for serum binding activity. In Immune Epitope Database, some epitopes containing 14–4 peptide were confirmed as MHC-II-restricted CD4 T cell epitope and had effects on releasing IL-2 or IFN. Conclusion The identified epitope should be a novel universal target for detection and vaccine design and its ability to generate immune protection needs further exploration. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-020-02725-5.
Collapse
Affiliation(s)
- Qiuxia Wang
- College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China.,Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Zhihao Sun
- College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China.,Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Jingzhi Li
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215000, People's Republic of China
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China.,Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Hongwei Ma
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215000, People's Republic of China
| | - Sujuan Chen
- College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, People's Republic of China. .,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China. .,Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, 225009, Jiangsu, People's Republic of China. .,Joint Laboratory Safety of International Cooperation of Agriculture & Agricultural-Products, Yangzhou, Jiangsu, 225009, People's Republic of China.
| | - Daxin Peng
- College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, People's Republic of China. .,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China. .,Jiangsu Research Centre of Engineering and Technology for Prevention and Control of Poultry Disease, Yangzhou, 225009, Jiangsu, People's Republic of China. .,Joint Laboratory Safety of International Cooperation of Agriculture & Agricultural-Products, Yangzhou, Jiangsu, 225009, People's Republic of China.
| | - Xiufan Liu
- College of Veterinary Medicine, Yangzhou University, 48 East Wenhui Road, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China.,Joint Laboratory Safety of International Cooperation of Agriculture & Agricultural-Products, Yangzhou, Jiangsu, 225009, People's Republic of China
| |
Collapse
|
8
|
Recombinant HA-based vaccine outperforms split and subunit vaccines in elicitation of influenza-specific CD4 T cells and CD4 T cell-dependent antibody responses in humans. NPJ Vaccines 2020; 5:77. [PMID: 32884842 PMCID: PMC7450042 DOI: 10.1038/s41541-020-00227-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Although traditional egg-based inactivated influenza vaccines can protect against infection, there have been significant efforts to develop improved formats to overcome disadvantages of this platform. Here, we have assessed human CD4 T cell responses to a traditional egg-based influenza vaccine with recently available cell-derived vaccines and recombinant baculovirus-derived vaccines. Adults were administered either egg-derived Fluzone®, mammalian cell-derived Flucelvax® or recombinant HA (Flublok®). CD4 T cell responses to each HA protein were assessed by cytokine EliSpot and intracellular staining assays. The specificity and magnitude of antibody responses were quantified by ELISA and HAI assays. By all criteria, Flublok vaccine exhibited superior performance in eliciting both CD4 T cell responses and HA-specific antibody responses, whether measured by mean response magnitude or percent of responders. Although the mechanism(s) underlying this advantage is not yet clear, it is likely that both qualitative and quantitative features of the vaccines impact the response.
Collapse
|
9
|
Sant AJ. The Way Forward: Potentiating Protective Immunity to Novel and Pandemic Influenza Through Engagement of Memory CD4 T Cells. J Infect Dis 2020; 219:S30-S37. [PMID: 30715376 PMCID: PMC6452298 DOI: 10.1093/infdis/jiy666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Potentially pandemic strains of influenza pose an undeniable threat to human populations. Therefore, it is essential to develop better strategies to enhance vaccine design and predict parameters that identify susceptible humans. CD4 T cells are a central component of protective immunity to influenza, delivering direct effector function and potentiating responses of other lymphoid cells. Humans have highly diverse influenza-specific CD4 T-cell populations that vary in stimulation history, specificity, and functionality. These complexities constitute a formidable obstacle to predicting immune responses to pandemic strains of influenza and derivation of optimal vaccine strategies. We suggest that more precise efforts to identify and enumerate both the positive and negative contributors of immunity in the CD4 T-cell compartment will aid in both predicting susceptible hosts and in development of vaccination strategies that will poise most human subjects to respond to pandemic influenza strains with protective immune responses.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, New York
| |
Collapse
|
10
|
Xu W, Weese JS, Ojkic D, Lung O, Handel K, Berhane Y. Phylogenetic Inference of H3N2 Canine Influenza A Outbreak in Ontario, Canada in 2018. Sci Rep 2020; 10:6309. [PMID: 32286409 PMCID: PMC7156495 DOI: 10.1038/s41598-020-63278-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/12/2020] [Indexed: 11/09/2022] Open
Abstract
The first Canadian H3N2 canine influenza A outbreak involving an Asian-origin H3N2 canine influenza virus (CIV) began in southwestern Ontario, Canada, in late December 2017. More H3N2 CIV cases were identified in central and eastern Ontario between March and October 2018. Based on epidemiological investigation, 5 clusters were identified (C1, C2, C3a, C3b, and C4); however, the origin of infection has only been revealed for epidemiological cluster C1. Here, we use phylogenetic analyses to unravel the links of virus transmission between the 5 epidemiological clusters and the origin of infection for all epidemiological clusters. Our results demonstrate that the Canadian H3N2 CIV sequences were grouped into four distinct phylogenetic clusters with minimal genetic diversity between these clusters. Large scale phylogenetic analysis of H3N2 CIV from around the globe showed that the Canadian CIVs formed a distinct new clade along with CIVs that have been circulating in the USA since 2017-2018 and in China since 2017. This clade shares a common ancestor of Asian origin. This study concludes that the H3N2 CIV outbreak in Ontario was driven by multiple introductions of South Korean/Chinese-origin H3N2 CIVs over 10 months.
Collapse
Affiliation(s)
- Wanhong Xu
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3M4, Canada
| | - J Scott Weese
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Davor Ojkic
- Animal Health Laboratory, University of Guelph, Guelph, Ontario, Canada
| | - Oliver Lung
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3M4, Canada
| | - Katherine Handel
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3M4, Canada
| | - Yohannes Berhane
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3M4, Canada. .,Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
11
|
Yang S, Li W, Fan Z, Zhai L, Chen J, Xiao X, Ma J, Song B, Ma J, Tong C, Yu L, Yu Y, Cao W, Cui Y. Identification of CD4 + T cell epitopes on glyceraldehyde-3-phosphate dehydrogenase-C of Staphylococcus aureus in Babl/c mice. Microb Pathog 2020; 144:104167. [PMID: 32222538 DOI: 10.1016/j.micpath.2020.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 11/15/2022]
Abstract
Glyceraldehyde-3-phosphate dehydrogenase-C (GapC) is a highly conserved surface protein of Staphylococcus aureus, with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activity, which represents an excellent vaccine candidate antigen. It can induce protective immune responses to S. aureus infections. However, CD4+ T cell epitopes of GapC that induce CD4+ T cell immune responses are currently unclear. In this study, we used bioinformatics prediction algorithms to predict CD4+ T cell epitopes of GapC. Ten peptides were synthesized to investigate the candidate epitopes. Our results showed that the peptides, G4 (GapC 104-123) and G10 (GapC 314-333) were able to induce proliferation of CD4+ T cells and secrete high levels of interferon (IFN)-γ, respectively. In addition, they significantly reduced bacterial loads in tissue and induced immunoprotective effects. It is suggested that G4 and G10 are Th1-type epitopes of S. aureus GapC. This study provides the potential development of the design of epitope-based vaccine against S. aureus.
Collapse
Affiliation(s)
- Siyu Yang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Wanyu Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Zhaowei Fan
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Lu Zhai
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jing Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Xue Xiao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jun Ma
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Baifen Song
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Jinzhu Ma
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Liquan Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Yongzhong Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Weifan Cao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Yudong Cui
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China; College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| |
Collapse
|
12
|
Ryt-Hansen P, Pedersen AG, Larsen I, Kristensen CS, Krog JS, Wacheck S, Larsen LE. Substantial Antigenic Drift in the Hemagglutinin Protein of Swine Influenza A Viruses. Viruses 2020; 12:E248. [PMID: 32102230 PMCID: PMC7077184 DOI: 10.3390/v12020248] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/16/2022] Open
Abstract
The degree of antigenic drift in swine influenza A viruses (swIAV) has historically been regarded as minimal compared to that of human influenza A virus strains. However, as surveillance activities on swIAV have increased, more isolates have been characterized, revealing a high level of genetic and antigenic differences even within the same swIAV lineage. The objective of this study was to investigate the level of genetic drift in one enzootically infected swine herd over one year. Nasal swabs were collected monthly from sows (n = 4) and piglets (n = 40) in the farrowing unit, and from weaners (n = 20) in the nursery. Virus from 1-4 animals were sequenced per month. Analyses of the sequences revealed that the hemagglutinin (HA) gene was the main target for genetic drift with a substitution rate of 7.6 × 10-3 substitutions/site/year and evidence of positive selection. The majority of the mutations occurred in the globular head of the HA protein and in antigenic sites. The phylogenetic tree of the HA sequences displayed a pectinate typology, where only a single lineage persists and forms the ancestor for subsequent lineages. This was most likely caused by repeated selection of a single immune-escape variant, which subsequently became the founder of the next wave of infections.
Collapse
Affiliation(s)
- Pia Ryt-Hansen
- National Veterinary Institute, Technical University of Denmark, Kemitorvet Building 204, DK-2800 Kongens Lyngby, Denmark
- Dpt. of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, DK-1870 Frederiksberg C, Denmark; (I.L.); (L.E.L.)
| | - Anders Gorm Pedersen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Kemitorvet Building 208, DK-2800 Kongens Lyngby, Denmark;
| | - Inge Larsen
- Dpt. of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, DK-1870 Frederiksberg C, Denmark; (I.L.); (L.E.L.)
| | | | - Jesper Schak Krog
- Statens Serum Institut, Artillerivej 5, DK-2300 Copenhagen S, Denmark;
| | - Silke Wacheck
- Ceva Santé Animale 10 Avenue de la Ballastière, 33500 Libourne, France;
| | - Lars Erik Larsen
- Dpt. of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, DK-1870 Frederiksberg C, Denmark; (I.L.); (L.E.L.)
| |
Collapse
|
13
|
Heterologous viral protein interactions within licensed seasonal influenza virus vaccines. NPJ Vaccines 2020; 5:3. [PMID: 31934357 PMCID: PMC6954117 DOI: 10.1038/s41541-019-0153-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 10/16/2019] [Indexed: 01/10/2023] Open
Abstract
Currently, licensed influenza virus vaccines are designed and tested only for their ability to elicit hemagglutinin (HA)-reactive, neutralizing antibodies. Despite this, the purification process in vaccine manufacturing often does not completely remove other virion components. In the studies reported here, we have examined the viral protein composition of a panel of licensed vaccines from different manufacturers and licensed in different years. Using western blotting, we found that, beyond HA proteins, there are detectable quantities of neuraminidase (NA), nucleoprotein (NP), and matrix proteins (M1) from both influenza A and influenza B viruses in the vaccines but that the composition differed by source and method of vaccine preparation. We also found that disparities in viral protein composition were associated with distinct patterns of elicited antibody specificities. Strikingly, our studies also revealed that many viral proteins contained in the vaccine form heterologous complexes. When H1 proteins were isolated by immunoprecipitation, NA (N1), M1 (M1-A), H3, and HA-B proteins were co-isolated with the H1. Further biochemical studies suggest that these interactions persist for at least 4 h at 37 °C and that the membrane/intracytoplasmic domains in the intact HA proteins are important for the intermolecular interactions detected. These studies indicate that, if such interactions persist after vaccines reach the draining lymph node, both dendritic cells and HA-specific B cells may take up multiple viral proteins simultaneously. Whether these interactions are beneficial or harmful to the developing immune response will depend on the functional potential of the elicited virus-specific CD4 T cells. Licensed influenza virus vaccines are evaluated for their ability to elicit neutralizing antibodies specific for hemagglutinin (HA), but the manufacturing process does not completely exclude other virion components from the formulations. Andrea Sant and colleagues now report the presence of several viral proteins, such as M1, NA, H3, and HA-B, in licensed formulations from different manufacturers and spanning stocks from several years. These viral proteins form heterologous complexes, and immunization of mice with some of the formulations analyzed elicited antibody responses specific to these viral proteins. These findings reveal heterogeneity across licensed influenza virus vaccine formulations, potentially due to variations in production processes, and raise the possibility that the presence of these additional viral protein complexes could influence the elicited immune responses following immunization, particularly in the context of multivalent strategies involving mixing of different formulations.
Collapse
|
14
|
Ryt-Hansen P, Pedersen AG, Larsen I, Krog JS, Kristensen CS, Larsen LE. Acute Influenza A virus outbreak in an enzootic infected sow herd: Impact on viral dynamics, genetic and antigenic variability and effect of maternally derived antibodies and vaccination. PLoS One 2019; 14:e0224854. [PMID: 31725751 PMCID: PMC6855628 DOI: 10.1371/journal.pone.0224854] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Influenza A virus (IAV) is a highly contagious pathogen in pigs. Swine IAV (swIAV) infection causes respiratory disease and is thereby a challenge for animal health, animal welfare and the production economy. In Europe, the most widespread strategy for controlling swIAV is implementation of sow vaccination programs, to secure delivery of protective maternally derived antibodies (MDAs) to the newborn piglets. In this study we report a unique case, where a persistently swIAV (A/sw/Denmark/P5U4/2016(H1N1)) infected herd experienced an acute outbreak with a new swIAV subtype (A/sw/Denmark/HB4280U1/2017(H1N2)) and subsequently decided to implement a mass sow vaccination program. Clinical registrations, nasal swabs and blood samples were collected from four different batches of pigs before and after vaccination. Virus isolation, sequencing of the virus strain and hemagglutinin inhibition (HI) tests were performed on samples collected before and during the outbreak and after implementation of mass sow vaccination. After implementation of the sow mass vaccination, the time of infection was delayed and the viral load significantly decreased. An increased number of pigs, however, tested positive at two consecutive sampling times indicating prolonged shedding. In addition, a significantly smaller proportion of the 10–12 weeks old pigs were seropositive by the end of the study, indicating an impaired induction of antibodies against swIAV in the presence of MDAs. Sequencing of the herd strains revealed major differences in the hemagglutinin gene of the strain isolated before- and during the acute outbreak despite that, the two strains belonged to the same HA lineage. The HI tests confirmed a limited degree of cross-reaction between the two strains. Furthermore, the sequencing results of the hemagglutinin gene obtained before and after implementation of mass sow vaccination revealed an increased substitution rate and an increase in positively selected sites in the globular head of the hemagglutinin after vaccination.
Collapse
Affiliation(s)
- Pia Ryt-Hansen
- National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
- * E-mail:
| | - Anders Gorm Pedersen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Inge Larsen
- University of Copenhagen, Dpt. of Veterinary and Animal Sciences, Frederiksberg C, Denmark
| | - Jesper Schak Krog
- National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Lars Erik Larsen
- National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
- University of Copenhagen, Dpt. of Veterinary and Animal Sciences, Frederiksberg C, Denmark
| |
Collapse
|
15
|
Pizzolla A, Wakim LM. Memory T Cell Dynamics in the Lung during Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:374-381. [PMID: 30617119 DOI: 10.4049/jimmunol.1800979] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/08/2018] [Indexed: 01/06/2023]
Abstract
Influenza A virus is highly contagious, infecting 5-15% of the global population every year. It causes significant morbidity and mortality, particularly among immunocompromised and at-risk individuals. Influenza virus is constantly evolving, undergoing continuous, rapid, and unpredictable mutation, giving rise to novel viruses that can escape the humoral immunity generated by current influenza virus vaccines. Growing evidence indicates that influenza-specific T cells resident along the respiratory tract are highly effective at providing potent and rapid protection against this inhaled pathogen. As these T cells recognize fragments of the virus that are highly conserved and less prone to mutation, they have the potential to provide cross-strain protection against a wide breadth of influenza viruses, including newly emerging strains. In this review, we will discuss how influenza-specific memory T cells in the lung are established and maintained and how we can harness this knowledge to design broadly protective influenza A virus vaccines.
Collapse
Affiliation(s)
- Angela Pizzolla
- Department of Microbiology and Immunology, University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| |
Collapse
|
16
|
Jansen JM, Gerlach T, Elbahesh H, Rimmelzwaan GF, Saletti G. Influenza virus-specific CD4+ and CD8+ T cell-mediated immunity induced by infection and vaccination. J Clin Virol 2019; 119:44-52. [DOI: 10.1016/j.jcv.2019.08.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/13/2023]
|
17
|
Asthagiri Arunkumar G, McMahon M, Pavot V, Aramouni M, Ioannou A, Lambe T, Gilbert S, Krammer F. Vaccination with viral vectors expressing NP, M1 and chimeric hemagglutinin induces broad protection against influenza virus challenge in mice. Vaccine 2019; 37:5567-5577. [PMID: 31399277 PMCID: PMC6717082 DOI: 10.1016/j.vaccine.2019.07.095] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/06/2019] [Accepted: 07/29/2019] [Indexed: 12/25/2022]
Abstract
Seasonal influenza virus infections cause significant morbidity and mortality every year. Annual influenza virus vaccines are effective but only when well matched with circulating strains. Therefore, there is an urgent need for better vaccines that induce broad protection against drifted seasonal and emerging pandemic influenza viruses. One approach to design such vaccines is based on targeting conserved regions of the influenza virus hemagglutinin. Sequential vaccination with chimeric hemagglutinin constructs can refocus antibody responses towards the conserved immunosubdominant stalk domain of the hemagglutinin, rather than the variable immunodominant head. A complementary approach for a universal influenza A virus vaccine is to induce T-cell responses to conserved internal influenza virus antigens. For this purpose, replication deficient recombinant viral vectors based on Chimpanzee Adenovirus Oxford 1 and Modified Vaccinia Ankara virus are used to express the viral nucleoprotein and the matrix protein 1. In this study, we combined these two strategies and evaluated the efficacy of viral vectors expressing both chimeric hemagglutinin and nucleoprotein plus matrix protein 1 in a mouse model against challenge with group 2 influenza viruses including H3N2, H7N9 and H10N8. We found that vectored vaccines expressing both sets of antigens provided enhanced protection against H3N2 virus challenge when compared to vaccination with viral vectors expressing only one set of antigens. Vaccine induced antibody responses against divergent group 2 hemagglutinins, nucleoprotein and matrix protein 1 as well as robust T-cell responses to the nucleoprotein and matrix protein 1 were detected. Of note, it was observed that while antibodies to the H3 stalk were already boosted to high levels after two vaccinations with chimeric hemagglutinins (cHAs), three exposures were required to induce strong reactivity across subtypes. Overall, these results show that a combinations of different universal influenza virus vaccine strategies can induce broad antibody and T-cell responses and can provide increased protection against influenza.
Collapse
Affiliation(s)
- Guha Asthagiri Arunkumar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Meagan McMahon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Vincent Pavot
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | - Andriani Ioannou
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Sarah Gilbert
- The Jenner Institute, University of Oxford, Oxford, UK.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
18
|
Meckiff BJ, Ladell K, McLaren JE, Ryan GB, Leese AM, James EA, Price DA, Long HM. Primary EBV Infection Induces an Acute Wave of Activated Antigen-Specific Cytotoxic CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:1276-1287. [PMID: 31308093 PMCID: PMC6697742 DOI: 10.4049/jimmunol.1900377] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/20/2019] [Indexed: 12/14/2022]
Abstract
Primary EBV infection drives highly cytotoxic virus-specific CD4+ T cell responses. EBV-specific memory CD4+ T cells are polyfunctional but lack cytotoxic activity. Acute EBV-specific CD4-CTLs differ transcriptionally from classical memory CD4-CTLs.
CD4+ T cells are essential for immune protection against viruses, yet their multiple roles remain ill-defined at the single-cell level in humans. Using HLA class II tetramers, we studied the functional properties and clonotypic architecture of EBV-specific CD4+ T cells in patients with infectious mononucleosis, a symptomatic manifestation of primary EBV infection, and in long-term healthy carriers of EBV. We found that primary infection elicited oligoclonal expansions of TH1-like EBV-specific CD4+ T cells armed with cytotoxic proteins that responded immediately ex vivo to challenge with EBV-infected B cells. Importantly, these acutely generated cytotoxic CD4+ T cells were highly activated and transcriptionally distinct from classically described cytotoxic CD4+ memory T cells that accumulate during other persistent viral infections, including CMV and HIV. In contrast, EBV-specific memory CD4+ T cells displayed increased cytokine polyfunctionality but lacked cytotoxic activity. These findings suggested an important effector role for acutely generated cytotoxic CD4+ T cells that could potentially be harnessed to improve the efficacy of vaccines against EBV.
Collapse
Affiliation(s)
- Benjamin J Meckiff
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; and
| | - James E McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; and
| | - Gordon B Ryan
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Alison M Leese
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Eddie A James
- Tetramer Core Laboratory, Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; and
| | - Heather M Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
19
|
Nelson SA, Sant AJ. Imprinting and Editing of the Human CD4 T Cell Response to Influenza Virus. Front Immunol 2019; 10:932. [PMID: 31134060 PMCID: PMC6514101 DOI: 10.3389/fimmu.2019.00932] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022] Open
Abstract
Immunity to influenza is unique among pathogens, in that immune memory is established both via intermittent lung localized infections with highly variable influenza virus strains and by intramuscular vaccinations with inactivated protein-based vaccines. Studies in the past decades have suggested that the B cell responses to influenza infection and vaccination are highly biased by an individual's early history of influenza infection. This reactivity likely reflects both the competitive advantage that memory B cells have in an immune response and the relatively limited diversity of epitopes in influenza hemagglutinin that are recognized by B cells. In contrast, CD4 T cells recognize a wide array of epitopes, with specificities that are heavily influenced by the diversity of influenza antigens available, and a multiplicity of functions that are determined by both priming events and subsequent confrontations with antigens. Here, we consider the events that prime and remodel the influenza-specific CD4 T cell response in humans that have highly diverse immune histories and how the CD4 repertoire may be edited in terms of functional potential and viral epitope specificity. We discuss the consequences that imprinting and remodeling may have on the potential of different human hosts to rapidly respond with protective cellular immunity to infection. Finally, these issues are discussed in the context of future avenues of investigation and vaccine strategies.
Collapse
Affiliation(s)
| | - Andrea J. Sant
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
20
|
Sant AJ, DiPiazza AT, Nayak JL, Rattan A, Richards KA. CD4 T cells in protection from influenza virus: Viral antigen specificity and functional potential. Immunol Rev 2019; 284:91-105. [PMID: 29944766 DOI: 10.1111/imr.12662] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CD4 T cells convey a number of discrete functions to protective immunity to influenza, a complexity that distinguishes this arm of adaptive immunity from B cells and CD8 T cells. Although the most well recognized function of CD4 T cells is provision of help for antibody production, CD4 T cells are important in many aspects of protective immunity. Our studies have revealed that viral antigen specificity is a key determinant of CD4 T cell function, as illustrated both by mouse models of infection and human vaccine responses, a factor whose importance is due at least in part to events in viral antigen handling. We discuss research that has provided insight into the diverse viral epitope specificity of CD4 T cells elicited after infection, how this primary response is modified as CD4 T cells home to the lung, establish memory, and after challenge with a secondary and distinct influenza virus strain. Our studies in human subjects point out the challenges facing vaccine efforts to facilitate responses to novel and avian strains of influenza, as well as strategies that enhance the ability of CD4 T cells to promote protective antibody responses to both seasonal and potentially pandemic strains of influenza.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Anthony T DiPiazza
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jennifer L Nayak
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.,Division of Infectious Diseases, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Ajitanuj Rattan
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
21
|
Wen F, Smith MR, Zhao H. Construction and Screening of an Antigen-Derived Peptide Library Displayed on Yeast Cell Surface for CD4+ T Cell Epitope Identification. Methods Mol Biol 2019; 2024:213-234. [PMID: 31364052 DOI: 10.1007/978-1-4939-9597-4_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Antigenic peptides (termed T cell epitopes) are assembled with major histocompatibility complex (MHC) molecules and presented on the surface of antigen-presenting cells (APCs) for T cell recognition. T cells engage these peptide-MHCs using T cell receptors (TCRs). Because T cell epitopes determine the specificity of a T cell immune response, their prediction and identification are important steps in developing peptide-based vaccines and immunotherapies. In recent years, a number of computational methods have been developed to predict T cell epitopes by evaluating peptide-MHC binding; however, the success of these methods has been limited for MHC class II (MHCII) due to the structural complexity of MHCII antigen presentation. Moreover, while peptide-MHC binding is a prerequisite for a T cell epitope, it alone is not sufficient. Therefore, T cell epitope identification requires further functional verification of the MHC-binding peptide using professional APCs, which are difficult to isolate, expand, and maintain. To address these issues, we have developed a facile, accurate, and high-throughput method for T cell epitope mapping by screening antigen-derived peptide libraries in complex with MHC protein displayed on yeast cell surface. Here, we use hemagglutinin and influenza A virus X31/A/Aichi/68 as examples to describe the key steps in identification of CD4+ T cell epitopes from a single antigenic protein and the entire genome of a pathogen, respectively. Methods for single-chain peptide MHC vector design, yeast surface display, peptide library generation in Escherichia coli, and functional screening in Saccharomyces cerevisiae are discussed.
Collapse
Affiliation(s)
- Fei Wen
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mason R Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Huimin Zhao
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Chemistry, Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
22
|
Influenza and sudden unexpected death: the possible role of peptide cross-reactivity. INFECTION INTERNATIONAL 2018. [DOI: 10.2478/ii-2018-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Abstract
This study investigates the hypothesis that cross-reactions may occur between human cardiac proteins and influenza antigens, thus possibly representing the molecular mechanism underlying influenzaassociated sudden unexpected death (SUD). Using titin protein as a research model, data were obtained on (1) the occurrence of the titin octapeptide AELLVLLE or its mimic AELLVALE in influenza A virus hemagglutinin (HA) sequences; (2) the immunological potential of AELLVLLE and its mimic AELLVALE; (3) the possible role of the flanking amino acid aa) context of the two octapeptide determinants in eliciting cross-reactivity between the human cardiac titin protein and HA antigens.
Collapse
|
23
|
Sant AJ, Richards KA, Nayak J. Distinct and complementary roles of CD4 T cells in protective immunity to influenza virus. Curr Opin Immunol 2018; 53:13-21. [PMID: 29621639 PMCID: PMC6141328 DOI: 10.1016/j.coi.2018.03.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/17/2018] [Accepted: 03/19/2018] [Indexed: 02/01/2023]
Abstract
CD4 T cells play a multiplicity of roles in protective immunity to influenza. Included in these functions are help for high affinity antibody production, enhancement of CD8 T cell expansion, function and memory, acceleration of the early innate response to infection and direct cytotoxicity. The influenza-specific CD4 T cell repertoire in humans established through exposures to infection and vaccination has been found to be highly variable in abundance, specificity and functionality. Deficits in particular subsets of CD4 T cells recruited into the response result in diminished antibody responses and protection from infection. Therefore, improved strategies for vaccination should include better methods to identify deficiencies in the circulating CD4 T cell repertoire, and vaccine constructs that increase the representation of CD4 T cells of the correct specificity and functionality.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA.
| | - Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA
| | - Jennifer Nayak
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, USA; Department of Microbiology and Immunology, University of Rochester Medical Center, USA; Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, USA
| |
Collapse
|
24
|
Kumar A, Meldgaard TS, Bertholet S. Novel Platforms for the Development of a Universal Influenza Vaccine. Front Immunol 2018; 9:600. [PMID: 29628926 PMCID: PMC5877485 DOI: 10.3389/fimmu.2018.00600] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/09/2018] [Indexed: 12/19/2022] Open
Abstract
Despite advancements in immunotherapeutic approaches, influenza continues to cause severe illness, particularly among immunocompromised individuals, young children, and elderly adults. Vaccination is the most effective way to reduce rates of morbidity and mortality caused by influenza viruses. Frequent genetic shift and drift among influenza-virus strains with the resultant disparity between circulating and vaccine virus strains limits the effectiveness of the available conventional influenza vaccines. One approach to overcome this limitation is to develop a universal influenza vaccine that could provide protection against all subtypes of influenza viruses. Moreover, the development of a novel or improved universal influenza vaccines may be greatly facilitated by new technologies including virus-like particles, T-cell-inducing peptides and recombinant proteins, synthetic viruses, broadly neutralizing antibodies, and nucleic acid-based vaccines. This review discusses recent scientific advances in the development of next-generation universal influenza vaccines.
Collapse
Affiliation(s)
- Arun Kumar
- GSK, Research and Development Center, Siena, Italy.,Linköping University, Linköping, Sweden
| | - Trine Sundebo Meldgaard
- GSK, Research and Development Center, Siena, Italy.,DTU Nanotech, Technical University of Denmark, Copenhagen, Denmark
| | - Sylvie Bertholet
- GSK, Research and Development Center, Siena, Italy.,GSK, Research and Development Center, Rockville, MD, United States
| |
Collapse
|
25
|
Lohia N, Baranwal M. Immune responses to highly conserved influenza A virus matrix 1 peptides. Microbiol Immunol 2017; 61:225-231. [PMID: 28429374 DOI: 10.1111/1348-0421.12485] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/12/2017] [Accepted: 04/18/2017] [Indexed: 01/07/2023]
Abstract
Influenza vaccine development is considered to be complicated and challenging. Constantly evolving influenza viruses require continuous global monitoring and reformulation of the vaccine strains. Peptides that are conserved among different strains and subtypes of influenza A virus are strongly considered to be attractive targets for development of cross protective influenza vaccines that stimulate cellular responses. In this study, three highly conserved (>90%) matrix 1 peptides that contain multiple T cell epitopes, ILGFVFTLTVPSERGLQRRRF (PM 1), LIRHENRMVLASTTAKA (PM 2) and LQAYQKRMGVQMQR (PM 3), were assessed for their immunogenic potential in vitro by subjecting peripheral blood mononuclear cells from healthy volunteers to repetitive stimulation with these chemically synthesised peptides and measuring their IFN-γ concentrations, proliferation by ELISA, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, respectively. Seven samples were screened for immunogenicity of PM 1 and PM 2, and six for that of PM 3. All six samples had positive responses (IFN-γ secretion) to PM 3 stimulation, as did five and three for PM 2 and PM 1 respectively. In contrast, seven (PM 1 and PM 2) and four (PM 3) samples showed proliferative response as compared with unstimulated cells. The encouraging immunogenic response generated by these highly conserved matrix 1 peptides indicates they are prospective candidates for development of broadly reactive influenza vaccines.
Collapse
Affiliation(s)
- Neha Lohia
- Department of Biotechnology, Thapar University, Patiala147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar University, Patiala147004, India
| |
Collapse
|
26
|
Absence of autoreactive CD4 + T-cells targeting HLA-DQA1*01:02/DQB1*06:02 restricted hypocretin/orexin epitopes in narcolepsy type 1 when detected by EliSpot. J Neuroimmunol 2017; 309:7-11. [DOI: 10.1016/j.jneuroim.2017.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 11/20/2022]
|
27
|
Chen L, Anthony A, Oveissi S, Huang M, Zanker D, Xiao K, Wu C, Zou Q, Chen W. Broad-Based CD4 + T Cell Responses to Influenza A Virus in a Healthy Individual Who Lacks Typical Immunodominance Hierarchy. Front Immunol 2017; 8:375. [PMID: 28421076 PMCID: PMC5377932 DOI: 10.3389/fimmu.2017.00375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/15/2017] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus (IAV) infection is a significant cause of morbidity and mortality worldwide. CD4+ T cell responses have been shown to be important for influenza protection in mouse models and in human volunteers. IAV antigen-specific CD4+ T cell responses were found to focus on matrix 1 (M1) and nucleoprotein (NP) at the protein antigen level. At the epitope level, only several epitopes within M1 and NP were recognized by CD4+ T cells. And the epitope-specific CD4+ T cell responses showed a typical immunodominance hierarchy in most of the healthy individuals studied. In this study, we reported one case of atypical immunodominance hierarchy of CD4+ T cell responses to IAV. M1 and NP were still the immunodominant targets of CD4+ T cell responses. However, CD4+ T cell responses specific to 11 epitopes derived from M1 and NP were detected and showed no significant immunodominance hierarchy. Such an atypical pattern is likely determined by the individual's HLA alleles. These findings will help us better understand the anti-IAV immunity as a whole and improve future vaccines against IAV.
Collapse
Affiliation(s)
- Li Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China.,T Cell Laboratory, School of Molecular Science, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia.,Department of Blood Transfusion, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Anjaleena Anthony
- T Cell Laboratory, School of Molecular Science, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Sara Oveissi
- T Cell Laboratory, School of Molecular Science, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Miaojuan Huang
- T Cell Laboratory, School of Molecular Science, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Damien Zanker
- T Cell Laboratory, School of Molecular Science, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Kun Xiao
- T Cell Laboratory, School of Molecular Science, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Chao Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Weisan Chen
- T Cell Laboratory, School of Molecular Science, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
28
|
Maljkovic Berry I, Melendrez MC, Li T, Hawksworth AW, Brice GT, Blair PJ, Halsey ES, Williams M, Fernandez S, Yoon IK, Edwards LD, Kuschner R, Lin X, Thomas SJ, Jarman RG. Frequency of influenza H3N2 intra-subtype reassortment: attributes and implications of reassortant spread. BMC Biol 2016; 14:117. [PMID: 28034300 PMCID: PMC5200972 DOI: 10.1186/s12915-016-0337-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/03/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Increasing evidence suggests that influenza reassortment not only contributes to the emergence of new human pandemics but also plays an important role in seasonal influenza epidemics, disease severity, evolution, and vaccine efficacy. We studied this process within 2091 H3N2 full genomes utilizing a combination of the latest reassortment detection tools and more conventional phylogenetic analyses. RESULTS We found that the amount of H3N2 intra-subtype reassortment depended on the number of sampled genomes, occurred with a steady frequency of 3.35%, and was not affected by the geographical origins, evolutionary patterns, or previous reassortment history of the virus. We identified both single reassortant genomes and reassortant clades, each clade representing one reassortment event followed by successful spread of the reassorted variant in the human population. It was this spread that was mainly responsible for the observed high presence of H3N2 intra-subtype reassortant genomes. The successfully spread variants were generally sampled within one year of their formation, highlighting the risk of their rapid spread but also presenting an opportunity for their rapid detection. Simultaneous spread of several different reassortant lineages was observed, and despite their limited average lifetime, second and third generation reassortment was detected, as well as reassortment between viruses belonging to different vaccine-associated clades, likely displaying differing antigenic properties. Some of the spreading reassortants remained confined to certain geographical regions, while others, sharing common properties in amino acid positions of the HA, NA, and PB2 segments, were found throughout the world. CONCLUSIONS Detailed surveillance of seasonal influenza reassortment patterns and variant properties may provide unique information needed for prediction of spread and construction of future influenza vaccines.
Collapse
Affiliation(s)
| | | | - Tao Li
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Anthony W Hawksworth
- Operational Infectious Diseases Directorate, Naval Health Research Center, San Diego, CA, USA
| | - Gary T Brice
- Operational Infectious Diseases Directorate, Naval Health Research Center, San Diego, CA, USA
| | - Patrick J Blair
- Operational Infectious Diseases Directorate, Naval Health Research Center, San Diego, CA, USA
| | | | | | - Stefan Fernandez
- Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - In-Kyu Yoon
- Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
- Present Address: International Vaccine Institute, Seoul, Republic of Korea
| | - Leslie D Edwards
- Office of Medical Services, US Department of State, Washington, DC, USA
| | - Robert Kuschner
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Xiaoxu Lin
- Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | | |
Collapse
|
29
|
Gutiérrez AH, Loving C, Moise L, Terry FE, Brockmeier SL, Hughes HR, Martin WD, De Groot AS. In Vivo Validation of Predicted and Conserved T Cell Epitopes in a Swine Influenza Model. PLoS One 2016; 11:e0159237. [PMID: 27411061 PMCID: PMC4943726 DOI: 10.1371/journal.pone.0159237] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/29/2016] [Indexed: 01/10/2023] Open
Abstract
Swine influenza is a highly contagious respiratory viral infection in pigs that is responsible for significant financial losses to pig farmers annually. Current measures to protect herds from infection include: inactivated whole-virus vaccines, subunit vaccines, and alpha replicon-based vaccines. As is true for influenza vaccines for humans, these strategies do not provide broad protection against the diverse strains of influenza A virus (IAV) currently circulating in U.S. swine. Improved approaches to developing swine influenza vaccines are needed. Here, we used immunoinformatics tools to identify class I and II T cell epitopes highly conserved in seven representative strains of IAV in U.S. swine and predicted to bind to Swine Leukocyte Antigen (SLA) alleles prevalent in commercial swine. Epitope-specific interferon-gamma (IFNγ) recall responses to pooled peptides and whole virus were detected in pigs immunized with multi-epitope plasmid DNA vaccines encoding strings of class I and II putative epitopes. In a retrospective analysis of the IFNγ responses to individual peptides compared to predictions specific to the SLA alleles of cohort pigs, we evaluated the predictive performance of PigMatrix and demonstrated its ability to distinguish non-immunogenic from immunogenic peptides and to identify promiscuous class II epitopes. Overall, this study confirms the capacity of PigMatrix to predict immunogenic T cell epitopes and demonstrate its potential for use in the design of epitope-driven vaccines for swine. Additional studies that match the SLA haplotype of animals with the study epitopes will be required to evaluate the degree of immune protection conferred by epitope-driven DNA vaccines in pigs.
Collapse
Affiliation(s)
- Andres H. Gutiérrez
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, United States of America
| | - Crystal Loving
- Virus and Prion Diseases Research Unit, NADC, USDA ARS, Ames, IA, United States of America
| | - Leonard Moise
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, United States of America
- EpiVax Inc., Providence, RI, United States of America
| | | | - Susan L. Brockmeier
- Virus and Prion Diseases Research Unit, NADC, USDA ARS, Ames, IA, United States of America
| | - Holly R. Hughes
- Virus and Prion Diseases Research Unit, NADC, USDA ARS, Ames, IA, United States of America
| | | | - Anne S. De Groot
- Institute for Immunology and Informatics, Department of Cell and Molecular Biology, University of Rhode Island, Providence, RI, United States of America
- EpiVax Inc., Providence, RI, United States of America
- * E-mail:
| |
Collapse
|
30
|
Identification of a Highly Conserved Epitope on Avian Influenza Virus Non-Structural Protein 1 Using a Peptide Microarray. PLoS One 2016; 11:e0149868. [PMID: 26938453 PMCID: PMC4777286 DOI: 10.1371/journal.pone.0149868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/05/2016] [Indexed: 11/20/2022] Open
Abstract
Avian influenza virus (AIV) non-structural protein 1 (NS1) is a multifunctional protein. It is present at high levels in infected cells and can be used for AIV detection and diagnosis. In this study, we generated monoclonal antibody (MAb) D7 against AIV NS1 protein by immunization of BALB/c mice with purified recombinant NS1 protein expressed in Escherichia coli. Isotype determination revealed that the MAb was IgG1/κ-type subclass. To identify the epitope of the MAb D7, the NS1 protein was truncated into a total of 225 15-mer peptides with 14 amino acid overlaps, which were spotted for a peptide microarray. The results revealed that the MAb D7 recognized the consensus DAPF motif. Furthermore, the AIV NS1 protein with the DAPF motif deletion was transiently expressed in 293T cells and failed to react with MAb D7. Subsequently, the DAPF motif was synthesized with an elongated GSGS linker at both the C- and N-termini. The MAb D7 reacted with the synthesized peptide both in enzyme-linked immunosorbent assay (ELISA) and dot-blot assays. From these results, we concluded that DAPF motif is the epitope of MAb D7. To our knowledge, this is the first report of a 4-mer epitope on the NS1 protein of AIV that can be recognized by MAb using a peptide microarray, which is able to simplify epitope identification, and that could serve as the basis for immune responses against avian influenza.
Collapse
|
31
|
Hegde NR. Cell culture-based influenza vaccines: A necessary and indispensable investment for the future. Hum Vaccin Immunother 2016; 11:1223-34. [PMID: 25875691 DOI: 10.1080/21645515.2015.1016666] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The traditional platform of using embryonated chicken eggs for the production of influenza vaccines has several drawbacks including the inability to meet the volume of required doses in the case of widespread epidemics and pandemics. Cell culture platforms have therefore been explored in the last 2 decades, and have attracted further attention following the H1N1 pandemic outbreak. This platform, while not the most economical for large-scale production, has several advantages, and can supplement the vaccine requirement when needed. Recent developments in production technologies have contributed greatly to fine-tuning this platform. In combination with other technologies such as live attenuated and recombinant protein or virus-like particle vaccines, and different adjuvants and delivery systems, cell culture-based influenza vaccine platform can be used both for production of seasonal vaccine, and to mitigate vaccine shortages in pandemic situations.
Collapse
Affiliation(s)
- Nagendra R Hegde
- a Ella Foundation; Genome Valley; Turkapally , Shameerpet Mandal , Hyderabad , India
| |
Collapse
|
32
|
DiPiazza A, Richards KA, Knowlden ZAG, Nayak JL, Sant AJ. The Role of CD4 T Cell Memory in Generating Protective Immunity to Novel and Potentially Pandemic Strains of Influenza. Front Immunol 2016; 7:10. [PMID: 26834750 PMCID: PMC4725218 DOI: 10.3389/fimmu.2016.00010] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/11/2016] [Indexed: 11/24/2022] Open
Abstract
Recent events have made it clear that potentially pandemic strains of influenza regularly pose a threat to human populations. Therefore, it is essential that we develop better strategies to enhance vaccine design and evaluation to predict those that will be poor responders to vaccination and to identify those that are at particular risk of disease-associated complications following infection. Animal models have revealed the discrete functions that CD4 T cells play in developing immune response and to influenza immunity. However, humans have a complex immunological history with influenza through periodic infection and vaccination with seasonal variants, leading to the establishment of heterogeneous memory populations of CD4 T cells that participate in subsequent responses. The continual evolution of the influenza-specific CD4 T cell repertoire involves both specificity and function and overlays other restrictions on CD4 T cell activity derived from viral antigen handling and MHC class II:peptide epitope display. Together, these complexities in the influenza-specific CD4 T cell repertoire constitute a formidable obstacle to predicting protective immune response to potentially pandemic strains of influenza and in devising optimal vaccine strategies to potentiate these responses. We suggest that more precise efforts to identify and enumerate both the positive and negative contributors within the CD4 T cell compartment will aid significantly in the achievement of these goals.
Collapse
Affiliation(s)
- Anthony DiPiazza
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center , Rochester, NY , USA
| | - Katherine A Richards
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center , Rochester, NY , USA
| | - Zackery A G Knowlden
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center , Rochester, NY , USA
| | - Jennifer L Nayak
- Department of Pediatrics, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center , Rochester, NY , USA
| | - Andrea J Sant
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center , Rochester, NY , USA
| |
Collapse
|
33
|
Wong TM, Ross TM. Use of computational and recombinant technologies for developing novel influenza vaccines. Expert Rev Vaccines 2015; 15:41-51. [PMID: 26595182 DOI: 10.1586/14760584.2016.1113877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Influenza vaccine design has changed considerably with advancements in bioinformatics and computational biology. Improved surveillance efforts provide up-to-date information about influenza sequence diversity and assist with monitoring the spread of epidemics and vaccine efficacy rates. The advent of next-generation sequencing, epitope scanning and high-throughput analysis all help decipher influenza-associated protein interactions as well as predict immune responsiveness based on host genetic diversity. Computational approaches are utilized in nearly all aspects of vaccine design, from modeling, compatibility predictions, and optimization of antigens in various platforms. This overview discusses how computational techniques strengthen vaccine efforts against highly diverse influenza species.
Collapse
Affiliation(s)
- Terianne M Wong
- a Center for Vaccines and Immunology, Department of Infectious Diseases , University of Georgia , Athens , GA , USA
| | - Ted M Ross
- a Center for Vaccines and Immunology, Department of Infectious Diseases , University of Georgia , Athens , GA , USA
| |
Collapse
|
34
|
Wen X, Sun J, Wang X, Bao H, Zhao Y, Zeng X, Xu X, Ma Y, Gu L, Chen H. Identification of a novel linear epitope on the NS1 protein of avian influenza virus. BMC Microbiol 2015; 15:168. [PMID: 26289074 PMCID: PMC4545905 DOI: 10.1186/s12866-015-0507-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 08/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The NS1 protein of avian influenza virus (AIV) is an important virulent factor of AIV. It has been shown to counteract host type I interferon response, to mediate host cell apoptosis, and to regulate the process of protein synthesis. The identification of AIV epitopes on NS1 protein is important for understanding influenza virus pathogenesis. RESULTS In this paper, we describe the generation, identification, and epitope mapping of a NS1 protein-specific monoclonal antibody (MAb) D9. First, to induce the production of MAbs, BALB/c mice were immunized with a purified recombinant NS1 expressed in E. coli. The spleen cells from the immunized mice were fused with myeloma cells SP2/0, and through screening via indirect ELISAs, a MAb, named D9, was identified. Western blot assay results showed that MAb D9 reacted strongly with the recombinant NS1. Confocal laser scanning microscopy showed that this MAb also reacts with NS1 expressed in 293T cells that had been transfected with eukaryotic recombinant plasmids. Results from screening a phage display random 7-mer peptide library with MAb D9 demonstrated that it recognizes phages displaying peptides with the consensus peptide WNLNTV--VS, which closely matches the (182)WNDNTVRVS(190) of AIV NS1. Further identification of the displayed epitope was performed with a set of truncated polypeptides expressed as glutathione S-transferase fusion proteins, and the motif (182)WNDNT(186) was defined as the minimal unit of the linear B cell epitope recognized by MAb D9 in western blot assays. Moreover, homology analysis showed that this epitope is a conserved motif among AIV. CONCLUSIONS We identified a conserved linear epitope, WNDNT, on the AIV NS1 protein that is recognized by MAb D9. This MAb and its epitope may facilitate future studies on NS1 function and aid the development of new diagnostic methods for AIV detection.
Collapse
Affiliation(s)
- Xuexia Wen
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China. .,College of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China.
| | - Jiashan Sun
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Xiurong Wang
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Hongmei Bao
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Yuhui Zhao
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Xianying Zeng
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Xiaolong Xu
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Yong Ma
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Linlin Gu
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Hualan Chen
- Animal Influenza Laboratory of the Ministry of Agriculture and State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| |
Collapse
|
35
|
In Silico Identification of Highly Conserved Epitopes of Influenza A H1N1, H2N2, H3N2, and H5N1 with Diagnostic and Vaccination Potential. BIOMED RESEARCH INTERNATIONAL 2015; 2015:813047. [PMID: 26346523 PMCID: PMC4544958 DOI: 10.1155/2015/813047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 06/13/2015] [Accepted: 06/14/2015] [Indexed: 12/15/2022]
Abstract
The unpredictable, evolutionary nature of the influenza A virus (IAV) is the primary problem when generating a vaccine and when designing diagnostic strategies; thus, it is necessary to determine the constant regions in viral proteins. In this study, we completed an in silico analysis of the reported epitopes of the 4 IAV proteins that are antigenically most significant (HA, NA, NP, and M2) in the 3 strains with the greatest world circulation in the last century (H1N1, H2N2, and H3N2) and in one of the main aviary subtypes responsible for zoonosis (H5N1). For this purpose, the HMMER program was used to align 3,016 epitopes reported in the Immune Epitope Database and Analysis Resource (IEDB) and distributed in 34,294 stored sequences in the Pfam database. Eighteen epitopes were identified: 8 in HA, 5 in NA, 3 in NP, and 2 in M2. These epitopes have remained constant since they were first identified (~91 years) and are present in strains that have circulated on 5 continents. These sites could be targets for vaccination design strategies based on epitopes and/or as markers in the implementation of diagnostic techniques.
Collapse
|
36
|
Using epidemics to map H3 equine influenza virus determinants of antigenicity. Virology 2015; 481:187-98. [DOI: 10.1016/j.virol.2015.02.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/08/2014] [Accepted: 02/14/2015] [Indexed: 01/25/2023]
|
37
|
Adaptive evolution of a novel avian-origin influenza A/H7N9 virus. Genomics 2014; 104:545-53. [PMID: 25449177 DOI: 10.1016/j.ygeno.2014.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 10/25/2014] [Accepted: 10/29/2014] [Indexed: 11/21/2022]
Abstract
In China, the recent outbreak of novel influenza A/H7N9 virus has been assumed to be severe, and it may possibly turn brutal in the near future. In order to develop highly protective vaccines and drugs for the A/H7N9 virus, it is critical to find out the selection pressure of each amino acid site. In the present study, six different statistical methods consisting of four independent codon-based maximum likelihood (CML) methods, one hierarchical Bayesian (HB) method and one branch-site (BS) method, were employed to determine if each amino acid site of A/H7N9 virus is under natural selection pressure. Functions for both positively and negatively selected sites were inferred by annotating these sites with experimentally verified amino acid sites. Comprehensively, the single amino acid site 627 of PB2 protein was inferred as positively selected and it function was identified as a T-cell epitope (TCE). Among the 26 negatively selected amino acid sites of PB2, PB1, PA, HA, NP, NA, M1 and NS2 proteins, only 16 amino acid sites were identified to be involved in TCEs. In addition, 7 amino acid sites including, 608 and 609 of PA, 480 of NP, and 24, 25, 109 and 205 of M1, were identified to be involved in both B-cell epitopes (BCEs) and TCEs. Conversely, the function of positions 62 of PA, and, 43 and 113 of HA was unknown. In conclusion, the seven amino acid sites engaged in both BCEs and TCEs were identified as highly suitable targets, as these sites will be predicted to play a principal role in inducing strong humoral and cellular immune responses against A/H7N9 virus.
Collapse
|
38
|
Immunodominant CD4+ T-cell responses to influenza A virus in healthy individuals focus on matrix 1 and nucleoprotein. J Virol 2014; 88:11760-73. [PMID: 25078703 DOI: 10.1128/jvi.01631-14] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antigen-specific CD4(+) T cells are essential for effective virus-specific host responses, with recent human challenge studies (in volunteers) establishing their importance for influenza A virus (IAV)-specific immunity. However, while many IAV CD4(+) T cell epitopes have been identified, few are known to stimulate immunodominant CD4(+) T cell responses. Moreover, much remains unclear concerning the major antigen(s) responded to by the human CD4(+) T cells and the extents and magnitudes of these responses. We initiated a systematic screen of immunodominant CD4(+) T cell responses to IAV in healthy individuals. Using in vitro expanded-multispecificity IAV-specific T cell lines and individual IAV protein antigens produced by recombinant vaccinia viruses, we found that the internal matrix protein 1 (M1) and nucleoprotein (NP) were the immunodominant targets of CD4(+) T cell responses. Ten epitopes derived from M1 and NP were definitively characterized. Furthermore, epitope sequence conservation analysis established that immunodominance correlated with an increased frequency of mutations, reflecting the fact that these prominent epitopes are under greater selective pressure. Such evidence that particular CD4(+) T cells are important for protection/recovery is of value for the development of novel IAV vaccines and for our understanding of different profiles of susceptibility to these major pathogens. Importance: Influenza virus causes half a million deaths annually. CD4(+) T cell responses have been shown to be important for protection against influenza and for recovery. CD4(+) T cell responses are also critical for efficient CD8(+) T cell response and antibody response. As immunodominant T cells generally play a more important role, characterizing these immunodominant responses is critical for influenza vaccine development. We show here that the internal matrix protein 1 (M1) and nucleoprotein (NP), rather than the surface proteins reported previously, are the immunodominant targets of CD4(+) T cell responses. Interestingly, these immunodominant epitope regions accumulated many mutations over time, which likely indicates increased immune pressure. These findings have significant implications for the design of T cell-based influenza vaccines.
Collapse
|
39
|
Lohia N, Baranwal M. Conserved peptides containing overlapping CD4+ and CD8+ T-cell epitopes in the H1N1 influenza virus: an immunoinformatics approach. Viral Immunol 2014; 27:225-34. [PMID: 24821387 DOI: 10.1089/vim.2013.0135] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pandemic threats of the H1N1 influenza virus have drawn attention to developing a universal vaccine against circulating and future strains of this virus. An immunoinformatics study was conducted to identify conserved peptides containing CD4+ and CD8+ T-cell epitopes from all the hemagglutinin (HA) and neuraminidase (NA) protein sequences available until February 2013 to cover the seasonal as well as the pandemic strains of the H1N1 virus. In the present study, six different immunoinformatics prediction programs were used in order to define the epitopes. Five conserved peptides of HA and six of NA protein were obtained that contained overlapping CD4+ and CD8+ T-cell epitopes. These identified peptides have a binding affinity for a large number of major histocompatibility complex (MHC) alleles. WHGSNRPWVSF of NA protein is a new peptide whose T-cell response has not been previously reported. Population coverage studies have shown that these peptide fragments have the capacity to induce a potent immune response among individuals from different populations around the world. Hence, these HA and NA peptides may be considered as interesting candidates for vaccine design.
Collapse
Affiliation(s)
- Neha Lohia
- Department of Biotechnology, Thapar University , Patiala, India
| | | |
Collapse
|
40
|
Xiong F, Xia L, Wang J, Wu B, Wang D, Yuan L, Cheng Y, Zhu H, Che X, Zhang Q, Zhao G, Wang Y. A high-affinity CDR-grafted antibody against influenza A H5N1 viruses recognizes a conserved epitope of H5 hemagglutinin. PLoS One 2014; 9:e88777. [PMID: 24558425 PMCID: PMC3928294 DOI: 10.1371/journal.pone.0088777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/15/2014] [Indexed: 01/19/2023] Open
Abstract
Highly pathogenic avian influenza (HPAI) H5N1 virus infection is still a potential threat to public health worldwide. While vaccines and antiviral drugs are currently under development, neutralizing antibodies could offer an alternative strategy to prevent and treat H5N1 virus infection. In the present study, we had developed a humanized antibody against H5N1 viruses from mouse-derived hybridoma in order to minimize its immunogenicity for potential clinical application. The humanized antibody hH5M9 was generated by transferring the mouse complementarity determining region (CDR) residues together with four key framework region (FR) residues onto the FR of the human antibody. This humanized antibody exhibited high affinity and specificity comparable to the parental mouse or chimeric counterpart with broad and strong neutralization activity against all H5N1 clades and subclades except for Egypt clades investigated. Furthermore, through epitope mapping we identified a linear epitope on the top region of hemagglutinin (HA) that was H5N1 specific and conserved. Our results for the first time reported a humanized antibody against H5N1 viruses by CDR grafting method. With the expected lower immunogenicity, this humanized antibody was expected to be more efficacious than murine or human-mouse chimeric antibodies for future application in humans.
Collapse
Affiliation(s)
- Feifei Xiong
- School of Life Science and Technology, Tongji University, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Liliang Xia
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Jingfang Wang
- Shanghai Center for Systems Biomedicine, Shanghai Jiaotong University, Shanghai, China
| | - Biao Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Dengyu Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Longfang Yuan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Yating Cheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Hongying Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Xiaoyan Che
- Central Laboratory, Zhujiang Hospital, The Southern Medical University, Guangzhou, China
| | - Qinghua Zhang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Guoping Zhao
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Ying Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
41
|
Hu YJ, Tu PC, Lin CS, Guo ST. Identification and chronological analysis of genomic signatures in influenza A viruses. PLoS One 2014; 9:e84638. [PMID: 24416256 PMCID: PMC3885579 DOI: 10.1371/journal.pone.0084638] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 11/26/2013] [Indexed: 01/05/2023] Open
Abstract
An increase in the availability of data on the influenza A viruses (IAV) has enabled the identification of the potential determinants of IAV host specificity using computational approaches. In this study, we proposed an alternative approach, based on the adjusted Rand index (ARI), for the evaluation of genomic signatures of IAVs and their ability to distinguish hosts they infected. Our experiments showed that the host-specific signatures identified using the ARI were more characteristic of their hosts than those identified using previous measures. Our results provided updates on the host-specific genomic signatures in the internal proteins of the IAV based on the sequence data as of February 2013 in the National Center for Biotechnology Information (NCBI). Unlike other approaches for signature recognition, our approach considered not only the ability of signatures to distinguish hosts (according to the ARI), but also the chronological relationships among proteins. We identified novel signatures that could be mapped to known functional domains, and introduced a chronological analysis to investigate the changes in host-specific genomic signatures over time. Our chronological analytical approach provided results on the adaptive variability of signatures, which correlated with previous studies' findings, and indicated prospective adaptation trends that warrant further investigation.
Collapse
Affiliation(s)
- Yuh-Jyh Hu
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu, Taiwan
- Department of Computer Science, National Chiao Tung University, Hsinchu, Taiwan
- * E-mail:
| | - Po-Chin Tu
- Department of Computer Science, National Chiao Tung University, Hsinchu, Taiwan
| | - Chun-Sheng Lin
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Szu-Ting Guo
- Department of Computer Science, National Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
42
|
Terajima M, Babon JAB, Co MDT, Ennis FA. Cross-reactive human B cell and T cell epitopes between influenza A and B viruses. Virol J 2013; 10:244. [PMID: 23886073 PMCID: PMC3726517 DOI: 10.1186/1743-422x-10-244] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 07/24/2013] [Indexed: 01/26/2023] Open
Abstract
Influenza A and B viruses form different genera, which were originally distinguished by antigenic differences in their nucleoproteins and matrix 1 proteins. Cross-protection between these two genera has not been observed in animal experiments, which is consistent with the low homology in viral proteins common to both viruses except for one of three polymerase proteins, polymerase basic 1 (PB1). Recently, however, antibody and CD4+ T cell epitopes conserved between the two genera were identified in humans. A protective antibody epitope was located in the stalk region of the surface glycoprotein, hemagglutinin, and a CD4+ T cell epitope was located in the fusion peptide of the hemagglutinin. The fusion peptide was also found to contain antibody epitopes in humans and animals. A short stretch of well-conserved peptide was also identified in the other surface glycoprotein, neuraminidase, and antibodies binding to this peptide were generated by peptide immunization in rabbits. Although PB1, the only protein which has relatively high overall sequence homology between influenza A and B viruses, is not considered an immunodominant protein in the T cell responses to influenza A virus infection, amino acid sequence comparisons show that a considerable number of previously identified T cell epitopes in the PB1 of influenza A viruses are conserved in the PB1 of influenza B viruses. These data indicate that B and T cell cross-reactivity exists between influenza A and B viruses, which may have modulatory effects on the disease process and recovery. Although the antibody titers and the specific T cell frequencies induced by natural infection or standard vaccination may not be high enough to provide cross protection in humans, it might be possible to develop immunization strategies to induce these cross-reactive responses more efficiently.
Collapse
Affiliation(s)
- Masanori Terajima
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA.
| | | | | | | |
Collapse
|
43
|
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
|
44
|
Arunachalam R. Detection of site-specific positive Darwinian selection on pandemic influenza A/H1N1 virus genome: integrative approaches. Genetica 2013; 141:143-55. [PMID: 23529677 DOI: 10.1007/s10709-013-9713-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 03/18/2013] [Indexed: 12/17/2022]
Abstract
In the twenty-first century, the first pandemic novel human influenza A/H1N1virus (NIV) outbreak was reported at Mexico and USA on March and early April, 2009 respectively. The outbreak occurred among human populations due to the presence of meager or no immune response against newly emerged viruses. The success of vaccines and drugs depends on their low susceptibility to the formation of escape mutants in virus. Identification of excess, non-synonymous substitutions over synonymous ones is a main indicator of positive Darwinian selection in protein-coding genes of NIVs. The positive Darwinian selection operating on each site of proteins were inferred by computing ω, the ratio of the non-synonymous/synonymous substitutions [dN/dS (or) Ka/Ks], which was calculated by three different methods in terms of codon-based maximum likelihood, branch-site and empirical Bayesian methods under various models. Totally, nine sites from PB2, PB1, HA, M2 and NS1 are inferred as positively selected. The function for amino acid sites of NIVs proteins under positive selection are inferred by comparing the sites with experimentally determined functionally known amino acid sites. Completely 4 positively selected sites of PB1, HA and M2 are found to be involved in B-cell epitopes (BCEs). Interestingly, most of these sites are also involving in T-cell epitopes (TCEs). However, more sites under positive selection forces are involved in TCEs than those of BCEs. Amino acid sites engaged in both BCEs and TCEs should be measured as highly suitable targets, because these sites could induce the strong humoral and cellular immune responses against targets.
Collapse
Affiliation(s)
- Ramaiah Arunachalam
- Sri Paramakalyani Centre for Environmental Sciences, Manonmaniam Sundaranar University, Alwarkurichi 627412, Tamil Nadu, India.
| |
Collapse
|
45
|
Ruiz-Riol M, Mothe B, Gandhi RT, Bhardwaj N, Scadden DT, Sanchez-Merino V, Brander C. Influenza, but not HIV-specific CTL epitopes, elicits delayed-type hypersensitivity (DTH) reactions in HIV-infected patients. Eur J Immunol 2013; 43:1545-54. [PMID: 23504637 DOI: 10.1002/eji.201242732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 02/15/2013] [Accepted: 03/08/2013] [Indexed: 01/19/2023]
Abstract
The induction of cytotoxic T lymphocytes (CTLs) is believed to be an important defense mechanism against viral infections. The availability of simple, sensitive, specific and physiologically informative in vivo tests, applicable to humans, would greatly elucidate the nature of protective immune responses and facilitate immune monitoring in large vaccine trials. Here we studied the possibility of using defined HLA-A*02:01-restricted CTL epitopes from influenza matrix protein (GL9, GILGFVFTL) and HIV Gag p17 (SL9, SLYNTVATL) to elicit a cutaneous delayed-type hypersensitivity (DTH) reaction. Our results show that the GL9 but not the SL9 epitope was able to induce a DTH reaction. HIV infection status, HIV RNA level and CD4(+) T-cell counts were not predictive of the extent of DTH reactions. However, a markedly reduced expression of skin homing markers CD103 and cutaneous lymphocyte associated Ag (CLA) on epitope-specific CTL populations was associated with a lack of SL9 DTH reactivity. These data demonstrate that DTH reactions can be elicited by optimally defined CTL epitopes per se and point towards specific homing markers that are required for such reactions. These data may offer new insights into the immune pathogenesis of HIV infection and provide the basis of novel immune monitoring approaches for large-scale HIV vaccine trials.
Collapse
Affiliation(s)
- Marta Ruiz-Riol
- IrsiCaixa AIDS Research Institute - HIVACAT, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
46
|
Wen F, Zhao H. Construction and screening of an antigen-derived peptide library displayed on yeast cell surface for CD4+ T cell epitope identification. Methods Mol Biol 2013; 1061:245-264. [PMID: 23963942 DOI: 10.1007/978-1-62703-589-7_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Identification of T cell epitopes is a critical, but often difficult step in studying T cell function and developing peptide-based vaccines and immunotherapies. Unlike antibodies that recognize free soluble antigens, T cell receptor (TCR) recognizes its epitope bound to major histocompatibility complex (MHC) expressed on antigen presenting cells (APCs). In addition, the examination of T cell epitope activity requires the use of professional APCs, which are difficult to isolate, expand, and maintain. To address these issues, we have developed a facile, accurate, and high-throughput method for T cell epitope mapping by screening antigen-derived peptide libraries in complex with MHC protein displayed on yeast cell surface. Here, we use hemagglutinin and influenza A virus X31/A/Aichi/68 as examples to describe the key steps in identification of CD4+ T cell epitopes from a single antigenic protein and the entire genome of a pathogen, respectively. Methods for single-chain peptide-MHC complex vector design, yeast surface display, peptide library generation in Escherichia coli, and functional screening in Saccharomyces cerevisiae are discussed.
Collapse
Affiliation(s)
- Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
47
|
Nayak JL, Fitzgerald TF, Richards KA, Yang H, Treanor JJ, Sant AJ. CD4+ T-cell expansion predicts neutralizing antibody responses to monovalent, inactivated 2009 pandemic influenza A(H1N1) virus subtype H1N1 vaccine. J Infect Dis 2012; 207:297-305. [PMID: 23148285 PMCID: PMC3532833 DOI: 10.1093/infdis/jis684] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background. The ability of influenza vaccines to elicit CD4+ T cells and the relationship between induction of CD4+ T cells and vaccine-induced neutralizing antibody responses has been controversial. The emergence of swine-origin 2009 pandemic influenza A virus subtype H1N1 (A[H1N1]pdm09) provided a unique opportunity to examine responses to an influenza vaccine composed of both novel and previously encountered antigens and to probe the relationship between B-cell and T-cell responses to vaccination. Methods. We tracked CD4+ T-cell and antibody responses of human subjects vaccinated with monovalent subunit A(H1N1)pdm09 vaccine. The specificity and magnitude of the CD4+ T-cell response was evaluated using cytokine enzyme-linked immunosorbent spot assays in conjugation with peptide pools representing distinct influenza virus proteins. Results. Our studies revealed that vaccination induced readily detectable CD4+ T cells specific for conserved portions of hemagglutinin (HA) and the internal viral proteins. Interestingly, expansion of HA-specific CD4+ T cells was most tightly correlated with the antibody response. Conclusions. These results indicate that CD4+ T-cell expansion may be a limiting factor in development of neutralizing antibody responses to pandemic influenza vaccines and suggest that approaches to facilitate CD4+ T-cell recruitment may increase the neutralizing antibody produced in response to vaccines against novel influenza strains.
Collapse
Affiliation(s)
- Jennifer L Nayak
- Department of Pediatrics, Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | | | | | | | |
Collapse
|
48
|
A human CD4+ T cell epitope in the influenza hemagglutinin is cross-reactive to influenza A virus subtypes and to influenza B virus. J Virol 2012; 86:9233-43. [PMID: 22718815 DOI: 10.1128/jvi.06325-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The hemagglutinin protein (HA) of the influenza virus family is a major antigen for protective immunity. Thus, it is a relevant target for developing vaccines. Here, we describe a human CD4(+) T cell epitope in the influenza virus HA that lies in the fusion peptide of the HA. This epitope is well conserved in all 16 subtypes of the HA protein of influenza A virus and the HA protein of influenza B virus. By stimulating peripheral blood mononuclear cells (PBMCs) from a healthy adult donor with peptides covering the entire HA protein based on the sequence of A/Japan/305/1957 (H2N2), we generated a T cell line specific to this epitope. This CD4(+) T cell line recognizes target cells infected with influenza A virus seasonal H1N1 and H3N2 strains, a reassortant H2N1 strain, the 2009 pandemic H1N1 strain, and influenza B virus in cytotoxicity assays and intracellular-cytokine-staining assays. It also lysed target cells infected with avian H5N1 virus. We screened healthy adult PBMCs for T cell responses specific to this epitope and found individuals who had ex vivo gamma interferon (IFN-γ) responses to the peptide epitope in enzyme-linked immunospot (ELISPOT) assays. Almost all donors who responded to the epitope had the HLA-DRB1*09 allele, a relatively common HLA allele. Although natural infection or standard vaccination may not induce strong T and B cell responses to this highly conserved epitope in the fusion peptide, it may be possible to develop a vaccination strategy to induce these CD4(+) T cells, which are cross-reactive to both influenza A and B viruses.
Collapse
|
49
|
Powell TJ, Fox A, Peng Y, Quynh Mai LT, Lien VTK, Hang NLK, Wang L, Lee LYH, Simmons CP, McMichael AJ, Farrar JJ, Askonas BA, Duong TN, Thai PQ, Thu Yen NT, Rowland-Jones SL, Hien NT, Horby P, Dong T. Identification of H5N1-specific T-cell responses in a high-risk cohort in vietnam indicates the existence of potential asymptomatic infections. J Infect Dis 2011; 205:20-7. [PMID: 22080094 DOI: 10.1093/infdis/jir689] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Most reported human H5N1 viral infections have been severe and were detected after hospital admission. A case ascertainment bias may therefore exist, with mild cases or asymptomatic infections going undetected. We sought evidence of mild or asymptomatic H5N1 infection by examining H5N1-specific T-cell and antibody responses in a high-risk cohort in Vietnam. METHODS Peripheral blood mononuclear cells were tested using interferon-γ enzyme-linked immunospot T assays measuring the response to peptides of influenza H5, H3, and H1 hemagglutinin (HA), N1 and N2 neuraminidase, and the internal proteins of H3N2. Horse erythrocyte hemagglutination inhibition assay was performed to detect antibodies against H5N1. RESULTS Twenty-four of 747 individuals demonstrated H5-specific T-cell responses but little or no cross-reactivity with H3 or H1 HA peptides. H5N1 peptide-specific T-cell lines that did not cross-react with H1 or H3 influenza virus HA peptides were generated. Four individuals also had antibodies against H5N1. CONCLUSIONS This is the first report of ex vivo H5 HA-specific T-cell responses in a healthy but H5N1-exposed population. Our results indicate that the presence of H5N1-specific T cells could be an additional diagnostic tool for asymptomatic H5N1 infection.
Collapse
Affiliation(s)
- Timothy J Powell
- MRC Human Immunology Unit, University of Oxford, John Radcliffe Hospital, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Homan EJ, Bremel RD. Patterns of predicted T-cell epitopes associated with antigenic drift in influenza H3N2 hemagglutinin. PLoS One 2011; 6:e26711. [PMID: 22039539 PMCID: PMC3200361 DOI: 10.1371/journal.pone.0026711] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 10/03/2011] [Indexed: 12/19/2022] Open
Abstract
Antigenic drift allowing escape from neutralizing antibodies is an important feature of transmission and survival of influenza viruses in host populations. Antigenic drift has been studied in particular detail for influenza A H3N2 and well defined antigenic clusters of this virus documented. We examine how host immunogenetics contributes to determination of the antibody spectrum, and hence the immune pressure bringing about antigenic drift. Using uTOPE™ bioinformatics analysis of predicted MHC binding, based on amino acid physical property principal components, we examined the binding affinity of all 9-mer and 15-mer peptides within the hemagglutinin 1 (HA1) of 447 H3N2 virus isolates to 35 MHC-I and 14 MHC-II alleles. We provide a comprehensive map of predicted MHC-I and MHC-II binding affinity for a broad array of HLA alleles for the H3N2 influenza HA1 protein. Each HLA allele exhibited a characteristic predicted binding pattern. Cluster analysis for each HLA allele shows that patterns based on predicted MHC binding mirror those described based on antibody binding. A single amino acid mutation or position displacement can result in a marked difference in MHC binding and hence potential T-helper function. We assessed the impact of individual amino acid changes in HA1 sequences between 10 virus isolates from 1968-2002, representative of antigenic clusters, to understand the changes in MHC binding over time. Gain and loss of predicted high affinity MHC-II binding sites with cluster transitions were documented. Predicted high affinity MHC-II binding sites were adjacent to antibody binding sites. We conclude that host MHC diversity may have a major determinant role in the antigenic drift of influenza A H3N2.
Collapse
Affiliation(s)
- E Jane Homan
- ioGenetics LLC, Madison, Wisconsin, United States of America.
| | | |
Collapse
|