1
|
Amoah S, Cao W, Sayedahmed EE, Wang Y, Kumar A, Mishina M, Eddins DJ, Wang WC, Burroughs M, Sheth M, Lee J, Shieh WJ, Ray SD, Bohannon CD, Ranjan P, Sharma SD, Hoehner J, Arthur RA, Gangappa S, Wakamatsu N, Johnston HR, Pohl J, Mittal SK, Sambhara S. The frequency and function of nucleoprotein-specific CD8 + T cells are critical for heterosubtypic immunity against influenza virus infection. J Virol 2024; 98:e0071124. [PMID: 39082839 PMCID: PMC11334528 DOI: 10.1128/jvi.00711-24] [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: 04/24/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) mediate host defense against viral and intracellular bacterial infections and tumors. However, the magnitude of CTL response and their function needed to confer heterosubtypic immunity against influenza virus infection are unknown. We addressed the role of CD8+ T cells in the absence of any cross-reactive antibody responses to influenza viral proteins using an adenoviral vector expressing a 9mer amino acid sequence recognized by CD8+ T cells. Our results indicate that both CD8+ T cell frequency and function are crucial for heterosubtypic immunity. Low morbidity, lower viral lung titers, low to minimal lung pathology, and better survival upon heterosubtypic virus challenge correlated with the increased frequency of NP-specific CTLs. NP-CD8+ T cells induced by differential infection doses displayed distinct RNA transcriptome profiles and functional properties. CD8+ T cells induced by a high dose of influenza virus secreted significantly higher levels of IFN-γ and exhibited higher levels of cytotoxic function. The mice that received NP-CD8+ T cells from the high-dose virus recipients through adoptive transfer had lower viral titers following viral challenge than those induced by the low dose of virus, suggesting differential cellular programming by antigen dose. Enhanced NP-CD8+ T-cell functions induced by a higher dose of influenza virus strongly correlated with the increased expression of cellular and metabolic genes, indicating a shift to a more glycolytic metabolic phenotype. These findings have implications for developing effective T cell vaccines against infectious diseases and cancer. IMPORTANCE Cytotoxic T lymphocytes (CTLs) are an important component of the adaptive immune system that clears virus-infected cells or tumor cells. Hence, developing next-generation vaccines that induce or recall CTL responses against cancer and infectious diseases is crucial. However, it is not clear if the frequency, function, or both are essential in conferring protection, as in the case of influenza. In this study, we demonstrate that both CTL frequency and function are crucial for providing heterosubtypic immunity to influenza by utilizing an Ad-viral vector expressing a CD8 epitope only to rule out the role of antibodies, single-cell RNA-seq analysis, as well as adoptive transfer experiments. Our findings have implications for developing T cell vaccines against infectious diseases and cancer.
Collapse
Affiliation(s)
- Samuel Amoah
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Weiping Cao
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Yuanyuan Wang
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Amrita Kumar
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Margarita Mishina
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Devon J. Eddins
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wen-Chien Wang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Mark Burroughs
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mili Sheth
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Justin Lee
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wun-Ju Shieh
- Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sean D. Ray
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Caitlin D. Bohannon
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Priya Ranjan
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Suresh D. Sharma
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jessica Hoehner
- Emory Integrated Computational Core, Emory Integrated Core Facilities, Emory University, Atlanta, Georgia, USA
| | - Robert A. Arthur
- Emory Integrated Computational Core, Emory Integrated Core Facilities, Emory University, Atlanta, Georgia, USA
| | - Shivaprakash Gangappa
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nobuko Wakamatsu
- Indiana Animal Disease Diagnostic Laboratory, Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - H. Richard Johnston
- Department of Human Genetics, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Jan Pohl
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Liu Z, Kabir MT, Chen S, Zhang H, Wakim LM, Rehm BHA. Intranasal Epitope-Polymer Vaccine Lodges Resident Memory T Cells Protecting Against Influenza Virus. Adv Healthc Mater 2024; 13:e2304188. [PMID: 38411375 PMCID: PMC11469178 DOI: 10.1002/adhm.202304188] [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: 11/27/2023] [Revised: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Intranasal vaccines, unlike injectable vaccines, boost immunity along the respiratory tract; this can significantly limit respiratory virus replication and shedding. There remains a need to develop mucosal adjuvants and vaccine delivery systems that are both safe and effective following intranasal administration. Here, biopolymer particles (BP) densely coated with repeats of MHC class I restricted immunodominant epitopes derived from influenza A virus namely NP366, a nucleoprotein-derived epitope and PA224, a polymerase acidic subunit derived epitope, are bioengineered. These BP-NP366/PA224 can be manufactured at a high yield and are obtained at ≈93% purity, exhibiting ambient-temperature stability. Immunological characterization includes comparing systemic and mucosal immune responses mounted following intramuscular or intranasal immunization. Immunization with BP-NP366/PA224 without adjuvant triggers influenza-specific CD8+ T cell priming and memory CD8+ T cell development. Co-delivery with the adjuvant poly(I:C) significantly boosts the size and functionality of the influenza-specific pulmonary resident memory CD8+ T cell pool. Intranasal, but not intramuscular delivery of BP-NP366/PA224 with poly(I:C), provides protection against influenza virus challenge. Overall, the BP approach demonstrates as a suitable antigen formulation for intranasal delivery toward induction of systemic protective T cell responses against influenza virus.
Collapse
Affiliation(s)
- Ziyang Liu
- Department of Microbiology and ImmunologyThe University of MelbourneThe Peter Doherty Institute for Infection and ImmunityMelbourneVictoria3000Australia
| | - Md. Tanvir Kabir
- Centre for Cell Factories and BiopolymersGriffith Institute for Drug DiscoveryGriffith UniversityDon Young RoadNathanQueensland4111Australia
| | - Shuxiong Chen
- Centre for Cell Factories and BiopolymersGriffith Institute for Drug DiscoveryGriffith UniversityDon Young RoadNathanQueensland4111Australia
| | - Heran Zhang
- Department of Microbiology and ImmunologyThe University of MelbourneThe Peter Doherty Institute for Infection and ImmunityMelbourneVictoria3000Australia
| | - Linda M. Wakim
- Department of Microbiology and ImmunologyThe University of MelbourneThe Peter Doherty Institute for Infection and ImmunityMelbourneVictoria3000Australia
| | - Bernd H. A. Rehm
- Centre for Cell Factories and BiopolymersGriffith Institute for Drug DiscoveryGriffith UniversityDon Young RoadNathanQueensland4111Australia
| |
Collapse
|
3
|
Zheng MZ, Tan TK, Villalon-Letelier F, Lau H, Deng YM, Fritzlar S, Valkenburg SA, Gu H, Poon LL, Reading PC, Townsend AR, Wakim LM. Single-cycle influenza virus vaccine generates lung CD8 + Trm that cross-react against viral variants and subvert virus escape mutants. SCIENCE ADVANCES 2023; 9:eadg3469. [PMID: 37683004 PMCID: PMC10491285 DOI: 10.1126/sciadv.adg3469] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/08/2023] [Indexed: 09/10/2023]
Abstract
Influenza virus-specific tissue-resident memory (Trm) CD8+ T cells located along the respiratory tract provide cross-strain protection against a breadth of influenza viruses. We show that immunization with a single-cycle influenza virus vaccine candidate (S-FLU) results in the deposition of influenza virus nucleoprotein (NP)-specific CD8+ Trm along the respiratory tract that were more cross-reactive against viral variants and less likely to drive the development of cytotoxic T lymphocyte (CTL) escape mutants, as compared to the lung memory NP-specific CD8+ T cell pool established following influenza infection. This immune profile was linked to the limited inflammatory response evoked by S-FLU vaccination, which increased TCR repertoire diversity within the memory CD8+ T cell compartment. Cumulatively, this work shows that S-FLU vaccination evokes a clonally diverse, cross-reactive memory CD8+ T cell pool, which protects against severe disease without driving the virus to rapidly evolve and escape, and thus represents an attractive vaccine for use against rapidly mutating influenza viruses.
Collapse
Affiliation(s)
- Ming Z. M. Zheng
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Tiong Kit Tan
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, UK
| | - Fernando Villalon-Letelier
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Hilda Lau
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Yi-Mo Deng
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Sophie A. Valkenburg
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Haogao Gu
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Leo L. M. Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Centre for Immunology & Infection, Hong Kong Science Park, Hong Kong SAR, China
| | - Patrick C. Reading
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| | - Alain R. Townsend
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, OX3 9DS Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences, Oxford Institute, University of Oxford, OX3 7FZ Oxford, UK
| | - Linda M. Wakim
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
| |
Collapse
|
4
|
Price MJ, Scharer CD, Kania AK, Randall TD, Boss JM. Conserved Epigenetic Programming and Enhanced Heme Metabolism Drive Memory B Cell Reactivation. THE JOURNAL OF IMMUNOLOGY 2021; 206:1493-1504. [PMID: 33627377 DOI: 10.4049/jimmunol.2000551] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/25/2021] [Indexed: 02/01/2023]
Abstract
Memory B cells (MBCs) have enhanced capabilities to differentiate to plasma cells and generate a rapid burst of Abs upon secondary stimulation. To determine if MBCs harbor an epigenetic landscape that contributes to increased differentiation potential, we derived the chromatin accessibility and transcriptomes of influenza-specific IgM and IgG MBCs compared with naive cells. MBCs possessed an accessible chromatin architecture surrounding plasma cell-specific genes, as well as altered expression of transcription factors and genes encoding cell cycle, chemotaxis, and signal transduction processes. Intriguingly, this MBC signature was conserved between humans and mice. MBCs of both species possessed a heightened heme signature compared with naive cells. Differentiation in the presence of hemin enhanced oxidative phosphorylation metabolism and MBC differentiation into Ab-secreting plasma cells. Thus, these data define conserved MBC transcriptional and epigenetic signatures that include a central role for heme and multiple other pathways in augmenting MBC reactivation potential.
Collapse
Affiliation(s)
- Madeline J Price
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Anna K Kania
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Troy D Randall
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322; .,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
5
|
Roy S, Williams CM, Wijesundara DK, Furuya Y. Impact of Pre-Existing Immunity to Influenza on Live-Attenuated Influenza Vaccine (LAIV) Immunogenicity. Vaccines (Basel) 2020; 8:E683. [PMID: 33207559 PMCID: PMC7711626 DOI: 10.3390/vaccines8040683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022] Open
Abstract
During the previous influenza seasons, between 2010 and 2016, the live attenuated influenza vaccine (LAIV) provided variable efficacy against influenza in the U.S., causing the recommendation against the use of the LAIV. In striking contrast, pre-clinical studies have repeatedly demonstrated superior efficacy of LAIV against mismatched influenza viruses, compared to inactivated influenza vaccines (IIV). This disparity in reported vaccine efficacies between pre-clinical and clinical studies may in part be explained by limitations of the animal models of influenza. In particular, the absence of pre-existing immunity in animal models has recently emerged as a potential explanation for the discrepancies between preclinical findings and human studies. This commentary focuses on the potential impact of pre-existing immunity on LAIV induced immunogenicity with an emphasis on cross-protective immunity.
Collapse
Affiliation(s)
- Sreeja Roy
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| | - Clare M. Williams
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| | - Danushka K. Wijesundara
- The School of Chemistry and Molecular Biosciences, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland 4072, Australia;
| | - Yoichi Furuya
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA; (S.R.); (C.M.W.)
| |
Collapse
|
6
|
Stone SL, Peel JN, Scharer CD, Risley CA, Chisolm DA, Schultz MD, Yu B, Ballesteros-Tato A, Wojciechowski W, Mousseau B, Misra RS, Hanidu A, Jiang H, Qi Z, Boss JM, Randall TD, Brodeur SR, Goldrath AW, Weinmann AS, Rosenberg AF, Lund FE. T-bet Transcription Factor Promotes Antibody-Secreting Cell Differentiation by Limiting the Inflammatory Effects of IFN-γ on B Cells. Immunity 2019; 50:1172-1187.e7. [PMID: 31076359 PMCID: PMC6929688 DOI: 10.1016/j.immuni.2019.04.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/04/2019] [Accepted: 04/12/2019] [Indexed: 01/07/2023]
Abstract
Although viral infections elicit robust interferon-γ (IFN-γ) and long-lived antibody-secreting cell (ASC) responses, the roles for IFN-γ and IFN-γ-induced transcription factors (TFs) in ASC development are unclear. We showed that B cell intrinsic expression of IFN-γR and the IFN-γ-induced TF T-bet were required for T-helper 1 cell-induced differentiation of B cells into ASCs. IFN-γR signaling induced Blimp1 expression in B cells but also initiated an inflammatory gene program that, if not restrained, prevented ASC formation. T-bet did not affect Blimp1 upregulation in IFN-γ-activated B cells but instead regulated chromatin accessibility within the Ifng and Ifngr2 loci and repressed the IFN-γ-induced inflammatory gene program. Consistent with this, B cell intrinsic T-bet was required for formation of long-lived ASCs and secondary ASCs following viral, but not nematode, infection. Therefore, T-bet facilitates differentiation of IFN-γ-activated inflammatory effector B cells into ASCs in the setting of IFN-γ-, but not IL-4-, induced inflammatory responses.
Collapse
Affiliation(s)
- Sara L Stone
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jessica N Peel
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Christopher A Risley
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Danielle A Chisolm
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael D Schultz
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Bingfei Yu
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Wojciech Wojciechowski
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Betty Mousseau
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ravi S Misra
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Adedayo Hanidu
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Huiping Jiang
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Zhenhao Qi
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Troy D Randall
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott R Brodeur
- Boerhinger Ingelheim Pharmaceutical Inc., Ridgefield, CT 06877, USA
| | - Ananda W Goldrath
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Amy S Weinmann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alexander F Rosenberg
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Informatics Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Frances E Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
7
|
Gonçalves E, Bonduelle O, Soria A, Loulergue P, Rousseau A, Cachanado M, Bonnabau H, Thiebaut R, Tchitchek N, Behillil S, van der Werf S, Vogt A, Simon T, Launay O, Combadière B. Innate gene signature distinguishes humoral versus cytotoxic responses to influenza vaccination. J Clin Invest 2019; 129:1960-1971. [PMID: 30843873 DOI: 10.1172/jci125372] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Systems vaccinology allows cutting-edge analysis of innate biomarkers of vaccine efficacy. We have been exploring novel strategies to shape the adaptive immune response, by targeting innate immune cells through novel immunization routes. METHODS This randomized phase I/II clinical study (n=60 healthy subjects aged 18-45 years old) used transcriptomic analysis to discover early biomarkers of immune response quality after transcutaneous (t.c.), intradermal (i.d.), and intramuscular (i.m.) administration of a trivalent influenza vaccine (TIV season 2012-2013) (1:1:1 ratio). Safety and immunogenicity (hemagglutinin inhibition (HI), microneutralization (MN) antibodies and CD4, CD8 effector T cells) were measured at baseline Day (D)0 and at D21. Blood transcriptome was analyzed at D0 and D1. RESULTS TIV-specific CD8+GranzymeB+(GRZ) T cells appeared in more individuals immunized by the t.c. and i.d. routes, while immunization by the i.d. and i.m. routes prompted high levels of HI antibody titers and MN against A/H1N1 and A/H3N2 influenza viral strains. The early innate gene signature anticipated immunological outcome by discriminating two clusters of individuals with either distinct humoral or CD8 cytotoxic responses. Several pathways explained this dichotomy confirmed by nine genes and serum level of CXCL10 were correlated with either TIV-specific cytotoxic CD8+GRZ+ T-cell or antibody responses. A logistic regression analysis demonstrated that these nine genes and serum levels of CXCL10 (D1/D0) best foreseen TIV-specific CD8+GRZ+ T-cell and antibody responses at D21. CONCLUSION This study provides new insight into the impact of immunization routes and innate signature in the quality of adaptive immune responses.
Collapse
Affiliation(s)
- Eléna Gonçalves
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France
| | - Olivia Bonduelle
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France
| | - Angèle Soria
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France.,Service de Dermatologie et Allergologie, Hôpital Tenon, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Pierre Loulergue
- Université Paris Descartes, Sorbonne Paris Cité, Centre d'Investigation Clinique Cochin Pasteur, INSERM CIC 1417, French Clinical Research Infrastructure Network, Innovative Clinical Research Network in Vaccinology, AP-HP, Hôpital Cochin, Paris, France
| | - Alexandra Rousseau
- Department of Clinical Pharmacology and Clinical Research Platform of East of Paris, Assistance Publique-Hôpitaux de Paris, Paris, France. Sorbonne Université, Paris, France
| | - Marine Cachanado
- Department of Clinical Pharmacology and Clinical Research Platform of East of Paris, Assistance Publique-Hôpitaux de Paris, Paris, France. Sorbonne Université, Paris, France
| | - Henri Bonnabau
- INSERM U1219, INRIA SISTM, Université de Bordeaux, Bordeaux France
| | | | - Nicolas Tchitchek
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Institut de Biologie François Jacob, 92265 Fontenay-aux-Roses, France
| | - Sylvie Behillil
- Institut Pasteur, CNR des Virus des Infections Respiratoires, Département de Virologie and Centre National de Recherche Scientifique UMR CNRS 3569, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Unité de Génétique Moléculaire des Virus à ARN, Paris, France
| | - Sylvie van der Werf
- Institut Pasteur, CNR des Virus des Infections Respiratoires, Département de Virologie and Centre National de Recherche Scientifique UMR CNRS 3569, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Unité de Génétique Moléculaire des Virus à ARN, Paris, France
| | - Annika Vogt
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France.,Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tabassome Simon
- Department of Clinical Pharmacology and Clinical Research Platform of East of Paris, Assistance Publique-Hôpitaux de Paris, Paris, France. Sorbonne Université, Paris, France
| | - Odile Launay
- Université Paris Descartes, Sorbonne Paris Cité, Centre d'Investigation Clinique Cochin Pasteur, INSERM CIC 1417, French Clinical Research Infrastructure Network, Innovative Clinical Research Network in Vaccinology, AP-HP, Hôpital Cochin, Paris, France
| | - Behazine Combadière
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France
| |
Collapse
|
8
|
Epstein SL. Universal Influenza Vaccines: Progress in Achieving Broad Cross-Protection In Vivo. Am J Epidemiol 2018; 187:2603-2614. [PMID: 30084906 DOI: 10.1093/aje/kwy145] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/09/2018] [Indexed: 01/08/2023] Open
Abstract
Despite all we have learned since 1918 about influenza virus and immunity, available influenza vaccines remain inadequate to control outbreaks of unexpected strains. Universal vaccines not requiring strain matching would be a major improvement. Their composition would be independent of predicting circulating viruses and thus potentially effective against unexpected drift or pandemic strains. This commentary explores progress with candidate universal vaccines based on various target antigens. Candidates include vaccines based on conserved viral proteins such as nucleoprotein and matrix, on the conserved hemagglutinin (HA) stem, and various combinations. Discussion covers the differing evidence for each candidate vaccine demonstrating protection in animals against influenza viruses of widely divergent HA subtypes and groups; durability of protection; routes of administration, including mucosal, providing local immunity; and reduction of transmission. Human trials of some candidate universal vaccines have been completed or are underway. Interestingly, the HA stem, like nucleoprotein and matrix, induces immunity that permits some virus replication and emergence of escape mutants fit enough to cause disease. Vaccination with multiple target antigens will thus have advantages over use of single antigens. Ultimately, a universal vaccine providing long-term protection against all influenza virus strains might contribute to pandemic control and routine vaccination.
Collapse
Affiliation(s)
- Suzanne L Epstein
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
9
|
Keshavarz M, Mirzaei H, Salemi M, Momeni F, Mousavi MJ, Sadeghalvad M, Arjeini Y, Solaymani-Mohammadi F, Sadri Nahand J, Namdari H, Mokhtari-Azad T, Rezaei F. Influenza vaccine: Where are we and where do we go? Rev Med Virol 2018; 29:e2014. [PMID: 30408280 DOI: 10.1002/rmv.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022]
Abstract
The alarming rise of morbidity and mortality caused by influenza pandemics and epidemics has drawn attention worldwide since the last few decades. This life-threatening problem necessitates the development of a safe and effective vaccine to protect against incoming pandemics. The currently available flu vaccines rely on inactivated viral particles, M2e-based vaccine, live attenuated influenza vaccine (LAIV) and virus like particle (VLP). While inactivated vaccines can only induce systemic humoral responses, LAIV and VLP vaccines stimulate both humoral and cellular immune responses. Yet, these vaccines have limited protection against newly emerging viral strains. These strains, however, can be targeted by universal vaccines consisting of conserved viral proteins such as M2e and capable of inducing cross-reactive immune response. The lack of viral genome in VLP and M2e-based vaccines addresses safety concern associated with existing attenuated vaccines. With the emergence of new recombinant viral strains each year, additional effort towards developing improved universal vaccine is warranted. Besides various types of vaccines, microRNA and exosome-based vaccines have been emerged as new types of influenza vaccines which are associated with new and effective properties. Hence, development of a new generation of vaccines could contribute to better treatment of influenza.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Salemi
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Fatemeh Momeni
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Mousavi
- Department of Immunology and Allergy, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.,Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Sadeghalvad
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Arjeini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Solaymani-Mohammadi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haideh Namdari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Extensive T cell cross-reactivity between diverse seasonal influenza strains in the ferret model. Sci Rep 2018; 8:6112. [PMID: 29666412 PMCID: PMC5904180 DOI: 10.1038/s41598-018-24394-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/27/2018] [Indexed: 12/03/2022] Open
Abstract
Influenza virus causes widespread, yearly epidemics by accumulating surface protein mutations to escape neutralizing antibodies established from prior exposure. In contrast to antibody epitopes, T cell mediated immunity targets influenza epitopes that are more highly conserved and have potential for cross-protection. The extent of T cell cross-reactivity between a diverse array of contemporary and historical influenza strains was investigated in ferrets challenged with 2009 pandemic H1N1 influenza or the seasonal H3N2 strain, A/Perth/16/2009. Post-challenge cell-mediated immune responses demonstrated extensive cross-reactivity with a wide variety of contemporary and historical influenza A strains as well as influenza B. Responses in peripheral blood were undetectable by 36d post-challenge, but cross-reactivity persisted in spleen. The strongest responses targeted peptides from the NP protein and demonstrated cross-reactivity in both the CD4+ and CD8+ T cell populations. Cross-reactive CD4+ T cells also targeted HA and NA epitopes, while cross-reactive CD8+ T cells targeted internal M1, NS2, and PA. T cell epitopes demonstrated extensive cross-reactivity between diverse influenza strains in outbred animals, with NP implicated as a significant antigenic target demonstrating extensive cross-reactivity for both CD4+ and CD8+ T cells.
Collapse
|
11
|
Pizzolla A, Nguyen TH, Sant S, Jaffar J, Loudovaris T, Mannering SI, Thomas PG, Westall GP, Kedzierska K, Wakim LM. Influenza-specific lung-resident memory T cells are proliferative and polyfunctional and maintain diverse TCR profiles. J Clin Invest 2018; 128:721-733. [PMID: 29309047 DOI: 10.1172/jci96957] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/14/2017] [Indexed: 01/01/2023] Open
Abstract
The human lung harbors a large population of resident memory T cells (Trm cells). These cells are perfectly positioned to mediate rapid protection against respiratory pathogens such as influenza virus, a highly contagious respiratory pathogen that continues to be a major public health burden. Animal models show that influenza-specific lung CD8+ Trm cells are indispensable for crossprotection against pulmonary infection with different influenza virus strains. However, it is not known whether influenza-specific CD8+ Trm cells present within the human lung have the same critical role in modulating the course of the disease. Here, we showed that human lung contains a population of CD8+ Trm cells that are highly proliferative and have polyfunctional progeny. We observed that different influenza virus-specific CD8+ T cell specificities differentiated into Trm cells with varying efficiencies and that the size of the influenza-specific CD8+ T cell population persisting in the lung directly correlated with the efficiency of differentiation into Trm cells. To our knowledge, we provide the first ex vivo dissection of paired T cell receptor (TCR) repertoires of human influenza-specific CD8+ Trm cells. Our data reveal diverse TCR profiles within the human lung Trm cells and a high degree of clonal sharing with other CD8+ T cell populations, a feature important for effective T cell function and protection against the generation of viral-escape mutants.
Collapse
Affiliation(s)
- Angela Pizzolla
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Thi Ho Nguyen
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sneha Sant
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jade Jaffar
- Lung Transplant Service, Alfred Hospital, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Tom Loudovaris
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Glen P Westall
- Lung Transplant Service, Alfred Hospital, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Jung YJ, Lee YT, Ngo VL, Cho YH, Ko EJ, Hong SM, Kim KH, Jang JH, Oh JS, Park MK, Kim CH, Sun J, Kang SM. Heat-killed Lactobacillus casei confers broad protection against influenza A virus primary infection and develops heterosubtypic immunity against future secondary infection. Sci Rep 2017; 7:17360. [PMID: 29234060 PMCID: PMC5727132 DOI: 10.1038/s41598-017-17487-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/23/2017] [Indexed: 02/08/2023] Open
Abstract
Lactic acid bacteria (LAB) are the common probiotics. Here, we investigated the antiviral protective effects of heat-killed LAB strain Lactobacillus casei DK128 (DK128) on influenza viruses. Intranasal treatment of mice with DK128 conferred protection against different subtypes of influenza viruses by lessening weight loss and lowering viral loads. Protection via heat-killed DK128 was correlated with an increase in alveolar macrophage cells in the lungs and airways, early induction of virus specific antibodies, reduced levels of pro-inflammatory cytokines and innate immune cells. Importantly, the mice that were protected against primary viral infection as a result of heat-killed DK128 pretreatment developed subsequent heterosubtypic immunity against secondary virus infection. For protection against influenza virus via heat-killed DK128 pretreatment, B cells and partially CD4 T cells but not CD8 T cells were required as inferred from studies using knockout mouse models. Our study provides insight into how hosts can be equipped with innate and adaptive immunity via heat-killed DK128 treatment to protect against influenza virus, supporting that heat-killed LAB may be developed as anti-virus probiotics.
Collapse
Affiliation(s)
- Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Vu Le Ngo
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Young-Hee Cho
- Department of Animal Resource Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 330-714, Korea
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Sung-Moon Hong
- Department of Animal Resource Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 330-714, Korea
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA
| | - Ji-Hun Jang
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, 331-858, Korea
| | - Joon-Suk Oh
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, 331-858, Korea
| | - Min-Kyung Park
- Department of Human Nutrition and Food Science, Chungwoon University, Namjang-Ri, Hongsung-Eup, Hongsung-Kun, Chungnam, 350-701, Korea
| | - Cheol-Hyun Kim
- Department of Animal Resource Science, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, 330-714, Korea
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, 30303, USA.
| |
Collapse
|
13
|
Yan AWC, Cao P, Heffernan JM, McVernon J, Quinn KM, La Gruta NL, Laurie KL, McCaw JM. Modelling cross-reactivity and memory in the cellular adaptive immune response to influenza infection in the host. J Theor Biol 2016; 413:34-49. [PMID: 27856216 DOI: 10.1016/j.jtbi.2016.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/02/2016] [Accepted: 11/05/2016] [Indexed: 01/05/2023]
Abstract
The cellular adaptive immune response plays a key role in resolving influenza infection. Experiments where individuals are successively infected with different strains within a short timeframe provide insight into the underlying viral dynamics and the role of a cross-reactive immune response in resolving an acute infection. We construct a mathematical model of within-host influenza viral dynamics including three possible factors which determine the strength of the cross-reactive cellular adaptive immune response: the initial naive T cell number, the avidity of the interaction between T cells and the epitopes presented by infected cells, and the epitope abundance per infected cell. Our model explains the experimentally observed shortening of a second infection when cross-reactivity is present, and shows that memory in the cellular adaptive immune response is necessary to protect against a second infection.
Collapse
Affiliation(s)
- Ada W C Yan
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Pengxing Cao
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jane M Heffernan
- Department of Mathematics and Statistics, York University, Toronto, Ontario, Canada M3J 1P3; Modelling Infection and Immunity Lab, Centre for Disease Modelling, York Institute for Health Research, York University, Toronto, Ontario, Canada M3J 1P3
| | - Jodie McVernon
- Doherty Epidemiology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC 3010, Australia; Modelling and Simulation, Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Kylie M Quinn
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Nicole L La Gruta
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Karen L Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; School of Applied and Biomedical Sciences, Federation University, Churchill, VIC 3842, Australia; Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - James M McCaw
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC 3010, Australia; Modelling and Simulation, Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia.
| |
Collapse
|
14
|
Lee YN, Lee YT, Kim MC, Gewirtz AT, Kang SM. A Novel Vaccination Strategy Mediating the Induction of Lung-Resident Memory CD8 T Cells Confers Heterosubtypic Immunity against Future Pandemic Influenza Virus. THE JOURNAL OF IMMUNOLOGY 2016; 196:2637-45. [PMID: 26864033 DOI: 10.4049/jimmunol.1501637] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 01/08/2016] [Indexed: 12/21/2022]
Abstract
The currently used vaccine strategy to combat influenza A virus (IAV) aims to provide highly specific immunity to circulating seasonal IAV strains. However, the outbreak of 2009 influenza pandemic highlights the danger in this strategy. In this study, we tested the hypothesis that universal vaccination that offers broader but weaker protection would result in cross protective T cell responses after primary IAV infection, which would subsequently provide protective immunity against future pandemic strains. Specifically, we used tandem repeat extracellular domain of M2 (M2e) epitopes on virus-like particles (M2e5x VLP) that induced heterosubtypic immunity by eliciting Abs to a conserved M2e epitope. M2e5x VLP was found to be superior to strain-specific current split vaccine in conferring heterosubtypic cross protection and in equipping the host with cross-protective lung-resident nucleoprotein-specific memory CD8(+) T cell responses to a subsequent secondary infection with a new pandemic potential strain. Immune correlates for subsequent heterosubtypic immunity by M2e5x VLP vaccination were found to be virus-specific CD8(+) T cells secreting IFN-γ and expressing lung-resident memory phenotypic markers CD69(+) and CD103(+) as well as M2e Abs. Hence, vaccination with M2e5x VLP may be developable as a new strategy to combat future pandemic outbreaks.
Collapse
Affiliation(s)
- Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and Animal and Plant Quarantine Agency, Anyang, Gyeonggi-do 14089, Republic of Korea
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| |
Collapse
|
15
|
Inactivated Influenza Vaccine That Provides Rapid, Innate-Immune-System-Mediated Protection and Subsequent Long-Term Adaptive Immunity. mBio 2015; 6:e01024-15. [PMID: 26507227 PMCID: PMC4626850 DOI: 10.1128/mbio.01024-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The continual threat to global health posed by influenza has led to increased efforts to improve the effectiveness of influenza vaccines for use in epidemics and pandemics. We show in this study that formulation of a low dose of inactivated detergent-split influenza vaccine with a Toll-like receptor 2 (TLR2) agonist-based lipopeptide adjuvant (R4Pam2Cys) provides (i) immediate, antigen-independent immunity mediated by the innate immune system and (ii) significant enhancement of antigen-dependent immunity which exhibits an increased breadth of effector function. Intranasal administration of mice with vaccine formulated with R4Pam2Cys but not vaccine alone provides protection against both homologous and serologically distinct (heterologous) viral strains within a day of administration. Vaccination in the presence of R4Pam2Cys subsequently also induces high levels of systemic IgM, IgG1, and IgG2b antibodies and pulmonary IgA antibodies that inhibit hemagglutination (HA) and neuraminidase (NA) activities of homologous but not heterologous virus. Improved primary virus nucleoprotein (NP)-specific CD8+ T cell responses are also induced by the use of R4Pam2Cys and are associated with robust recall responses to provide heterologous protection. These protective effects are demonstrated in wild-type and antibody-deficient animals but not in those depleted of CD8+ T cells. Using a contact-dependent virus transmission model, we also found that heterologous virus transmission from vaccinated mice to naive mice is significantly reduced. These results demonstrate the potential of adding a TLR2 agonist to an existing seasonal influenza vaccine to improve its utility by inducing immediate short-term nonspecific antiviral protection and also antigen-specific responses to provide homologous and heterologous immunity. The innate and adaptive immune systems differ in mechanisms, specificities, and times at which they take effect. The innate immune system responds within hours of exposure to infectious agents, while adaptive immunity takes several days to become effective. Here we show, by using a simple lipopeptide-based TLR2 agonist, that an influenza detergent-split vaccine can be made to simultaneously stimulate and amplify both systems to provide immediate antiviral protection while giving the adaptive immune system time to implement long-term immunity. Both types of immunity induced by this approach protect against vaccine-matched as well as unrelated virus strains and potentially even against strains yet to be encountered. Conferring dual functionality to influenza vaccines is beneficial for improving community protection, particularly during periods between the onset of an outbreak and the time when a vaccine becomes available or in scenarios in which mass vaccination with a strain to which the population is immunologically naive is imperative.
Collapse
|
16
|
Antibody-targeted vaccination to lung dendritic cells generates tissue-resident memory CD8 T cells that are highly protective against influenza virus infection. Mucosal Immunol 2015; 8:1060-71. [PMID: 25586557 DOI: 10.1038/mi.2014.133] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/24/2014] [Indexed: 02/04/2023]
Abstract
Influenza virus gains entry into the body by inhalation and initiates its replication cycle within the lung. The early stage of infection, while the virus is confined to the lung mucosa, provides the ideal window of opportunity for an effective immune response to control the infection. Tissue-resident memory (Trm) CD8 T cells, located in a variety of tissues including the lung, are ideally situated to act during this window and stall the infection. The factors involved in the differentiation of lung Trm cells remain poorly defined. We demonstrate that recognition of antigen presented locally by dendritic cells (DCs) and transforming growth factor-β (TGFβ) signaling are both required. We exploited this knowledge to develop an antibody-targeted vaccination approach to generate lung Trm cells. Delivering antigen exclusively to respiratory DCs results in the development of lung CD8 Trm cells that are highly protective against lethal influenza challenge. Our results describe an effective vaccination strategy that protects against influenza virus infection.
Collapse
|
17
|
Choi EH, Song MS, Park SJ, Pascua PNQ, Baek YH, Kwon HI, Kim EH, Kim S, Jang HK, Poo H, Kim CJ, Choi YK. Development of a dual-protective live attenuated vaccine against H5N1 and H9N2 avian influenza viruses by modifying the NS1 gene. Arch Virol 2015; 160:1729-40. [PMID: 25959557 DOI: 10.1007/s00705-015-2442-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/26/2015] [Indexed: 11/26/2022]
Abstract
An increasing number of outbreaks of avian influenza H5N1 and H9N2 viruses in poultry have caused serious economic losses and raised concerns for human health due to the risk of zoonotic transmission. However, licensed H5N1 and H9N2 vaccines for animals and humans have not been developed. Thus, to develop a dual H5N1 and H9N2 live-attenuated influenza vaccine (LAIV), the HA and NA genes from a virulent mouse-adapted avian H5N2 (A/WB/Korea/ma81/06) virus and a recently isolated chicken H9N2 (A/CK/Korea/116/06) virus, respectively, were introduced into the A/Puerto Rico/8/34 backbone expressing truncated NS1 proteins (NS1-73, NS1-86, NS1-101, NS1-122) but still possessing a full-length NS gene. Two H5N2/NS1-LAIV viruses (H5N2/NS1-86 and H5N2/NS1-101) were highly attenuated compared with the full-length and remaining H5N2/NS-LAIV viruses in a mouse model. Furthermore, viruses containing NS1 modifications were found to induce more IFN-β activation than viruses with full-length NS1 proteins and were correspondingly attenuated in mice. Intranasal vaccination with a single dose (10(4.0) PFU/ml) of these viruses completely protected mice from a lethal challenge with the homologous A/WB/Korea/ma81/06 (H5N2), heterologous highly pathogenic A/EM/Korea/W149/06 (H5N1), and heterosubtypic highly virulent mouse-adapted H9N2 viruses. This study clearly demonstrates that the modified H5N2/NS1-LAIV viruses attenuated through the introduction of mutations in the NS1 coding region display characteristics that are desirable for live attenuated vaccines and hold potential as vaccine candidates for mammalian hosts.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Chickens
- Female
- Humans
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H9N2 Subtype/genetics
- Influenza A Virus, H9N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Poultry Diseases/immunology
- Poultry Diseases/prevention & control
- Poultry Diseases/virology
- Vaccination
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Viral Nonstructural Proteins/administration & dosage
- Viral Nonstructural Proteins/genetics
- Viral Nonstructural Proteins/immunology
Collapse
Affiliation(s)
- Eun-hye Choi
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, 12 Gaeshin-Dong Heungduk-Ku, Cheongju, 361-763, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Guo H, Baker SF, Martínez-Sobrido L, Topham DJ. Induction of CD8 T cell heterologous protection by a single dose of single-cycle infectious influenza virus. J Virol 2014; 88:12006-16. [PMID: 25100831 PMCID: PMC4178714 DOI: 10.1128/jvi.01847-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/01/2014] [Indexed: 12/13/2022] Open
Abstract
The effector functions of specific CD8 T cells are crucial in mediating influenza heterologous protection. However, new approaches for influenza vaccines that can trigger effective CD8 T cell responses have not been extensively explored. We report here the generation of single-cycle infectious influenza virus that lacks a functional hemagglutinin (HA) gene on an X31 genetic background and demonstrate its potential for triggering protective CD8 T cell immunity against heterologous influenza virus challenge. In vitro, X31-sciIV can infect MDCK cells, but infectious virions are not produced unless HA is transcomplemented. In vivo, intranasal immunization with X31-sciIV does not cause any clinical symptoms in mice but generates influenza-specific CD8 T cells in lymphoid (mediastinal lymph nodes and spleen) and nonlymphoid tissues, including lung and bronchoalveolar lavage fluid, as measured by H2-Db NP366 and PA224 tetramer staining. In addition, a significant proportion of X31-sciIV-induced antigen-specific respiratory CD8 T cells expressed VLA-1, a marker that is associated with heterologous influenza protection. Further, these influenza-specific CD8 T cells produce antiviral cytokines when stimulated with NP366 and PA224 peptides, indicating that CD8 T cells triggered by X31-sciIV are functional. When challenged with a lethal dose of heterologous PR8 virus, X31-sciIV-primed mice were fully protected from death. However, when CD8 T cells were depleted after priming or before priming, mice could not effectively control virus replication or survive the lethal challenge, indicating that X31-sciIV-induced memory CD8 T cells mediate the heterologous protection. Thus, our results demonstrate the potential for sciIV as a CD8 T cell-inducing vaccine. Importance: One of the challenges for influenza prevention is the existence of multiple influenza virus subtypes and variants and the fact that new strains can emerge yearly. Numerous studies have indicated that the effector functions of specific CD8 T cells are crucial in mediating influenza heterologous protection. However, influenza vaccines that can trigger effective CD8 T cell responses for heterologous protection have not been developed. We report here the generation of an X31 (H3N2) virus-derived single-cycle infectious influenza virus, X31-sciIV. A one-dose immunization with X31-sciIV is capable of inducing functional influenza virus-specific CD8 T cells that can be recruited into respiratory tissues and provide protection against lethal heterologous challenge. Without these cells, protection against lethal challenge was essentially lost. Our data indicate that an influenza vaccine that primarily relies on CD8 T cells for protection could be developed.
Collapse
Affiliation(s)
- Hailong Guo
- Center for Infectious Diseases and Vaccine Immunology, Rochester General Hospital Research Institute, Rochester, New York, USA
| | - Steven F Baker
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, New York, USA
| |
Collapse
|
19
|
Quiñones-Parra S, Loh L, Brown LE, Kedzierska K, Valkenburg SA. Universal immunity to influenza must outwit immune evasion. Front Microbiol 2014; 5:285. [PMID: 24971078 PMCID: PMC4054793 DOI: 10.3389/fmicb.2014.00285] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/23/2014] [Indexed: 12/23/2022] Open
Abstract
Although an influenza vaccine has been available for 70 years, influenza virus still causes seasonal epidemics and worldwide pandemics. Currently available vaccines elicit strain-specific antibody (Ab) responses to the surface haemagglutinin (HA) and neuraminidase (NA) proteins, but these can be ineffective against serologically-distinct viral variants and novel subtypes. Thus, there is a great need for cross-protective or "universal" influenza vaccines to overcome the necessity for annual immunization against seasonal influenza and to provide immunity to reduce the severity of infection with pandemic or outbreak viruses. It is well established that natural influenza infection can provide cross-reactive immunity that can reduce the impact of infection with distinct influenza type A strains and subtypes, including H1N1, H3N2, H2N2, H5N1, and H7N9. The key to generating universal influenza immunity through vaccination is to target functionally-conserved regions of the virus, which include epitopes on the internal proteins for cross-reactive T cell immunity or on the HA stem for broadly reactive Ab responses. In the wake of the 2009 H1N1 pandemic, broadly neutralizing antibodies (bnAbs) have been characterized and isolated from convalescent and vaccinated individuals, inspiring development of new vaccination techniques to elicit such responses. Induction of influenza-specific T cell responses through vaccination has also been recently examined in clinical trials. Strong evidence is available from human and animal models of influenza to show that established influenza-specific T cell memory can reduce viral shedding and symptom severity. However, the published evidence also shows that CD8(+) T cells can efficiently select immune escape mutants early after influenza virus infection. Here, we discuss universal immunity to influenza viruses mediated by both cross-reactive T cells and Abs, the mechanisms of immune evasion in influenza, and propose how to counteract commonly occurring immune-escape variants.
Collapse
Affiliation(s)
- Sergio Quiñones-Parra
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville VIC, Australia
| | - Liyen Loh
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville VIC, Australia
| | - Lorena E Brown
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville VIC, Australia
| | - Sophie A Valkenburg
- Centre for Influenza Research and School of Public Health, The University of Hong Kong Hong Kong, China
| |
Collapse
|
20
|
Abstract
The challenges in successful vaccination against influenza using conventional approaches lie in their variable efficacy in different age populations, the antigenic variability of the circulating virus, and the production and manufacturing limitations to ensure safe, timely, and adequate supply of vaccine. The conventional influenza vaccine platform is based on stimulating immunity against the major neutralizing antibody target, hemagglutinin (HA), by virus attenuation or inactivation. Improvements to this conventional system have focused primarily on improving production and immunogenicity. Cell culture, reverse genetics, and baculovirus expression technology allow for safe and scalable production, while adjuvants, dose variation, and alternate routes of delivery aim to improve vaccine immunogenicity. Fundamentally different approaches that are currently under development hope to signal new generations of influenza vaccines. Such approaches target nonvariable regions of antigenic proteins, with the idea of stimulating cross-protective antibodies and thus creating a "universal" influenza vaccine. While such approaches have obvious benefits, there are many hurdles yet to clear. Here, we discuss the process and challenges of the current influenza vaccine platform as well as new approaches that are being investigated based on the same antigenic target and newer technologies based on different antigenic targets.
Collapse
|
21
|
Reber A, Katz J. Immunological assessment of influenza vaccines and immune correlates of protection. Expert Rev Vaccines 2013; 12:519-36. [PMID: 23659300 DOI: 10.1586/erv.13.35] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Influenza vaccines remain the primary public health tool in reducing the ever-present burden of influenza and its complications. In seeking more immunogenic, more effective and more broadly cross-protective influenza vaccines, the landscape of influenza vaccines is rapidly expanding, both in near-term advances and next-generation vaccine design. Although the first influenza vaccines were licensed over 60 years ago, the hemagglutination-inhibition antibody titer is currently the only universally accepted immune correlate of protection against influenza. However, hemagglutination-inhibition titers appear to be less effective at predicting protection in populations at high risk for severe influenza disease; older adults, young children and those with certain medical conditions. The lack of knowledge and validated methods to measure alternate immune markers of protection against influenza remain a substantial barrier to the development of more immunogenic, broadly cross-reactive and effective influenza vaccines. Here, the authors review the knowledge of immune effectors of protection against influenza and discuss assessment methods for a broader range of immunological parameters that could be considered in the evaluation of traditional or new-generation influenza vaccines.
Collapse
Affiliation(s)
- Adrian Reber
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road Atlanta, GA 30333, USA
| | | |
Collapse
|
22
|
Gildea S, Quinlivan M, Murphy BA, Cullinane A. Humoral response and antiviral cytokine expression following vaccination of thoroughbred weanlings--a blinded comparison of commercially available vaccines. Vaccine 2013; 31:5216-22. [PMID: 24021309 DOI: 10.1016/j.vaccine.2013.08.083] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 08/19/2013] [Accepted: 08/27/2013] [Indexed: 11/29/2022]
Abstract
Previous studies in experimental ponies using interferon gamma (IFN-γ) as a marker for cell mediated immune (CMI) response demonstrated an increase in IFN-γ gene expression following vaccination with an ISCOM subunit, a canarypox recombinant and more recently, an inactivated whole virus vaccine. The objective of this study was to carry out an independent comparison of both humoral antibody and CMI responses elicited following vaccination with all these vaccine presentation systems. Antibody response of 44 Thoroughbred weanlings was monitored for three weeks following the second dose of primary vaccination (V2) by single radial haemolysis (SRH). The pattern of antibody response was similar for all vaccines. The antibody response of horses vaccinated with the inactivated whole virus vaccine (Duvaxyn IE-T Plus) was superior to that of the horses vaccinated with the ISCOM-matrix subunit (Equilis Prequenza Te) and canarypox recombinant (ProteqFlu-Te) vaccine. In this study 39% of weanlings failed to seroconvert following their first dose of primary vaccination (V1). Poor response to vaccination (H3N8) was observed among weanlings vaccinated with Equilis Prequenza Te and ProteqFlu-Te but not among those vaccinated with Duvaxyn IE-T Plus. PAXgene bloods were collected on days 0, 2, 7 and 14 following V1. Gene expression levels of IFN-γ, IL-1β (proinflammatory cytokine) and IL-4 (B cell stimulating cytokine) were measured using RT-PCR. Mean gene expression levels of IL-1β and IL-4 peaked on day 14 post vaccination. The increase in IL-4 gene expression by horses vaccinated with Equilis Prequenza Te was significantly greater to those vaccinated with the other two products. Vaccination with all three vaccines resulted in a significant increase in IFN-γ gene expression which peaked at 7 days post V1. Overall, there was no significant difference in IFN-γ gene expression by the horses vaccinated with the whole inactivated, the subunit and the canarypox recombinant vaccines included in this study.
Collapse
Affiliation(s)
- Sarah Gildea
- Virology Unit, The Irish Equine Centre, Johnstown, Naas, Co., Kildare, Ireland
| | | | | | | |
Collapse
|
23
|
MacLeod MKL, David A, Jin N, Noges L, Wang J, Kappler JW, Marrack P. Influenza nucleoprotein delivered with aluminium salts protects mice from an influenza A virus that expresses an altered nucleoprotein sequence. PLoS One 2013; 8:e61775. [PMID: 23613928 PMCID: PMC3629017 DOI: 10.1371/journal.pone.0061775] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 03/13/2013] [Indexed: 11/19/2022] Open
Abstract
Influenza virus poses a difficult challenge for protective immunity. This virus is adept at altering its surface proteins, the proteins that are the targets of neutralizing antibody. Consequently, each year a new vaccine must be developed to combat the current recirculating strains. A universal influenza vaccine that primes specific memory cells that recognise conserved parts of the virus could prove to be effective against both annual influenza variants and newly emergent potentially pandemic strains. Such a vaccine will have to contain a safe and effective adjuvant that can be used in individuals of all ages. We examine protection from viral challenge in mice vaccinated with the nucleoprotein from the PR8 strain of influenza A, a protein that is highly conserved across viral subtypes. Vaccination with nucleoprotein delivered with a universally used and safe adjuvant, composed of insoluble aluminium salts, provides protection against viruses that either express the same or an altered version of nucleoprotein. This protection correlated with the presence of nucleoprotein specific CD8 T cells in the lungs of infected animals at early time points after infection. In contrast, immunization with NP delivered with alum and the detoxified LPS adjuvant, monophosphoryl lipid A, provided some protection to the homologous viral strain but no protection against infection by influenza expressing a variant nucleoprotein. Together, these data point towards a vaccine solution for all influenza A subtypes.
Collapse
Affiliation(s)
- Megan K. L. MacLeod
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
- * E-mail: (PM); (MKLM)
| | - Alexandria David
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
| | - Niyun Jin
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
| | - Laura Noges
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
| | - Jieru Wang
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - John W. Kappler
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
- Program in Biomolecular Structure, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
- Department of Medicine, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
| | - Philippa Marrack
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, Colorado, United States of America
- Program in Biomolecular Structure, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, School of Medicine, Aurora, Colorado, United States of America
- * E-mail: (PM); (MKLM)
| |
Collapse
|
24
|
Sage LK, Fox JM, Tompkins SM, Tripp RA. Subsisting H1N1 influenza memory responses are insufficient to protect from pandemic H1N1 influenza challenge in C57BL/6 mice. J Gen Virol 2013; 94:1701-1711. [PMID: 23580424 DOI: 10.1099/vir.0.049494-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The 2009 swine-origin pandemic H1N1 (pH1N1) influenza virus transmitted and caused disease in many individuals immune to pre-2009 H1N1 influenza virus. Whilst extensive studies on antibody-mediated pH1N1 cross-reactivity have been described, few studies have focused on influenza-specific memory T-cells. To address this, the immune response in pre-2009 H1N1 influenza-immune mice was evaluated after pH1N1 challenge and disease pathogenesis was determined. The results show that despite homology shared between pre-2009 H1N1 and pH1N1 strains, the effector memory T-cell response to pre-2009 H1N1 was generally ineffective, a finding that correlated with lung virus persistence. Additionally, pH1N1 challenge generated T-cells reactive to new pH1N1 epitopes. These studies highlight the importance of vaccinating against immunodominant T-cell epitopes to provide for a more effective strategy to control influenza virus through heterosubtypic immunity.
Collapse
Affiliation(s)
- Leo K Sage
- University of Georgia, College of Veterinary Medicine, Department of Infectious Diseases, Athens, GA, USA
| | - Julie M Fox
- University of Georgia, College of Veterinary Medicine, Department of Infectious Diseases, Athens, GA, USA
| | - Stephen M Tompkins
- University of Georgia, College of Veterinary Medicine, Department of Infectious Diseases, Athens, GA, USA
| | - Ralph A Tripp
- University of Georgia, College of Veterinary Medicine, Department of Infectious Diseases, Athens, GA, USA
| |
Collapse
|
25
|
Laidlaw BJ, Decman V, Ali MAA, Abt MC, Wolf AI, Monticelli LA, Mozdzanowska K, Angelosanto JM, Artis D, Erikson J, Wherry EJ. Cooperativity between CD8+ T cells, non-neutralizing antibodies, and alveolar macrophages is important for heterosubtypic influenza virus immunity. PLoS Pathog 2013; 9:e1003207. [PMID: 23516357 PMCID: PMC3597515 DOI: 10.1371/journal.ppat.1003207] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 01/10/2013] [Indexed: 11/29/2022] Open
Abstract
Seasonal epidemics of influenza virus result in ∼36,000 deaths annually in the United States. Current vaccines against influenza virus elicit an antibody response specific for the envelope glycoproteins. However, high mutation rates result in the emergence of new viral serotypes, which elude neutralization by preexisting antibodies. T lymphocytes have been reported to be capable of mediating heterosubtypic protection through recognition of internal, more conserved, influenza virus proteins. Here, we demonstrate using a recombinant influenza virus expressing the LCMV GP33-41 epitope that influenza virus-specific CD8+ T cells and virus-specific non-neutralizing antibodies each are relatively ineffective at conferring heterosubtypic protective immunity alone. However, when combined virus-specific CD8 T cells and non-neutralizing antibodies cooperatively elicit robust protective immunity. This synergistic improvement in protective immunity is dependent, at least in part, on alveolar macrophages and/or other lung phagocytes. Overall, our studies suggest that an influenza vaccine capable of eliciting both CD8+ T cells and antibodies specific for highly conserved influenza proteins may be able to provide heterosubtypic protection in humans, and act as the basis for a potential “universal” vaccine. Influenza virus continues to pose a significant risk to global health and is responsible for thousands of deaths each year in the United States. This threat is largely due to the ability of the influenza virus to undergo rapid changes, allowing it to escape from immune responses elicited by previous infections or vaccinations. Certain internal determinants of the influenza virus are largely conserved across different viral strains and represent attractive targets for potential “universal” influenza vaccines. Here, we demonstrated that cross-subtype protection against the influenza virus could be obtained through simultaneous priming of multiple arms of the immune response against conserved elements of the influenza virus. These results suggest a novel strategy that could potentially form a primary component of a universal influenza vaccine capable of providing long-lasting protection.
Collapse
Affiliation(s)
- Brian J. Laidlaw
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Vilma Decman
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Mohammed-Alkhatim A. Ali
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michael C. Abt
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Amaya I. Wolf
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Laurel A. Monticelli
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | | | - Jill M. Angelosanto
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - David Artis
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jan Erikson
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - E. John Wherry
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
26
|
Sato K, Iwai A, Nakayama Y, Morimoto J, Takada A, Maruyama M, Kida H, Uede T, Miyazaki T. Osteopontin is critical to determine symptom severity of influenza through the regulation of NK cell population. Biochem Biophys Res Commun 2012; 417:274-9. [DOI: 10.1016/j.bbrc.2011.11.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 11/18/2011] [Indexed: 01/08/2023]
|
27
|
Kappes MA, Sandbulte MR, Platt R, Wang C, Lager KM, Henningson JN, Lorusso A, Vincent AL, Loving CL, Roth JA, Kehrli ME. Vaccination with NS1-truncated H3N2 swine influenza virus primes T cells and confers cross-protection against an H1N1 heterosubtypic challenge in pigs. Vaccine 2011; 30:280-8. [PMID: 22067263 DOI: 10.1016/j.vaccine.2011.10.098] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 10/21/2011] [Accepted: 10/30/2011] [Indexed: 10/15/2022]
Abstract
The diversity of contemporary swine influenza virus (SIV) strains impedes effective immunization of swine herds. Mucosally delivered, attenuated virus vaccines are one approach with potential to provide broad cross-protection. Reverse genetics-derived H3N2 SIV virus with truncated NS1 (NS1Δ126 TX98) is attenuated and immunogenic when delivered intranasally in young pigs. We analyzed T-cell priming and cross-protective efficacy in weanling piglets after intranasal inoculation with NS1Δ126 TX98 versus wild type TX98. In vivo replication of the truncation mutant was minimal compared to the wild type virus. T-cell responses were greater in magnitude in pigs infected with the wild type virus in in vitro restimulation assays. According to the expression of activation marker CD25, peripheral T cell recall responses in NS1Δ126 TX98 infected pigs were minimal. However, intracellular IFN-γ data indicate that the attenuated virus induced virus-specific CD4(+)CD8(-), CD4(+)CD8(+), CD4(-)CD8(+), and γδ T cells within 28 days. The IFN-γ response appeared to contract, as responses were reduced at later time points prior to challenge. CD4(+)CD8(+) cells isolated 5 days after heterosubtypic H1N1 challenge (day 70 overall) showed an elevated CD25 response to virus restimulation. Pigs previously infected with wild type TX98 were protected from replication of the H1N1 challenge virus. Vaccination with NS1Δ126 TX98 was associated with significantly lower levels of Th1-associated cytokines in infected lungs but provided partial cross-protection against the H1N1 challenge. These results demonstrate that NS1Δ SIV vaccines can elicit cell-mediated cross-protection against antigenically divergent strains.
Collapse
Affiliation(s)
- Matthew A Kappes
- Virus and Prion Diseases Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, PO Box 70, Ames, IA 50010, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Induction of virus-specific cytotoxic T lymphocytes as a basis for the development of broadly protective influenza vaccines. J Biomed Biotechnol 2011; 2011:939860. [PMID: 22007149 PMCID: PMC3189652 DOI: 10.1155/2011/939860] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/01/2011] [Accepted: 08/02/2011] [Indexed: 11/18/2022] Open
Abstract
There is considerable interest in the development of broadly protective influenza vaccines because of the continuous emergence of antigenic drift variants of seasonal influenza viruses and the threat posed by the emergence of antigenically distinct pandemic influenza viruses. It has been recognized more than three decades ago that influenza A virus-specific cytotoxic T lymphocytes recognize epitopes located in the relatively conserved proteins like the nucleoprotein and that they cross-react with various subtypes of influenza A viruses. This implies that these CD8+ T lymphocytes may contribute to protective heterosubtypic immunity induced by antecedent influenza A virus infections. In the present paper, we review the evidence for the role of virus-specific CD8+ T lymphocytes in protective immunity against influenza virus infections and discuss vaccination strategies that aim at the induction of cross-reactive virus-specific T-cell responses.
Collapse
|
29
|
Ichihashi T, Yoshida R, Sugimoto C, Takada A, Kajino K. Cross-protective peptide vaccine against influenza A viruses developed in HLA-A*2402 human immunity model. PLoS One 2011; 6:e24626. [PMID: 21949735 PMCID: PMC3176274 DOI: 10.1371/journal.pone.0024626] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 08/15/2011] [Indexed: 11/25/2022] Open
Abstract
Background The virus-specific cytotoxic T lymphocyte (CTL) induction is an important target for the development of a broadly protective human influenza vaccine, since most CTL epitopes are found on internal viral proteins and relatively conserved. In this study, the possibility of developing a strain/subtype-independent human influenza vaccine was explored by taking a bioinformatics approach to establish an immunogenic HLA-A24 restricted CTL epitope screening system in HLA-transgenic mice. Methodology/Principal Findings HLA-A24 restricted CTL epitope peptides derived from internal proteins of the H5N1 highly pathogenic avian influenza A virus were predicted by CTL epitope peptide prediction programs. Of 35 predicted peptides, six peptides exhibited remarkable cytotoxic activity in vivo. More than half of the mice which were subcutaneously vaccinated with the three most immunogenic and highly conserved epitopes among three different influenza A virus subtypes (H1N1, H3N2 and H5N1) survived lethal influenza virus challenge during both effector and memory CTL phases. Furthermore, mice that were intranasally vaccinated with these peptides remained free of clinical signs after lethal virus challenge during the effector phase. Conclusions/Significance This CTL epitope peptide selection system can be used as an effective tool for the development of a cross-protective human influenza vaccine. Furthermore this vaccine strategy can be applicable to the development of all intracellular pathogens vaccines to induce epitope-specific CTL that effectively eliminate infected cells.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cross Protection/immunology
- Epitopes/immunology
- HLA-A24 Antigen/genetics
- HLA-A24 Antigen/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H1N1 Subtype/enzymology
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza A Virus, H3N2 Subtype/enzymology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/pathogenicity
- Influenza A Virus, H5N1 Subtype/enzymology
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza A virus/enzymology
- Influenza A virus/immunology
- Influenza A virus/pathogenicity
- Influenza Vaccines/immunology
- Lung/virology
- Mice
- Mice, Transgenic
- Models, Animal
- Neuraminidase/immunology
- Reproducibility of Results
- T-Lymphocytes, Cytotoxic/immunology
- Time Factors
- Vaccination
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Toru Ichihashi
- Department of Collaboration and Education, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Reiko Yoshida
- Department of Global Epidemiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Chihiro Sugimoto
- Department of Collaboration and Education, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Ayato Takada
- Department of Global Epidemiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Kiichi Kajino
- Department of Collaboration and Education, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
- * E-mail:
| |
Collapse
|
30
|
Staneková Z, Varečková E. Conserved epitopes of influenza A virus inducing protective immunity and their prospects for universal vaccine development. Virol J 2010; 7:351. [PMID: 21118546 PMCID: PMC3009981 DOI: 10.1186/1743-422x-7-351] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 11/30/2010] [Indexed: 01/08/2023] Open
Abstract
Influenza A viruses belong to the best studied viruses, however no effective prevention against influenza infection has been developed. The emerging of still new escape variants of influenza A viruses causing epidemics and periodic worldwide pandemics represents a threat for human population. Therefore, current, hot task of influenza virus research is to look for a way how to get us closer to a universal vaccine. Combination of chosen conserved antigens inducing cross-protective antibody response with epitopes activating also cross-protective cytotoxic T-cells would offer an attractive strategy for improving protection against drift variants of seasonal influenza viruses and reduces the impact of future pandemic strains. Antigenically conserved fusion-active subunit of hemagglutinin (HA2 gp) and ectodomain of matrix protein 2 (eM2) are promising candidates for preparation of broadly protective HA2- or eM2-based vaccine that may aid in pandemic preparedness. Overall protective effect could be achieved by contribution of epitopes recognized by cytotoxic T-lymphocytes (CTL) that have been studied extensively to reach much broader control of influenza infection. In this review we present the state-of-art in this field. We describe known adaptive immune mechanisms mediated by influenza specific B- and T-cells involved in the anti-influenza immune defense together with the contribution of innate immunity. We discuss the mechanisms of neutralization of influenza infection mediated by antibodies, the role of CTL in viral elimination and new approaches to develop epitope based vaccine inducing cross-protective influenza virus-specific immune response.
Collapse
Affiliation(s)
- Zuzana Staneková
- Institute of Virology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovak Republic.
| | | |
Collapse
|
31
|
McKee AS, MacLeod MKL, Kappler JW, Marrack P. Immune mechanisms of protection: can adjuvants rise to the challenge? BMC Biol 2010; 8:37. [PMID: 20385031 PMCID: PMC2864095 DOI: 10.1186/1741-7007-8-37] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 04/12/2010] [Indexed: 12/18/2022] Open
Abstract
For many diseases vaccines are lacking or only partly effective. Research on protective immunity and adjuvants that generate vigorous immune responses may help generate effective vaccines against such pathogens.
Collapse
Affiliation(s)
- Amy S McKee
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Health, Denver, CO 80206, USA
| | | | | | | |
Collapse
|
32
|
Cell-mediated immune response and cross-protective efficacy of binary ethylenimine-inactivated bluetongue virus serotype-1 vaccine in sheep. Vaccine 2010; 28:2522-31. [DOI: 10.1016/j.vaccine.2010.01.039] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2009] [Revised: 01/14/2010] [Accepted: 01/16/2010] [Indexed: 11/22/2022]
|
33
|
Targets for the induction of protective immunity against influenza a viruses. Viruses 2010; 2:166-188. [PMID: 21994606 PMCID: PMC3185556 DOI: 10.3390/v2010166] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 01/04/2010] [Accepted: 01/13/2010] [Indexed: 01/09/2023] Open
Abstract
The current pandemic caused by the new influenza A(H1N1) virus of swine origin and the current pandemic threat caused by the highly pathogenic avian influenza A viruses of the H5N1 subtype have renewed the interest in the development of vaccines that can induce broad protective immunity. Preferably, vaccines not only provide protection against the homologous strains, but also against heterologous strains, even of another subtype. Here we describe viral targets and the arms of the immune response involved in protection against influenza virus infections such as antibodies directed against the hemagglutinin, neuraminidase and the M2 protein and cellular immune responses directed against the internal viral proteins.
Collapse
|
34
|
Sun Y, Liu J, Yang M, Gao F, Zhou J, Kitamura Y, Gao B, Tien P, Shu Y, Iwamoto A, Chen Z, Gao GF. Identification and structural definition of H5-specific CTL epitopes restricted by HLA-A*0201 derived from the H5N1 subtype of influenza A viruses. J Gen Virol 2009; 91:919-30. [PMID: 19955560 DOI: 10.1099/vir.0.016766-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The haemagglutinin (HA) glycoprotein of influenza A virus is a major antigen that initiates humoral immunity against infection; however, the cellular immune response against HA is poorly understood. Furthermore, HA-derived cytotoxic T-lymphocyte (CTL) epitopes are relatively rare in comparison to other internal gene products. Here, CTL epitopes of the HA serotype H5 protein were screened. By using in silico prediction, in vitro refolding and a T2 cell-binding assay, followed by immunization of HLA-A2.1/K(b) transgenic mice, an HLA-A*0201-restricted decameric epitope, RI-10 (H5 HA205-214, RLYQNPTTYI), was shown to elicit a robust CTL epitope-specific response. In addition, RI-10 and its variant, KI-10 (KLYQNPTTYI), were also demonstrated to be able to induce a higher CTL epitope-specific response than the influenza A virus dominant CTL epitope GL-9 (GILGFVFTL) in peripheral blood mononuclear cells of HLA-A*0201-positive patients who had recovered from H5N1 virus infection. Furthermore, the crystal structures of RI-10-HLA-A*0201 and KI-10-HLA-A*0201 complexes were determined at 2.3 and 2.2 A resolution, respectively, showing typical HLA-A*0201-restricted epitopes. The conformations of RI-10 and KI-10 in the antigen-presenting grooves in crystal structures of the two complexes show significant differences, despite their nearly identical sequences. These results provide implications for the discovery of diagnostic markers and the design of novel influenza vaccines.
Collapse
Affiliation(s)
- Yeping Sun
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kreijtz JHCM, Bodewes R, van den Brand JMA, de Mutsert G, Baas C, van Amerongen G, Fouchier RAM, Osterhaus ADME, Rimmelzwaan GF. Infection of mice with a human influenza A/H3N2 virus induces protective immunity against lethal infection with influenza A/H5N1 virus. Vaccine 2009; 27:4983-9. [PMID: 19538996 DOI: 10.1016/j.vaccine.2009.05.079] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 05/07/2009] [Accepted: 05/28/2009] [Indexed: 10/20/2022]
Abstract
The transmission of highly pathogenic avian influenza (HPAI) A viruses of the H5N1 subtype from poultry to man and the high case fatality rate fuels the fear for a pandemic outbreak caused by these viruses. However, prior infections with seasonal influenza A/H1N1 and A/H3N2 viruses induce heterosubtypic immunity that could afford a certain degree of protection against infection with the HPAI A/H5N1 viruses, which are distantly related to the human influenza A viruses. To assess the protective efficacy of such heterosubtypic immunity mice were infected with human influenza virus A/Hong Kong/2/68 (H3N2) 4 weeks prior to a lethal infection with HPAI virus A/Indonesia/5/05 (H5N1). Prior infection with influenza virus A/Hong Kong/2/68 reduced clinical signs, body weight loss, mortality and virus replication in the lungs as compared to naive mice infected with HPAI virus A/Indonesia/5/05. Priming by infection with respiratory syncytial virus, a non-related virus did not have a beneficial effect on the outcome of A/H5N1 infections, indicating that adaptive immune responses were responsible for the protective effect. In mice primed by infection with influenza A/H3N2 virus cytotoxic T lymphocytes (CTL) specific for NP(366-374) epitope ASNENMDAM and PA(224-232) SCLENFRAYV were observed. A small proportion of these CTL was cross-reactive with the peptide variant derived from the influenza A/H5N1 virus (ASNENMEVM and SSLENFRAYV respectively) and upon challenge infection with the influenza A/H5N1 virus cross-reactive CTL were selectively expanded. These CTL, in addition to those directed to conserved epitopes, shared by the influenza A/H3N2 and A/H5N1 viruses, most likely contributed to accelerated clearance of the influenza A/H5N1 virus infection. Although also other arms of the adaptive immune response may contribute to heterosubtypic immunity, the induction of virus-specific CTL may be an attractive target for development of broad protective vaccines. Furthermore the existence of pre-existing heterosubtypic immunity may dampen the impact a future influenza pandemic may have.
Collapse
Affiliation(s)
- J H C M Kreijtz
- Department of Virology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Prospects for an influenza vaccine that induces cross-protective cytotoxic T lymphocytes. Immunol Cell Biol 2009; 87:300-8. [PMID: 19308073 DOI: 10.1038/icb.2009.16] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Our approach to vaccination against influenza is unique. For no other pathogen do we construct and produce a new vaccine every year in the face of uncertainty about the strains that will be circulating when it is used. The huge global cooperative effort that underpins this process reflects our awareness of the need to control this major pathogen. Moreover, the threat of devastation by a pandemic due to a newly emerging viral subtype has triggered an intense effort to improve and accelerate the production of vaccines for use if a pandemic arises. However, type A influenza viruses responsible for seasonal epidemics and those with the potential to cause a pandemic share amino acid sequences that form the targets of cytotoxic T lymphocytes (CTL). CTL activated by currently circulating viruses, therefore, offer a possible means to limit the impact of infection with future variant seasonal strains and even new subtypes. This review examines how cross-protective CTL can be exploited to improve influenza vaccination and issues that need to be considered when attempting to induce this type of immunity. We discuss the role of CTL responses in viral control and review the current knowledge relating to specificity and longevity of memory CD8(+) T cells, how vaccine antigen can be loaded into antigen-presenting cells to prime these responses and factors influencing the class of response induced. Application of these principles to the next generation of influenza vaccines should lead to much greater control of infection.
Collapse
|
37
|
Development of stable influenza vaccine powder formulations: challenges and possibilities. Pharm Res 2008; 25:1256-73. [PMID: 18338241 PMCID: PMC2346510 DOI: 10.1007/s11095-008-9559-6] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2007] [Accepted: 02/13/2008] [Indexed: 01/24/2023]
Abstract
Influenza vaccination represents the cornerstone of influenza prevention. However, today all influenza vaccines are formulated as liquids that are unstable at ambient temperatures and have to be stored and distributed under refrigeration. In order to stabilize influenza vaccines, they can be brought into the dry state using suitable excipients, stabilizers and drying processes. The resulting stable influenza vaccine powder is independent of cold-chain facilities. This can be attractive for the integration of the vaccine logistics with general drug distribution in Western as well as developing countries. In addition, a stockpile of stable vaccine formulations of potential vaccines against pandemic viruses can provide an immediate availability and simple distribution of vaccine in a pandemic outbreak. Finally, in the development of new needle-free dosage forms, dry and stable influenza vaccine powder formulations can facilitate new or improved targeting strategies for the vaccine compound. This review represents the current status of dry stable inactivated influenza vaccine development. Attention is given to the different influenza vaccine types (i.e. whole inactivated virus, split, subunit or virosomal vaccine), the rationale and need for stabilized influenza vaccines, drying methods by which influenza vaccines can be stabilized (i.e. lyophilization, spray drying, spray-freeze drying, vacuum drying or supercritical fluid drying), the current status of dry influenza vaccine development and the challenges for ultimate market introduction of a stable and effective dry-powder influenza vaccine.
Collapse
|
38
|
Rimmelzwaan GF, Fouchier RAM, Osterhaus ADME. Influenza virus-specific cytotoxic T lymphocytes: a correlate of protection and a basis for vaccine development. Curr Opin Biotechnol 2008; 18:529-36. [PMID: 18083548 DOI: 10.1016/j.copbio.2007.11.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 10/30/2007] [Accepted: 11/12/2007] [Indexed: 10/22/2022]
Abstract
Since influenza A viruses of the H5N1 subtype continue to circulate in wild and domestic birds and cause an ever increasing number of human cases, it is feared that H5N1 viruses may cause the next influenza pandemic. Therefore, there is considerable interest in the development of vaccines that confer protection against infections with these viruses or ideally, protection against influenza viruses of different subtypes. For the development of broad-protective vaccines the induction of virus-specific cytotoxic T lymphocytes (CTL) may be an important target, since it has been demonstrated that CTL contribute to protective immunity and are largely directed to epitopes shared by influenza viruses of various subtypes. In the present paper, the possibility to develop (cross-reactive) CTL-inducing vaccines is discussed.
Collapse
Affiliation(s)
- Guus F Rimmelzwaan
- Erasmus Medical Center, Department of Virology, Rotterdam, The Netherlands
| | | | | |
Collapse
|
39
|
Kreijtz JHCM, Bodewes R, van Amerongen G, Kuiken T, Fouchier RAM, Osterhaus ADME, Rimmelzwaan GF. Primary influenza A virus infection induces cross-protective immunity against a lethal infection with a heterosubtypic virus strain in mice. Vaccine 2006; 25:612-20. [PMID: 17005299 DOI: 10.1016/j.vaccine.2006.08.036] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 08/18/2006] [Accepted: 08/24/2006] [Indexed: 10/24/2022]
Abstract
In order to assess the level of protection against a lethal influenza virus infection provided by a primary infection with a virus strain of another subtype, C57BL/6 mice were infected with the sublethal influenza virus X-31 (H3N2) and subsequently challenged with the lethal strain A/PR/8/34 (H1N1). The outcome of the challenge infection was compared with that in mice that did not experience an infection with influenza virus X-31 prior to the challenge infection. The X-31 experienced mice cleared the infection with influenza virus A/PR/8/34 in an accelerated fashion, displayed less clinical signs and a reduction of lesions in the lungs resulting in improved survival rates of these mice compared to the naive mice. The improved outcome of the challenge infection with influenza virus A/PR/8/34 in the X-31 experienced mice correlated with priming for anamnestic virus-specific CD8(+) cytotoxic T lymphocyte (CTL) responses as was demonstrated by the detection of CTL specific for the H-2D(b) restricted NP(366-374) epitope that was shared by the influenza viruses X-31 and A/PR/8/34. Thus previous exposure to influenza A viruses affords partial protection against infection in the absence of virus-neutralizing antibodies specific for the hemagglutinin and the neuraminidase. The implications of these observations are discussed in the light of the current pandemic threat and development of vaccines that aim at the induction of virus-specific CTL.
Collapse
Affiliation(s)
- J H C M Kreijtz
- Department of Virology, Erasmus Medical Centre, P.O. Box 1738, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
40
|
Vignuzzi M, Gerbaud S, van der Werf S, Escriou N. Naked RNA immunization with replicons derived from poliovirus and Semliki Forest virus genomes for the generation of a cytotoxic T cell response against the influenza A virus nucleoprotein. J Gen Virol 2001; 82:1737-1747. [PMID: 11413386 DOI: 10.1099/0022-1317-82-7-1737] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The potential of RNA-based vaccines was evaluated for the generation of a protective immune response in the mouse model of influenza type A virus infection using the internal nucleoprotein (NP) as antigen. This antigen is of particular interest, since it has the potential to elicit protective cytotoxic T lymphocytes (CTL) against heterologous strains of influenza A virus. In view of the short half-life of RNA, self-replicating RNAs or replicons of the positive-stranded genomes of Semliki Forest virus (SFV) and poliovirus were engineered to synthesize the influenza A virus NP in place of their structural proteins. NP expression was demonstrated by immunoprecipitation after transfection of cells with RNA from the SFV (rSFV-NP) and poliovirus (rDeltaP1-E-NP) genome-derived replicons transcribed in vitro. C57BL/6 mice were injected intramuscularly with these synthetic RNAs in naked form. Both replicons, rSFV-NP and rDeltaP1-E-NP, induced antibodies against the influenza virus NP, but only mice immunized with the rSFV-NP replicon developed a CTL response against the immunodominant H-2D(b) epitope NP366. Finally, the protective potential of the CTL response induced by immunization of mice with rSFV-NP RNA was demonstrated by the reduction of virus load in the lungs after challenge infection with mouse-adapted influenza A/PR/8/34 virus and was comparable to the protective potential of the response induced by plasmid DNA immunization. These results demonstrate that naked RNA immunization with self-replicating molecules can effectively induce both humoral and cellular immune responses and constitutes an alternative strategy to DNA immunization.
Collapse
Affiliation(s)
- Marco Vignuzzi
- Unité de Génétique Moléculaire des Virus Respiratoires, URA 1966 CNRS, Institut Pasteur, 25 rue du Dr Roux, F-75724 Paris cedex 15, France1
| | - Sylvie Gerbaud
- Unité de Génétique Moléculaire des Virus Respiratoires, URA 1966 CNRS, Institut Pasteur, 25 rue du Dr Roux, F-75724 Paris cedex 15, France1
| | - Sylvie van der Werf
- Unité de Génétique Moléculaire des Virus Respiratoires, URA 1966 CNRS, Institut Pasteur, 25 rue du Dr Roux, F-75724 Paris cedex 15, France1
| | - Nicolas Escriou
- Unité de Génétique Moléculaire des Virus Respiratoires, URA 1966 CNRS, Institut Pasteur, 25 rue du Dr Roux, F-75724 Paris cedex 15, France1
| |
Collapse
|
41
|
Chen Z, Berkower I, Ching WM, Wang RY, Alter HJ, Shih JW. Identification of a murine CD4+ T-lymphocyte response site in hepatitis C virus core protein. Mol Immunol 1996; 33:703-9. [PMID: 8760282 DOI: 10.1016/0161-5890(96)00010-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The T cell response to a recombinant HCV truncated core protein (cp1-10) was measured in a proliferation assay. Based on a 10-fold greater response to this truncated core protein than to its shorter form (cp1-8), a predominant epitope was mapped to the carboxyl quarter of this sequence. This epitope was further mapped to a synthetic peptide corresponding to amino acids 121-140 of the core protein. The peptide was antigenic for T cells of all three H-2 types tested, H-2 r, b and d, and the proliferating T cells were CD4+. Besides inducing specific proliferation in vitro, peptide aa121-140 can prime helper T cells in vivo. When boosted with core protein, mice primed with peptide produced 64-fold higher antibody titer than without priming in 1 week. The identification of a broadly immunogenic T cell helper epitope on core protein may be important for vaccine design against HCV.
Collapse
Affiliation(s)
- Z Chen
- Department of Transfusion Medicine, Warren G. Magnuson Clinical Center, National Institutes of Health, Bethesda, MD 20892-1184, USA
| | | | | | | | | | | |
Collapse
|
42
|
Kuroda K, Nakajima M, Minakata H, Hosaka Y. Identification of Kk-restricted T-cell epitope within influenza virus nucleoprotein. Virus Res 1995; 38:43-53. [PMID: 8546009 DOI: 10.1016/0168-1702(95)00042-o] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In order to determine the epitope structure in peptide NP50-63, which has been reported to be the only Kk-restricted T-cell antigen within the influenza virus (A/PR/8/34) nucleoprotein, a series of 13 peptides truncated from C- and N-termini of NP50-63 were synthesized and their sensitizing activities against Kk-restricted nucleoprotein-specific cytotoxic T lymphocytes (CTLs) were examined. One of the 13 peptides, NP50-57, sensitized L929 cells at the nM level, which was 100-1000 times lower in concentration than that at which the other peptides sensitized these cells. The presence of NP50-57 in A/PR/8/34-infected L929 cells was also investigated. Acid extracts of virus-infected cells were separated on a reverse-phase HPLC column and then anion-exchange column. By both separations, only one peak of sensitizing activity against nucleoprotein-specific CTLs was observed. The position of the peak coincided with that of the elution of NP50-57. These results strongly suggest that NP50-57 is the natural epitope in the antigenic structure, NP50-63.
Collapse
Affiliation(s)
- K Kuroda
- Department of Virology and Immunology, Osaka University of Pharmaceutical Sciences, Matsubara, Japan
| | | | | | | |
Collapse
|
43
|
Iinuma H, Nerome K, Yoshioka Y, Okinaga K. Characteristics of cytotoxic T lymphocytes directed to influenza virus haemagglutinin elicited by immunization with muramyldipeptide-influenza liposome vaccine. Scand J Immunol 1995; 41:1-10. [PMID: 7824883 DOI: 10.1111/j.1365-3083.1995.tb03526.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We examined the characterization of the antiviral T lymphocytes elicited by immunization with a novel liposome vaccine (MDP-virosome) constructed with synthetic muramyldipeptide; [6-0-(2-tetradecylhexadecanoyl)-N-acetylmuramyl-L-alanyl-D-isoglutamine] , cholesterol, influenza virus haemagglutinin and neuraminidase. The haemagglutinin glycoprotein first appeared to induce a significant subtype-specific cytotoxic activity through its arrangement on the inner and outer surfaces of the MDP-virosome. Splenocytes of BALB/c mice immunized with the virosome vaccine containing H3 haemagglutinin and N2 neuraminidase from human Hong Kong virus markedly lysed H3N2 virus-infected target cells, but not those infected with virus possessing a different subtype such as H1N1 surface antigens. Exposure of these splenic lymphocytes to virus antigen in vitro further enhanced their cytotoxic activity. The cytotoxic lymphocytes generated by the MDP-virosome vaccine expressed Thy 1 and CD4 antigens on their cell surface, and these activities were restricted by class II histocompatibility gene products. The marked reduction of pulmonary virus titres in infected mice caused by transferred immune spleen cells suggested that the MDP-virosome vaccination is able to protect against influenza virus infection through enhanced cellular immune responses.
Collapse
Affiliation(s)
- H Iinuma
- Second Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
44
|
Abstract
Twenty years ago, the terms 'altered self' and 'H-2 restriction', later modified to 'MHC-restriction', were coined to describe the finding by Peter Doherty and Rolf Zinkernagel that murine cytotoxic T cells (CTL) would lyse virus-infected target cells only if effector and target cells were H-2 compatible. This short review recalls those heady days and briefly recounts some of the later findings in three aspects of particular interest raised by the original finding: the nature of the T cell receptor, the composition and structure of the ligand on the target cell recognized by the TCR and the importance of CTL in the control and clearance of infections in general.
Collapse
Affiliation(s)
- G Ada
- Division of Cell Biology, John Curtin School of Medical Research, Australian National University, Canberra
| |
Collapse
|
45
|
Mbawuike IN, Dillion SB, Demuth SG, Jones CS, Cate TR, Couch RB. Influenza A subtype cross-protection after immunization of outbred mice with a purified chimeric NS1/HA2 influenza virus protein. Vaccine 1994; 12:1340-8. [PMID: 7856302 DOI: 10.1016/s0264-410x(94)80063-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Influenza A/PR/8/34-derived chimeric (D) protein (SK&F 106160) composed of the first 81 amino acids (aa) of NS1 fused to the conserved 157 C-terminal aa of HA2 (NS1 1-81-HA2 65-222) was previously shown to induce H-2d-restricted protective cytotoxic T-lymphocyte (CTL) immunity in inbred mice. However, D protein, like other small peptides, exhibited haplotype dependence and was not immunogenic in H-2b and H-2K mice. A potential use of this antigen in humans and the role of T cells in any protection were evaluated in outbred Swiss and inbred CBF6F1 (H-2d/b) mice. Mice immunized with D protein and challenged by small-particle aerosol with a lethal dose of influenza virus were significantly protected against mortality from influenza A/H1N1 and A/H2N2 (p < 0.05-< 0.0000001), but not from A/H3N2 and influenza B viruses when compared with control mice. D protein did not induce serum virus-neutralizing antibody but caused virus to be cleared faster in immunized mice. Protection was long-lasting. In vivo depletion of either Lyt2 (CD8+) or L3T4 (CD4+) T cells with monoclonal antibodies led to abrogation of in vitro-generated CTL activity in CF6F1 mice and significant reduction in the protective efficacy of D protein against virus challenge in both Swiss and CF6F1 mice. These results suggest that protection was mediated by CD8+ and/or CD4+ cells and not antibody. Thus D protein, via a conserved sequence on the HA2 polypeptide, has the potential to induce partially cross-reactive CTL that may protect against influenza virus disease in humans.
Collapse
Affiliation(s)
- I N Mbawuike
- Department of Microbiology and Immunology, Baylor College of Medicine, Houston, TX 77030-3498
| | | | | | | | | | | |
Collapse
|
46
|
Mbawuike IN, Wyde PR. Induction of CD8+ cytotoxic T cells by immunization with killed influenza virus and effect of cholera toxin B subunit. Vaccine 1993; 11:1205-13. [PMID: 7903015 DOI: 10.1016/0264-410x(93)90044-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The MHC class I cytotoxic T-lymphocyte (CTL) response in mice given formalin-inactivated influenza whole-virus vaccine (WVV) with or without cholera toxin B subunit (CTB) was studied. Intraperitoneal injection of Balb/c (H-2d) mice with high doses of A/Taiwan/1/86 (H1N1) WVV stimulated influenza A virus-specific CTL response in a dose-dependent manner. A dose of 4.4 or 44 micrograms induced CTL response equal to or greater than live influenza virus infection. Coadministration of vaccine with 5 or 25 micrograms of CTB resulted in a higher level of CTL than with vaccine alone. CTL lysed A/Taiwan and A/Shanghai (H3N2) virus-infected class I-expressing P815 (H-2d) but not virus-infected EL-4 (H-2b) target cells nor B/Yamagata virus-infected target cells. Virus-infected MHC class II- and class I-expressing A20 (H-2d) targets were also lysed. Depletion of Lyt-2+ (CD8+) T cells with monoclonal antibody completely abrogated lysis of P815 target cells and resulted only in a slight reduction of lysis of A20 target cells. Depletion of L3T4+ (CD4+) T cells or NK cells had minimal effect on lysis of either P815 or A20 target cells. Using limiting dilution analysis, the precursor CTL (pCTL) frequency paralleled CTL activity. Significant CTL activity was detected 7 months after immunization. These results demonstrate that adequate doses of influenza WVV with or without CTB can induce long-lasting influenza A cross-reactive MHC class I-restricted CD8+ CTL response in mice. Thus, coadministration of influenza WVV with CTB may lead to an effective vaccine that stimulates both CTL and antibody responses.
Collapse
Affiliation(s)
- I N Mbawuike
- Department of Microbiology and Immunology, Baylor College of Medicine, Houston, TX 77030-3498
| | | |
Collapse
|
47
|
Nonacs R, Humborg C, Tam JP, Steinman RM. Mechanisms of mouse spleen dendritic cell function in the generation of influenza-specific, cytolytic T lymphocytes. J Exp Med 1992; 176:519-29. [PMID: 1386874 PMCID: PMC2119320 DOI: 10.1084/jem.176.2.519] [Citation(s) in RCA: 108] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We have evaluated the capacity of dendritic cells to function as antigen-presenting cells (APCs) for influenza and have examined their mechanism of action. Virus-pulsed dendritic cells were 100 times more efficient than bulk spleen cells in stimulating cytotoxic T lymphocyte (CTL) formation. The induction of CTLs required neither exogenous lymphokines nor APCs in the responding T cell population. Infectious virus entered dendritic cells through intracellular acidic vacuoles and directed the synthesis of several viral proteins. If ultraviolet (UV)-inactivated or bromelain-treated viruses were used, viral protein synthesis could not be detected, and there was poor induction of CTLs. This indicated that dendritic cells were not capable of processing noninfectious virus onto major histocompatibility complex (MHC) class I molecules. However, UV-inactivated and bromelain-treated viruses were presented efficiently to class II-restricted CD4+ T cells. The CD4+ T cells crossreacted with different strains of influenza and markedly amplified CTL formation. Cell lines that lacked MHC class II, and consequently the capacity to stimulate CD4+ T cells, failed to induce CTLs unless helper lymphokines were added. Similarly, dendritic cells pulsed with the MHC class I-restricted nucleoprotein 147-155 peptide were poor stimulators in the absence of exogenous helper factors. We conclude that the function of dendritic cells as APCs for the generation of virus-specific CTLs in vitro depends measurably upon: (a) charging class I molecules with peptides derived from endogenously synthesized viral antigens, and (b) stimulating a strong CD4+ helper T cell response.
Collapse
Affiliation(s)
- R Nonacs
- Laboratory of Biochemistry, Rockefeller University, New York, New York 10021
| | | | | | | |
Collapse
|
48
|
Shirai M, Akatsuka T, Pendleton CD, Houghten R, Wychowski C, Mihalik K, Feinstone S, Berzofsky JA. Induction of cytotoxic T cells to a cross-reactive epitope in the hepatitis C virus nonstructural RNA polymerase-like protein. J Virol 1992; 66:4098-106. [PMID: 1376366 PMCID: PMC241212 DOI: 10.1128/jvi.66.7.4098-4106.1992] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) have been found to mediate protection in vivo against certain virus infections. CTL also may play an important role in control of infection by hepatitis C virus (HCV), but no CTL epitopes have yet been defined in any HCV protein. The nonstructural protein with homology to RNA polymerase should be a relatively conserved target protein for CTL. To investigate the epitope specificity of CTL specific for this protein, we used 28 peptides from this sequence to study murine CTL. Mice were immunized with a recombinant vaccinia virus expressing the HCV nonstructural region corresponding to the flavivirus NS5 gene (RNA polymerase), and the primed spleen cells were restimulated in vitro with peptides. CTL from H-2d mice responded to a single 16-residue synthetic peptide (HCV 2422 to 2437). This relatively conserved epitope was presented by H-2d class I major histocompatibility complex (MHC) molecules to conventional CD4- CD8+ CTL but was not recognized by CTL restricted by H-2b. Moreover, exon shuffle experiments using several transfectants expressing recombinant Dd/Ld and Kd demonstrated that this peptide is seen in association with alpha 1 and alpha 2 domains of the Dd class I MHC molecule. This peptide differs from the homologous segments of this nonstructural region from three other HCV isolates by one residue each. Variant peptides with single amino acid substitutions were made to test the effect of each residue on the ability to sensitize targets. Neither substitution affected recognition. Therefore, these conservative mutations affected peptide interaction neither with the Dd class I MHC molecule nor with the T-cell receptor. Because these CTL cross-react with all four sequenced isolates of HCV in the United States and Japan, if human CTL display similar cross-reactivity, this peptide may be valuable for studies of HCV diagnosis and vaccine development. Our study provides the first evidence that CD8+ CTL can recognize an epitope from the HCV sequence in association with a class I MHC molecule.
Collapse
Affiliation(s)
- M Shirai
- Molecular Immunogenetics and Vaccine Research Section, National Cancer Institute, Bethesda, Maryland 20892
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Tärnvik A, Eriksson M, Sandström G, Sjöstedt A. Francisella tularensis--a model for studies of the immune response to intracellular bacteria in man. Immunol Suppl 1992; 76:349-54. [PMID: 1526645 PMCID: PMC1421695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
50
|
Kos FJ, Müllbacher A. Specific epitope-induced conversion of CD8+ memory cells into effector cytotoxic T lymphocytes in vitro: presentation of peptide antigen by CD8+ T cells. Eur J Immunol 1992; 22:1595-601. [PMID: 1376266 DOI: 10.1002/eji.1830220637] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The requirements for the conversion of CD8+ memory T cells into effector class I major histocompatibility complex (MHC) Kd-restricted cytotoxic T (Tc) cells in vitro have been studied. Purified CD8+ splenocytes from influenza A/WSN-primed BALB/c (H-2d) mice stimulated with a synthetic nucleoprotein peptide 147-158 R156- (NPP) alone generated Tc cells specific for influenza virus-infected target cells. No additional requirements for accessory cells or their lymphokine products were necessary indicating that peptide antigen (Ag) in association with Kd was presented on CD8+ T cells. The evidence for presentation of NPP by CD8+ T cells was supported by the use of CD8+ memory T cells from semiallogeneic bone marrow radiation chimeras of P1----F1 type (H-2b----[H-2d x H-2b]F1). Memory CD8+ splenocytes from A/WSN-immune chimeras did not develop into secondary effector Tc cells as a result of a 4-day culture with NPP alone, however, were able to do so if NPP was presented by Kd-bearing Ag-presenting cells. In addition, these results exclude the possibility of direct recognition of free NPP molecules by the specific T cell receptor of CD8+ memory T cells. CD8+ memory splenocytes (H-2b) from chimeras were also able to develop into functionally active Tc cells as a result of presentation of Db-restricted synthetic peptide (NP 366-374) with a sequence derived from influenza virus nucleoprotein with high affinity for Db MHC class I molecules. Blockade of endogenously produced interleukin 2 (IL-2) activity by anti-IL-2 or anti-IL-2 receptor monoclonal antibody in the culture of CD8+ memory T cells during a 4-day NPP stimulation completely abolished Tc cell generation, indicating that the utilization of this lymphokine is absolutely required for the secondary Tc cell development. These findings demonstrate that CD8+ memory T cells per se are able to recognize the restimulating epitope as a result of its presentation by CD8+ T cells and develop into cytolytically active and highly specific Tc cells with no requirements for other cellular helper components or their lymphokine products.
Collapse
Affiliation(s)
- F J Kos
- Division of Cell Biology, John Curtin School of Medical Research, Australian National University, Canberra, ACT
| | | |
Collapse
|