1
|
Laphanuwat P, Gomes DCO, Akbar AN. Senescent T cells: Beneficial and detrimental roles. Immunol Rev 2023; 316:160-175. [PMID: 37098109 PMCID: PMC10952287 DOI: 10.1111/imr.13206] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/20/2023] [Accepted: 04/01/2023] [Indexed: 04/27/2023]
Abstract
As the thymus involutes during aging, the T-cell pool has to be maintained by the periodic expansion of preexisting T cells during adulthood. A conundrum is that repeated episodes of activation and proliferation drive the differentiation of T cells toward replicative senescence, due to telomere erosion. This review discusses mechanisms that regulate the end-stage differentiation (senescence) of T cells. Although these cells, within both CD4 and CD8 compartments, lose proliferative activity after antigen-specific challenge, they acquire innate-like immune function. While this may confer broad immune protection during aging, these senescent T cells may also cause immunopathology, especially in the context of excessive inflammation in tissue microenvironments.
Collapse
Affiliation(s)
- Phatthamon Laphanuwat
- Division of MedicineUniversity College LondonLondonUK
- Department of PharmacologyFaculty of Medicine, Khon Kaen UniversityKhon KaenThailand
| | - Daniel Claudio Oliveira Gomes
- Division of MedicineUniversity College LondonLondonUK
- Núcleo de Doenças InfecciosasUniversidade Federal do Espírito SantoVitoriaBrazil
- Núcleo de BiotecnologiaUniversidade Federal do Espírito SantoVitoriaBrazil
| | - Arne N. Akbar
- Division of MedicineUniversity College LondonLondonUK
| |
Collapse
|
2
|
Finn CM, Dhume K, Prokop E, Strutt TM, McKinstry KK. STAT1 Controls the Functionality of Influenza-Primed CD4 T Cells but Therapeutic STAT4 Engagement Maximizes Their Antiviral Impact. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1292-1304. [PMID: 36961447 PMCID: PMC10121883 DOI: 10.4049/jimmunol.2200407] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 02/22/2023] [Indexed: 03/25/2023]
Abstract
It is generally accepted that influenza A virus (IAV) infection promotes a Th1-like CD4 T cell response and that this effector program underlies its protective impact. Canonical Th1 polarization requires cytokine-mediated activation of the transcription factors STAT1 and STAT4 that synergize to maximize the induction of the "master regulator" Th1 transcription factor, T-bet. Here, we determine the individual requirements for these transcription factors in directing the Th1 imprint primed by influenza infection in mice by tracking virus-specific wild-type or T-bet-deficient CD4 T cells in which STAT1 or STAT4 is knocked out. We find that STAT1 is required to protect influenza-primed CD4 T cells from NK cell-mediated deletion and for their expression of hallmark Th1 attributes. STAT1 is also required to prevent type I IFN signals from inhibiting the induction of the Th17 master regulator, Rorγt, in Th17-prone T-bet-/- cells responding to IAV. In contrast, STAT4 expression does not appreciably impact the phenotypic or functional attributes of wild-type or T-bet-/- CD4 T cell responses. However, cytokine-mediated STAT4 activation in virus-specific CD4 T cells enhances their Th1 identity in a T-bet-dependent manner, indicating that influenza infection does not promote maximal Th1 induction. Finally, we show that the T-bet-dependent protective capacity of CD4 T cell effectors against IAV is optimized by engaging both STAT1 and STAT4 during Th1 priming, with important implications for vaccine strategies aiming to generate T cell immunity.
Collapse
Affiliation(s)
- Caroline M. Finn
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Kunal Dhume
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Emily Prokop
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Tara M. Strutt
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - K. Kai McKinstry
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
3
|
Sanchez MV, Ebensen T, Schulze K, Cargnelutti DE, Scodeller EA, Guzmán CA. Protective Efficacy of a Mucosal Influenza Vaccine Formulation Based on the Recombinant Nucleoprotein Co-Administered with a TLR2/6 Agonist BPPcysMPEG. Pharmaceutics 2023; 15:pharmaceutics15030912. [PMID: 36986773 PMCID: PMC10057018 DOI: 10.3390/pharmaceutics15030912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Current influenza vaccines target highly variable surface glycoproteins; thus, mismatches between vaccine strains and circulating strains often diminish vaccine protection. For this reason, there is still a critical need to develop effective influenza vaccines able to protect also against the drift and shift of different variants of influenza viruses. It has been demonstrated that influenza nucleoprotein (NP) is a strong candidate for a universal vaccine, which contributes to providing cross-protection in animal models. In this study, we developed an adjuvanted mucosal vaccine using the recombinant NP (rNP) and the TLR2/6 agonist S-[2,3-bispalmitoyiloxy-(2R)-propyl]-R-cysteinyl-amido-monomethoxyl-poly-ethylene-glycol (BPPcysMPEG). The vaccine efficacy was compared with that observed following parenteral vaccination of mice with the same formulation. Mice vaccinated with 2 doses of rNP alone or co-administered with BPPcysMPEG by the intranasal (i.n.) route showed enhanced antigen-specific humoral and cellular responses. Moreover, NP-specific humoral immune responses, characterized by significant NP-specific IgG and IgG subclass titers in sera and NP-specific IgA titers in mucosal territories, were remarkably increased in mice vaccinated with the adjuvanted formulation as compared with those of the non-adjuvanted vaccination group. The addition of BPPcysMPEG also improved NP-specific cellular responses in vaccinated mice, characterized by robust lymphoproliferation and mixed Th1/Th2/Th17 immune profiles. Finally, it is notable that the immune responses elicited by the novel formulation administered by the i.n. route were able to confer protection against the influenza H1N1 A/Puerto Rico/8/1934 virus.
Collapse
Affiliation(s)
- Maria Victoria Sanchez
- Laboratorio de Inmunología y Desarrollo de Vacunas, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CCT-CONICET, Universidad Nacional de Cuyo, Mendoza M5500, Argentina; (M.V.S.); (D.E.C.); (E.A.S.)
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (T.E.); (K.S.)
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (T.E.); (K.S.)
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (T.E.); (K.S.)
| | - Diego Esteban Cargnelutti
- Laboratorio de Inmunología y Desarrollo de Vacunas, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CCT-CONICET, Universidad Nacional de Cuyo, Mendoza M5500, Argentina; (M.V.S.); (D.E.C.); (E.A.S.)
| | - Eduardo A. Scodeller
- Laboratorio de Inmunología y Desarrollo de Vacunas, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CCT-CONICET, Universidad Nacional de Cuyo, Mendoza M5500, Argentina; (M.V.S.); (D.E.C.); (E.A.S.)
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (T.E.); (K.S.)
- Correspondence: ; Tel.: +49-531-61814600; Fax: +49-531-618414699
| |
Collapse
|
4
|
Swain SL, Jones MC, Devarajan P, Xia J, Dutton RW, Strutt TM, McKinstry KK. Durable CD4 T-Cell Memory Generation Depends on Persistence of High Levels of Infection at an Effector Checkpoint that Determines Multiple Fates. Cold Spring Harb Perspect Biol 2021; 13:a038182. [PMID: 33903157 PMCID: PMC8559547 DOI: 10.1101/cshperspect.a038182] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We have discovered that the determination of CD4 effector and memory fates after infection is regulated not only by initial signals from antigen and pathogen recognition, but also by a second round of such signals at a checkpoint during the effector response. Signals to effectors determine their subsequent fate, inducing further progression to tissue-restricted follicular helpers, cytotoxic CD4 effectors, and long-lived memory cells. The follicular helpers help the germinal center B-cell responses that give rise to high-affinity long-lived antibody responses and memory B cells that synergize with T-cell memory to provide robust long-lived protection. We postulate that inactivated vaccines do not provide extended signals from antigen and pathogen beyond a few days, and thus elicit ineffective CD4 T- and B-cell effector responses and memory. Defining the mechanisms that underlie effective responses should provide insights necessary to develop vaccine strategies that induce more effective and durable immunity.
Collapse
Affiliation(s)
- Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Michael C Jones
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Priyadharshini Devarajan
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Jingya Xia
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Richard W Dutton
- Department of Pathology, University of Massachusetts Medical School, 368 Plantation Ave, Worcester, Massachusetts 01655, USA
| | - Tara M Strutt
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA
| | - K Kai McKinstry
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32827, USA
| |
Collapse
|
5
|
Memory CD4 + T Cells in Immunity and Autoimmune Diseases. Cells 2020; 9:cells9030531. [PMID: 32106536 PMCID: PMC7140455 DOI: 10.3390/cells9030531] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 12/26/2022] Open
Abstract
CD4+ T helper (Th) cells play central roles in immunity in health and disease. While much is known about the effector function of Th cells in combating pathogens and promoting autoimmune diseases, the roles and biology of memory CD4+ Th cells are complex and less well understood. In human autoimmune diseases such as multiple sclerosis (MS), there is a critical need to better understand the function and biology of memory T cells. In this review article we summarize current concepts in the field of CD4+ T cell memory, including natural history, developmental pathways, subsets, and functions. Furthermore, we discuss advancements in the field of the newly-described CD4+ tissue-resident memory T cells and of CD4+ memory T cells in autoimmune diseases, two major areas of important unresolved questions in need of answering to advance new vaccine design and development of novel treatments for CD4+ T cell-mediated autoimmune diseases.
Collapse
|
6
|
Dai M, Xu C, Chen W, Liao M. Progress on chicken T cell immunity to viruses. Cell Mol Life Sci 2019; 76:2779-2788. [PMID: 31101935 PMCID: PMC11105491 DOI: 10.1007/s00018-019-03117-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/14/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022]
Abstract
Avian virus infection remains one of the most important threats to the poultry industry. Pathogens such as avian influenza virus (AIV), avian infectious bronchitis virus (IBV), and infectious bursal disease virus (IBDV) are normally controlled by antibodies specific for surface proteins and cellular immune responses. However, standard vaccines aimed at inducing neutralizing antibodies must be administered annually and can be rendered ineffective because immune-selective pressure results in the continuous mutation of viral surface proteins of different strains circulating from year to year. Chicken T cells have been shown to play a crucial role in fighting virus infection, offering lasting and cross-strain protection, and offer the potential for developing universal vaccines. This review provides an overview of our current knowledge of chicken T cell immunity to viruses. More importantly, we point out the limitations and barriers of current research and a potential direction for future studies.
Collapse
Affiliation(s)
- Manman Dai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
| | - Chenggang Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture, Guangzhou, People's Republic of China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China
| | - Weisan Chen
- T Cell Lab, Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Bundoora, Australia.
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, People's Republic of China.
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture, Guangzhou, People's Republic of China.
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, People's Republic of China.
| |
Collapse
|
7
|
Immunogenicity and safety of a quadrivalent plant-derived virus like particle influenza vaccine candidate-Two randomized Phase II clinical trials in 18 to 49 and ≥50 years old adults. PLoS One 2019; 14:e0216533. [PMID: 31166987 PMCID: PMC6550445 DOI: 10.1371/journal.pone.0216533] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/11/2019] [Indexed: 11/19/2022] Open
Abstract
Background New influenza vaccines eliciting more effective protection are needed, particularly for the elderly who paid a large and disproportional toll of hospitalization and dead during seasonal influenza epidemics. Low (≤15 μg/strain) doses of a new plant-derived virus-like-particle (VLP) vaccine candidate have been shown to induce humoral and cellular responses against both homologous and heterologous strains in healthy adults 18–64 years of age. The two clinical trials reported here addressed the safety and immunogenicity of higher doses (≥15 μg/strain) of quadrivalent VLP candidate vaccine on 18–49 years old and ≥50 years old subjects. We also investigated the impact of alum on the immunogenicity induced by lower doses of the vaccine candidate. Method In the first Phase II trial reported here (NCT02233816), 18–49 year old subjects received 15, 30 or 60 μg/strain of a hemagglutinin-bearing quadrivalent virus-like particle (QVLP) vaccine or placebo. In the second trial (NCT02236052), ≥50 years old subjects received QVLP as above or placebo with additional groups receiving 7.5 or 15 μg/strain with alum. Along with safety recording, the humoral and cell-mediated immune responses were analyzed. Results Local and systemic side-effects were similar to those reported previously. The QVLP vaccine induced significant homologous and heterologous antibody responses at the two higher doses, the addition of alum having little-to-no effect. Serologic outcomes tended to be lower in ≥50 years old subjects previously vaccinated. The candidate vaccine also consistently elicited both homologous and heterologous antigen-specific CD4+ T cells characterized by their production of interferon-gamma (IFN-γ), interleukine-2 (IL-2) and/or tumor-necrosis factor alpha (TNF-α) upon ex vivo antigenic restimulation. Conclusion Overall, the 30 μg dose produced the most consistent humoral and cellular responses in both 18–49 and ≥50 years old subjects, strongly supporting the clinical development of this candidate vaccine. That particular dose was chosen to test in the ongoing Phase III clinical trial.
Collapse
|
8
|
Mouse Models Reveal Role of T-Cytotoxic and T-Reg Cells in Immune Response to Influenza: Implications for Vaccine Design. Viruses 2019; 11:v11010052. [PMID: 30641955 PMCID: PMC6356589 DOI: 10.3390/v11010052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 01/04/2023] Open
Abstract
Immunopathologic examination of the lungs of mouse models of experimental influenza virus infection provides new insights into the immune response in this disease. First, there is rapidly developing perivascular and peribronchial infiltration of the lung with T-cells. This is followed by invasion of T-cells into the bronchiolar epithelium, and separation of epithelial cells from each other and from the basement membrane leading to defoliation of the bronchial epithelium. The intraepithelial reaction may involve either CD8 or CD4 T-cytotoxic cells and is analogous to a viral exanthema of the skin, such as measles and smallpox, which occur when the immune response against these infections is activated and the infected cells are attacked by T-cytotoxic cells. Then there is formation of B-cell follicles adjacent to bronchi, i.e., induced bronchial associated lymphoid tissue (iBALT). iBALT reacts like the cortex of a lymph node and is a site for a local immune response not only to the original viral infection, but also related viral infections (heterologous immunity). Proliferation of Type II pneumocytes and/or terminal bronchial epithelial cells may extend into the adjacent lung leading to large zones filled with tumor-like epithelial cells. The effective killing of influenza virus infected epithelial cells by T-cytotoxic cells and induction of iBALT suggests that adding the induction of these components might greatly increase the efficacy of influenza vaccination.
Collapse
|
9
|
Broad CD8 + T cell cross-recognition of distinct influenza A strains in humans. Nat Commun 2018; 9:5427. [PMID: 30575715 PMCID: PMC6303473 DOI: 10.1038/s41467-018-07815-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/23/2018] [Indexed: 11/29/2022] Open
Abstract
Newly-emerged and vaccine-mismatched influenza A viruses (IAVs) result in a rapid global spread of the virus due to minimal antibody-mediated immunity. In that case, established CD8+ T-cells can reduce disease severity. However, as mutations occur sporadically within immunogenic IAV-derived T-cell peptides, understanding of T-cell receptor (TCRαβ) cross-reactivity towards IAV variants is needed for a vaccine design. Here, we investigate TCRαβ cross-strain recognition across IAV variants within two immunodominant human IAV-specific CD8+ T-cell epitopes, HLA-B*37:01-restricted NP338-346 (B37-NP338) and HLA-A*01:01-restricted NP44-52 (A1-NP44). We find high abundance of cross-reactive TCRαβ clonotypes recognizing distinct IAV variants. Structures of the wild-type and variant peptides revealed preserved conformation of the bound peptides. Structures of a cross-reactive TCR-HLA-B37-NP338 complex suggest that the conserved conformation of the variants underpins TCR cross-reactivity. Overall, cross-reactive CD8+ T-cell responses, underpinned by conserved epitope structure, facilitates recognition of distinct IAV variants, thus CD8+ T-cell-targeted vaccines could provide protection across different IAV strains. Mutations within immunological epitope containing regions of influenza A virus can impair the established immune response between influenza strains and could impact rational vaccine design. Here Grant et al. examine the presence, structural impact and cross reactivity of two human immunodominant influenza epitope variants.
Collapse
|
10
|
Singh RK, Dhama K, Karthik K, Khandia R, Munjal A, Khurana SK, Chakraborty S, Malik YS, Virmani N, Singh R, Tripathi BN, Munir M, van der Kolk JH. A Comprehensive Review on Equine Influenza Virus: Etiology, Epidemiology, Pathobiology, Advances in Developing Diagnostics, Vaccines, and Control Strategies. Front Microbiol 2018; 9:1941. [PMID: 30237788 PMCID: PMC6135912 DOI: 10.3389/fmicb.2018.01941] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/31/2018] [Indexed: 01/23/2023] Open
Abstract
Among all the emerging and re-emerging animal diseases, influenza group is the prototype member associated with severe respiratory infections in wide host species. Wherein, Equine influenza (EI) is the main cause of respiratory illness in equines across globe and is caused by equine influenza A virus (EIV-A) which has impacted the equine industry internationally due to high morbidity and marginal morality. The virus transmits easily by direct contact and inhalation making its spread global and leaving only limited areas untouched. Hitherto reports confirm that this virus crosses the species barriers and found to affect canines and few other animal species (cat and camel). EIV is continuously evolving with changes at the amino acid level wreaking the control program a tedious task. Until now, no natural EI origin infections have been reported explicitly in humans. Recent advances in the diagnostics have led to efficient surveillance and rapid detection of EIV infections at the onset of outbreaks. Incessant surveillance programs will aid in opting a better control strategy for this virus by updating the circulating vaccine strains. Recurrent vaccination failures against this virus due to antigenic drift and shift have been disappointing, however better understanding of the virus pathogenesis would make it easier to design effective vaccines predominantly targeting the conserved epitopes (HA glycoprotein). Additionally, the cold adapted and canarypox vectored vaccines are proving effective in ceasing the severity of disease. Furthermore, better understanding of its genetics and molecular biology will help in estimating the rate of evolution and occurrence of pandemics in future. Here, we highlight the advances occurred in understanding the etiology, epidemiology and pathobiology of EIV and a special focus is on designing and developing effective diagnostics, vaccines and control strategies for mitigating the emerging menace by EIV.
Collapse
Affiliation(s)
- Raj K. Singh
- ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | | | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, West Tripura, India
| | - Yashpal S. Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | | | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | | | - Muhammad Munir
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, United Kingdom
| | - Johannes H. van der Kolk
- Division of Clinical Veterinary Medicine, Swiss Institute for Equine Medicine (ISME), Vetsuisse Faculty, University of Bern and Agroscope, Bern, Switzerland
| |
Collapse
|
11
|
Dhume K, McKinstry KK. Early programming and late-acting checkpoints governing the development of CD4 T-cell memory. Immunology 2018; 155:53-62. [PMID: 29701246 DOI: 10.1111/imm.12942] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 12/25/2022] Open
Abstract
CD4 T cells contribute to protection against pathogens through numerous mechanisms. Incorporating the goal of memory CD4 T-cell generation into vaccine strategies therefore offers a powerful approach to improve their efficacy, especially in situations where humoral responses alone cannot confer long-term immunity. These threats include viruses such as influenza that mutate coat proteins to avoid neutralizing antibodies, but that are targeted by T cells that recognize more conserved protein epitopes shared by different strains. A major barrier in the design of such vaccines is that the mechanisms controlling the efficiency with which memory cells form remain incompletely understood. Here, we discuss recent insights into fate decisions controlling memory generation. We focus on the importance of three general cues: interleukin-2, antigen and co-stimulatory interactions. It is increasingly clear that these signals have a powerful influence on the capacity of CD4 T cells to form memory during two distinct phases of the immune response. First, through 'programming' that occurs during initial priming, and second, through 'checkpoints' that operate later during the effector stage. These findings indicate that novel vaccine strategies must seek to optimize cognate interactions, during which interleukin-2-, antigen- and co-stimulation-dependent signals are tightly linked, well beyond initial antigen encounter to induce robust memory CD4 T cells.
Collapse
Affiliation(s)
- Kunal Dhume
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Karl Kai McKinstry
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
12
|
Zheng J, Perlman S. Immune responses in influenza A virus and human coronavirus infections: an ongoing battle between the virus and host. Curr Opin Virol 2018; 28:43-52. [PMID: 29172107 PMCID: PMC5835172 DOI: 10.1016/j.coviro.2017.11.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/02/2017] [Indexed: 12/25/2022]
Abstract
Respiratory viruses, especially influenza A viruses and coronaviruses such as MERS-CoV, represent continuing global threats to human health. Despite significant advances, much needs to be learned. Recent studies in virology and immunology have improved our understanding of the role of the immune system in protection and in the pathogenesis of these infections and of co-evolution of viruses and their hosts. These findings, together with sophisticated molecular structure analyses, omics tools and computer-based models, have helped delineate the interaction between respiratory viruses and the host immune system, which will facilitate the development of novel treatment strategies and vaccines with enhanced efficacy.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA 52242, United States
| | - Stanley Perlman
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
13
|
Pillet S, Aubin É, Trépanier S, Poulin JF, Yassine-Diab B, Ter Meulen J, Ward BJ, Landry N. Humoral and cell-mediated immune responses to H5N1 plant-made virus-like particle vaccine are differentially impacted by alum and GLA-SE adjuvants in a Phase 2 clinical trial. NPJ Vaccines 2018; 3:3. [PMID: 29387473 PMCID: PMC5780465 DOI: 10.1038/s41541-017-0043-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/11/2017] [Accepted: 12/15/2017] [Indexed: 12/19/2022] Open
Abstract
The hemagglutinination inhibition (HI) response remains the gold standard used for the licensure of influenza vaccines. However, cell-mediated immunity (CMI) deserves more attention, especially when evaluating H5N1 influenza vaccines that tend to induce poor HI response. In this study, we measured the humoral response (HI) and CMI (flow cytometry) during a Phase II dose-ranging clinical trial (NCT01991561). Subjects received two intramuscular doses, 21 days apart, of plant-derived virus-like particles (VLP) presenting the A/Indonesia/05/2005 H5N1 influenza hemagglutinin protein (H5) at the surface of the VLP (H5VLP). The vaccine was co-administrated with Alhydrogel® or with a glucopyranosyl lipid adjuvant-stable emulsion (GLA-SE). We demonstrated that low doses (3.75 or 7.5 μg H5VLP) of GLA-SE-adjuvanted vaccines induced HI responses that met criteria for licensure at both antigen doses tested. Alhydrogel adjuvanted vaccines induced readily detectable HI response that however failed to meet licensure criteria at any of three doses (10, 15 and 20 μg) tested. The H5VLP also induced a sustained (up to 6 months) polyfunctional and cross-reactive HA-specific CD4+ T cell response in all vaccinated groups. Interestingly, the frequency of central memory Th1-primed precursor cells before the boost significantly correlated with HI titers 21 days after the boost. The ability of the low dose GLA-SE-adjuvanted H5VLP to elicit both humoral response and a sustained cross-reactive CMI in healthy adults is very attractive and could result in significant dose-sparing in a pandemic situation.
Collapse
Affiliation(s)
- Stéphane Pillet
- 1Medicago Inc., Québec, G1V 3V9 QC Canada.,2Research Institute of the McGill University Health Centre, Montreal, H4A 3J1 QC Canada
| | - Éric Aubin
- 1Medicago Inc., Québec, G1V 3V9 QC Canada
| | | | | | | | - Jan Ter Meulen
- Immune Design, Seattle, WA 98102 USA.,Immune Design, San Francisco, CA 94080-7006 USA
| | - Brian J Ward
- 2Research Institute of the McGill University Health Centre, Montreal, H4A 3J1 QC Canada
| | | |
Collapse
|
14
|
Ward BJ, Pillet S, Charland N, Trepanier S, Couillard J, Landry N. The establishment of surrogates and correlates of protection: Useful tools for the licensure of effective influenza vaccines? Hum Vaccin Immunother 2018; 14:647-656. [PMID: 29252098 PMCID: PMC5861778 DOI: 10.1080/21645515.2017.1413518] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The search for a test that can predict vaccine efficacy is an important part of any vaccine development program. Although regulators hesitate to acknowledge any test as a true ‘correlate of protection’, there are many precedents for defining ‘surrogate’ assays. Surrogates can be powerful tools for vaccine optimization, licensure, comparisons between products and development of improved products. When such tests achieve ‘reference’ status however, they can inadvertently become barriers to new technologies that do not work the same way as existing vaccines. This is particularly true when these tests are based upon circularly-defined ‘reference’ or, even worse, proprietary reagents. The situation with inactivated influenza vaccines is a good example of this phenomenon. The most frequently used tests to define vaccine-induced immunity are all serologic assays: hemagglutination inhibition (HI), single radial hemolysis (SRH) and microneutralization (MN). The first two, and particularly the HI assay, have achieved reference status and criteria have been established in many jurisdictions for their use in licensing new vaccines and to compare the performance of different vaccines. However, all of these assays are based on biological reagents that are notoriously difficult to standardize and can vary substantially by geography, by chance (i.e. developing reagents in eggs that may not antigenitically match wild-type viruses) and by intention (ie: choosing reagents that yield the most favorable results). This review describes attempts to standardize these assays to improve their performance as surrogates, the dangers of over-reliance on ‘reference’ serologic assays, the ways that manufacturers can exploit the existing regulatory framework to make their products ‘look good’ and the implications of this long-established system for the introduction of novel influenza vaccines.
Collapse
Affiliation(s)
- Brian J Ward
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | - Stephane Pillet
- a Research Institute of the McGill University Health Centre, Infectious Diseases Division , Montreal , QC , Canada.,b Medicago Inc , Québec , QC , Canada
| | | | | | | | | |
Collapse
|
15
|
M2e-tetramer-specific memory CD4 T cells are broadly protective against influenza infection. Mucosal Immunol 2018; 11:273-289. [PMID: 28295019 DOI: 10.1038/mi.2017.14] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/24/2017] [Indexed: 02/04/2023]
Abstract
Matrix protein 2 ectodomain (M2e) is considered an attractive component of a broadly protective, universal influenza A vaccine. Here we challenge the canonical view that antibodies against M2e are the prime effectors of protection. Intranasal immunizations of Balb/c mice with CTA1-3M2e-DD-generated M2e-specific memory CD4 T cells that were I-Ad restricted and critically protected against infection, even in the complete absence of antibodies, as observed in JhD mice. Whereas some M2e-tetramer-specific memory CD4 T cells resided in spleen and lymph nodes, the majority were lung-resident Th17 cells, that rapidly expanded upon a viral challenge infection. Indeed, immunized IL-17A-/- mice were significantly less well protected compared with wild-type mice despite exhibiting comparable antibody levels. Similarly, poor protection was also observed in congenic Balb/B (H-2b) mice, which failed to develop M2e-specific CD4 T cells, but exhibited comparable antibody levels. Lung-resident CD69+ CD103low M2e-specific memory CD4 T cells were αβ TCR+ and 50% were Th17 cells that were associated with an early influx of neutrophils after virus challenge. Adoptively transferred M2e memory CD4 T cells were strong helper T cells, which accelerated M2e- but more importantly also hemagglutinin-specific IgG production. Thus, for the first time we demonstrate that M2e-specific memory CD4 T cells are broadly protective.
Collapse
|
16
|
Alam S, Chan C, Qiu X, Shannon I, White CL, Sant AJ, Nayak JL. Selective pre-priming of HA-specific CD4 T cells restores immunological reactivity to HA on heterosubtypic influenza infection. PLoS One 2017; 12:e0176407. [PMID: 28493882 PMCID: PMC5426616 DOI: 10.1371/journal.pone.0176407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/09/2017] [Indexed: 01/07/2023] Open
Abstract
A hallmark of the immune response to influenza is repeated encounters with proteins containing both genetically conserved and variable components. Therefore, the B and T cell repertoire is continually being remodeled, with competition between memory and naïve lymphocytes. Our previous work using a mouse model of secondary heterosubtypic influenza infection has shown that this competition results in a focusing of CD4 T cell response specificity towards internal virion proteins with a selective decrease in CD4 T cell reactivity to the novel HA epitopes. Strikingly, this shift in CD4 T cell specificity was associated with a diminished anti-HA antibody response. Here, we sought to determine whether the loss in HA-specific reactivity that occurs as a consequence of immunological memory could be reversed by selectively priming HA-specific CD4 T cells prior to secondary infection. Using a peptide-based priming strategy, we found that selective expansion of the anti-HA CD4 T cell memory repertoire enhanced HA-specific antibody production upon heterosubtypic infection. These results suggest that the potentially deleterious consequences of repeated exposure to conserved influenza internal virion proteins could be reversed by vaccination strategies that selectively arm the HA-specific CD4 T cell compartment. This could be a potentially useful pre-pandemic vaccination strategy to promote accelerated neutralizing antibody production on challenge with a pandemic influenza strain that contains few conserved HA epitopes.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- CD4-Positive T-Lymphocytes/immunology
- Epitopes/immunology
- Hemagglutinins, Viral/immunology
- Humans
- Immunologic Memory
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza Vaccines/immunology
- Influenza Vaccines/therapeutic use
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Pandemics
- Vaccination
Collapse
Affiliation(s)
- Shabnam Alam
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Cory Chan
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Ian Shannon
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Chantelle L. White
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Jennifer L. Nayak
- Department of Pediatrics, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Juno JA, van Bockel D, Kent SJ, Kelleher AD, Zaunders JJ, Munier CML. Cytotoxic CD4 T Cells-Friend or Foe during Viral Infection? Front Immunol 2017; 8:19. [PMID: 28167943 PMCID: PMC5253382 DOI: 10.3389/fimmu.2017.00019] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/05/2017] [Indexed: 01/03/2023] Open
Abstract
CD4 T cells with cytotoxic function were once thought to be an artifact due to long-term in vitro cultures but have in more recent years become accepted and reported in the literature in response to a number of viral infections. In this review, we focus on cytotoxic CD4 T cells in the context of human viral infections and in some infections that affect mice and non-human primates. We examine the effector mechanisms used by cytotoxic CD4 cells, the phenotypes that describe this population, and the transcription factors and pathways that lead to their induction following infection. We further consider the cells that are the predominant targets of this effector subset and describe the viral infections in which CD4 cytotoxic T lymphocytes have been shown to play a protective or pathologic role. Cytotoxic CD4 T cells are detected in the circulation at much higher levels than previously realized and are now recognized to have an important role in the immune response to viral infections.
Collapse
Affiliation(s)
- Jennifer A Juno
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne , Melbourne, VIC , Australia
| | - David van Bockel
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia , Sydney, NSW , Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia; Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - John J Zaunders
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - C Mee Ling Munier
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia , Sydney, NSW , Australia
| |
Collapse
|
18
|
Strutt TM, McKinstry KK, Kuang Y, Finn CM, Hwang JH, Dhume K, Sell S, Swain SL. Direct IL-6 Signals Maximize Protective Secondary CD4 T Cell Responses against Influenza. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:3260-3270. [PMID: 27647834 PMCID: PMC5101150 DOI: 10.4049/jimmunol.1600033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/22/2016] [Indexed: 01/08/2023]
Abstract
Memory T cells can often respond against pathogens that have evaded neutralizing Abs and are thus key to vaccine-induced protection, yet the signals needed to optimize their responses are unclear. In this study, we identify a dramatic and selective requirement for IL-6 to achieve optimal memory CD4 T cell recall following heterosubtypic influenza A virus (IAV) challenge of mice primed previously with wild-type or attenuated IAV strains. Through analysis of endogenous T cell responses and adoptive transfer of IAV-specific memory T cell populations, we find that without IL-6, CD4+, but not CD8+, secondary effector populations expand less and have blunted function and antiviral impact. Early and direct IL-6 signals to memory CD4 T cells are required to program maximal secondary effector responses at the site of infection during heterosubtypic challenge, indicating a novel role for a costimulatory cytokine in recall responses.
Collapse
Affiliation(s)
- Tara M Strutt
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827;
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Karl Kai McKinstry
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827;
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Yi Kuang
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605; and
| | - Caroline M Finn
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| | - Ji Hae Hwang
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| | - Kunal Dhume
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827
| | | | - Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605; and
| |
Collapse
|
19
|
Pillet S, Aubin É, Trépanier S, Bussière D, Dargis M, Poulin JF, Yassine-Diab B, Ward BJ, Landry N. A plant-derived quadrivalent virus like particle influenza vaccine induces cross-reactive antibody and T cell response in healthy adults. Clin Immunol 2016; 168:72-87. [PMID: 26987887 DOI: 10.1016/j.clim.2016.03.008] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/22/2016] [Accepted: 03/07/2016] [Indexed: 01/09/2023]
Abstract
Recent issues regarding efficacy of influenza vaccines have re-emphasized the need of new approaches to face this major public health issue. In a phase 1-2 clinical trial, healthy adults received one intramuscular dose of a seasonal influenza plant-based quadrivalent virus-like particle (QVLP) vaccine or placebo. The hemagglutination inhibition (HI) titers met all the European licensure criteria for the type A influenza strains at the 3μg/strain dose and for all four strains at the higher dosages 21days after immunization. High HI titers were maintained for most of the strains 6months after vaccination. QVLP vaccine induced a substantial and sustained increase of hemagglutinin-specific polyfunctional CD4 T cells, mainly transitional memory and TEMRA effector IFN-γ(+) CD4 T cells. A T cells cross-reactive response was also observed against A/Hong-Kong/1/1968 H3N2 and B/Massachusetts/2/2012. Plant-based QVLP offers an attractive alternative manufacturing method for producing effective and HA-strain matching seasonal influenza vaccines.
Collapse
Affiliation(s)
- Stéphane Pillet
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9; Research Institute of the McGill University Health Centre, 2155 Guy Street, 5th Floor, Montreal, QC, Canada, H3H 2R9
| | - Éric Aubin
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | - Sonia Trépanier
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | - Diane Bussière
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | - Michèle Dargis
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9
| | | | - Bader Yassine-Diab
- ImmuneCarta, 201 Avenue du Président-Kennedy, Montreal, QC, Canada, H2X 3Y7
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, 2155 Guy Street, 5th Floor, Montreal, QC, Canada, H3H 2R9
| | - Nathalie Landry
- Medicago Inc., 1020 route de l'Église office 600, Québec, QC, Canada, G1V 3V9.
| |
Collapse
|
20
|
Brown DM, Lampe AT, Workman AM. The Differentiation and Protective Function of Cytolytic CD4 T Cells in Influenza Infection. Front Immunol 2016; 7:93. [PMID: 27014272 PMCID: PMC4783394 DOI: 10.3389/fimmu.2016.00093] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/25/2016] [Indexed: 12/24/2022] Open
Abstract
CD4 T cells that recognize peptide antigen in the context of class II MHC can differentiate into various subsets that are characterized by their helper functions. However, increasing evidence indicates that CD4 cells with direct cytolytic activity (CD4 CTL) play a role in chronic as well as acute infections, such as influenza A virus (IAV) infection. In the last couple of decades, techniques to measure the frequency and activity of these cytolytic cells has demonstrated their abundance in infections, such as human immunodeficiency virus, mouse pox, murine gamma herpes virus, cytomegalovirus, Epstein-Barr virus, and influenza among others. We now appreciate a greater role for CD4 CTL as direct effectors in viral infections and antitumor immunity through their ability to acquire perforin-mediated cytolytic activity and contribution to lysis of virally infected targets or tumors. As early as the 1980s, CD4 T cell clones with cytolytic potential were identified after influenza virus infection, yet much of this early work was dependent on in vitro culture and little was known about the physiological relevance of CD4 CTL. Here, we discuss the direct role CD4 CTL play in protection against lethal IAV infection and the factors that drive the generation of perforin-mediated lytic activity in CD4 cells in vivo during IAV infection. While focusing on CD4 CTL generated during IAV infection, we pull comparisons from the literature in other antiviral and antitumor systems. Further, we highlight what is currently known about CD4 CTL secondary and memory responses, as well as vaccination strategies to induce these potent killer cells that provide an extra layer of cell-mediated immune protection against heterosubtypic IAV infection.
Collapse
Affiliation(s)
- Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Anna T Lampe
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Aspen M Workman
- Nebraska Center for Virology, University of Nebraska-Lincoln , Lincoln, NE , USA
| |
Collapse
|
21
|
Pillet S, Racine T, Nfon C, Di Lenardo TZ, Babiuk S, Ward BJ, Kobinger GP, Landry N. Plant-derived H7 VLP vaccine elicits protective immune response against H7N9 influenza virus in mice and ferrets. Vaccine 2015; 33:6282-9. [PMID: 26432915 DOI: 10.1016/j.vaccine.2015.09.065] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/18/2015] [Accepted: 09/19/2015] [Indexed: 12/23/2022]
Abstract
In March 2013, the Chinese Centre for Disease Control and Prevention confirmed the first reported case of human infection with an avian influenza A H7N9 virus. Infection with this virus often caused severe pneumonia and acute respiratory distress syndrome resulting in a case fatality rate >35%. The risk of pandemic highlighted, once again, the need for a more rapid and scalable vaccine response capability. Here, we describe the rapid (19 days) development of a plant-derived VLP vaccine based on the hemagglutinin sequence of influenza H7N9 A/Hangzhou/1/2013. The immunogenicity of the H7 VLP vaccine was assessed in mice and ferrets after one or two intramuscular dose(s) with and without adjuvant (alum or GLA-SE™). In ferrets, we also measured H7-specific cell-mediated immunity. The mice and ferrets were then challenged with H7N9 A/Anhui/1/2013 influenza virus. A single immunization with the adjuvanted vaccine elicited a strong humoral response and protected mice against an otherwise lethal challenge. Two doses of unadjuvanted vaccine significantly increased humoral response and resulted in 100% protection with significant reduction of clinical signs leading to nearly asymptomatic infections. In ferrets, a single immunization with the alum-adjuvanted H7 VLP vaccine induced strong humoral and CMI responses with antigen-specific activation of CD3(+) T cells. Compared to animals injected with placebo, ferrets vaccinated with alum-adjuvanted vaccine displayed no weight loss during the challenge. Moreover, the vaccination significantly reduced the viral load in lungs and nasal washes 3 days after the infection. This candidate plant-made H7 vaccine therefore induced protective responses after either one adjuvanted or two unadjuvanted doses. Studies are currently ongoing to better characterize the immune response elicited by the plant-derived VLP vaccines. Regardless, these data are very promising for the rapid production of an immunogenic and protective vaccine against this potentially pandemic virus.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/blood
- Body Weight
- Disease Models, Animal
- Female
- Ferrets
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunization Schedule
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/isolation & purification
- Injections, Intramuscular
- Lung/virology
- Male
- Mice, Inbred BALB C
- Nasal Cavity/virology
- Orthomyxoviridae Infections/pathology
- Orthomyxoviridae Infections/prevention & control
- Placebos/administration & dosage
- Plants, Genetically Modified
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Survival Analysis
- Nicotiana
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/isolation & purification
- Viral Load
Collapse
Affiliation(s)
- S Pillet
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada; Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - T Racine
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - C Nfon
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - T Z Di Lenardo
- Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - S Babiuk
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada; Department of Immunology, University of Manitoba, MB, Canada
| | - B J Ward
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada
| | - G P Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Immunology, University of Manitoba, MB, Canada; Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - N Landry
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada.
| |
Collapse
|
22
|
Abstract
Immunologic memory is the adaptive immune system's powerful ability to remember a previous antigen encounter and react with accelerated vigor upon antigen re-exposure. It provides durable protection against reinfection with pathogens and is the foundation for vaccine-induced immunity. Unlike the relatively restricted immunologic purview of memory B cells and CD8 T cells, the field of CD4 T-cell memory must account for multiple distinct lineages with diverse effector functions, the issue of lineage commitment and plasticity, and the variable distribution of memory cells within each lineage. Here, we discuss the evidence for lineage-specific CD4 T-cell memory and summarize the known factors contributing to memory-cell generation, plasticity, and long-term maintenance.
Collapse
Affiliation(s)
- David J Gasper
- Department of Pathobiological Sciences; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Melba Marie Tejera
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - M Suresh
- Department of Pathobiological Sciences; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
23
|
|
24
|
Mohn KGI, Bredholt G, Brokstad KA, Pathirana RD, Aarstad HJ, Tøndel C, Cox RJ. Longevity of B-cell and T-cell responses after live attenuated influenza vaccination in children. J Infect Dis 2014; 211:1541-9. [PMID: 25425696 PMCID: PMC4407761 DOI: 10.1093/infdis/jiu654] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 11/11/2014] [Indexed: 12/31/2022] Open
Abstract
Background. The live attenuated influenza vaccine (LAIV) is the preferred vaccine for children, but the mechanisms behind protective immune responses are unclear, and the duration of immunity remains to be elucidated. This study reports on the longevity of B-cell and T-cell responses elicited by the LAIV. Methods. Thirty-eight children (3–17 years old) were administered seasonal LAIV. Blood samples were collected before vaccination with sequential sampling up to 1 year after vaccination. Humoral responses were evaluated by a hemagglutination inhibition assay, and memory B-cell responses were evaluated by an enzyme-linked immunosorbent spot assay (ELISpot). T-cell responses were evaluated by interferon γ (IFN-γ) ELISpot analysis, and intracellular cytokine staining of CD4+ T cells for detection of IFN-γ, interleukin 2, and tumor necrosis factor α was performed using flow cytometry. Results. LAIV induced significant increases in B-cell and T-cell responses, which were sustained at least 1 year after vaccination. Strain variations were observed, in which the B strain elicited stronger responses. IFN-γ–expressing T cell counts increased significantly, and remained higher than prevaccination levels 1 year later. Expression of T-helper type 1 intracellular cytokines (interleukin 2, IFN-γ, and tumor necrosis factor α) increased after 1 dose and were boosted after the second dose. Hemagglutination inhibition titers were sustained for 1 year. Vaccine-induced memory B cell counts were significantly increased, and the response persisted for one year. Conclusions. LAIV elicited B-cell and T-cell responses that persisted for at least 1 year in children. This is a novel finding that will aid future vaccine policy.
Collapse
Affiliation(s)
| | - Geir Bredholt
- Influenza Center K. G. Jebsen Center for Influenza Vaccines
| | - Karl A Brokstad
- Broegelman Research Laboratory, Department of Clinical Science
| | | | - Hans J Aarstad
- Department of Clinical Medicine, University of Bergen Department of Otolaryngology/Head and Neck Surgery
| | - Camilla Tøndel
- Department of Clinical Medicine, University of Bergen Department of Pediatrics
| | - Rebecca J Cox
- Influenza Center K. G. Jebsen Center for Influenza Vaccines Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
25
|
Landry N, Pillet S, Favre D, Poulin JF, Trépanier S, Yassine-Diab B, Ward BJ. Influenza virus-like particle vaccines made in Nicotiana benthamiana elicit durable, poly-functional and cross-reactive T cell responses to influenza HA antigens. Clin Immunol 2014; 154:164-77. [PMID: 25128897 DOI: 10.1016/j.clim.2014.08.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/26/2014] [Accepted: 08/06/2014] [Indexed: 12/14/2022]
Abstract
Cell-mediated immunity plays a major role in long-lived, cross-reactive protection against influenza virus. We measured long-term poly-functional and cross-reactive T cell responses to influenza hemagglutinin (HA) elicited by a new plant-made Virus-Like Particle (VLP) vaccine targeting either H1N1 A/California/7/09 (H1) or H5N1 A/Indonesia/5/05 (H5). In two independent clinical trials, we characterized the CD4(+) and CD8(+) T cell homotypic and heterotypic responses 6 months after different vaccination regimens. Responses of VLP-vaccinated subjects were compared with placebo and/or a commercial trivalent inactivated vaccine (TIV:Fluzone™) recipients. Both H1 and H5 VLP vaccines elicited significantly greater poly-functional CD4(+) T cell responses than placebo and TIV. Poly-functional CD8(+) T cell responses were also observed after H1 VLP vaccination. Our results show that plant-made HA VLP vaccines elicit both strong antibody responses and poly-functional, cross-reactive memory T cells that persist for at least 6 months after vaccination.
Collapse
Affiliation(s)
- Nathalie Landry
- Medicago inc., 1020 Route de l'Église, Bureau 600, Québec, Qc G1V 3V9, Canada.
| | - Stéphane Pillet
- Medicago inc., 1020 Route de l'Église, Bureau 600, Québec, Qc G1V 3V9, Canada; Research Institute of the McGill University Health Center, Montreal General Hospital, 1650 Cedar Avenue, Montreal, Qc H3G 1A4, Canada.
| | - David Favre
- ImmuneCarta, 201 President-Kennedy, Suite PK-3900, Montreal, Qc H2X 3Y7, Canada.
| | - Jean-François Poulin
- ImmuneCarta, 201 President-Kennedy, Suite PK-3900, Montreal, Qc H2X 3Y7, Canada.
| | - Sonia Trépanier
- Medicago inc., 1020 Route de l'Église, Bureau 600, Québec, Qc G1V 3V9, Canada.
| | - Bader Yassine-Diab
- ImmuneCarta, 201 President-Kennedy, Suite PK-3900, Montreal, Qc H2X 3Y7, Canada.
| | - Brian J Ward
- Research Institute of the McGill University Health Center, Montreal General Hospital, 1650 Cedar Avenue, Montreal, Qc H3G 1A4, Canada.
| |
Collapse
|
26
|
Sanchez MV, Ebensen T, Schulze K, Cargnelutti D, Blazejewska P, Scodeller EA, Guzmán CA. Intranasal delivery of influenza rNP adjuvanted with c-di-AMP induces strong humoral and cellular immune responses and provides protection against virus challenge. PLoS One 2014; 9:e104824. [PMID: 25140692 PMCID: PMC4139298 DOI: 10.1371/journal.pone.0104824] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/17/2014] [Indexed: 12/17/2022] Open
Abstract
There is a critical need for new influenza vaccines able to protect against constantly emerging divergent virus strains. This will be sustained by the induction of vigorous cellular responses and humoral immunity capable of acting at the portal of entry of this pathogen. In this study we evaluate the protective efficacy of intranasal vaccination with recombinant influenza nucleoprotein (rNP) co-administrated with bis-(3′,5′)-cyclic dimeric adenosine monophosphate (c-di-AMP) as adjuvant. Immunization of BALB/c mice with two doses of the formulation stimulates high titers of NP-specific IgG in serum and secretory IgA at mucosal sites. This formulation also promotes a strong Th1 response characterized by high secretion of INF-γ and IL-2. The immune response elicited promotes efficient protection against virus challenge. These results suggest that c-di-AMP is a potent mucosal adjuvant which may significantly contribute towards the development of innovative mucosal vaccines against influenza.
Collapse
Affiliation(s)
- Maria Victoria Sanchez
- Laboratory of Virology, Institute of Experimental Medicine and Biology of Cuyo (IMBECU-CCT, CONICET), Mendoza, Argentina
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail:
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Diego Cargnelutti
- Laboratory of Virology, Institute of Experimental Medicine and Biology of Cuyo (IMBECU-CCT, CONICET), Mendoza, Argentina
| | - Paulina Blazejewska
- Boehringer Ingelheim Veterinary Research Center GmbH & Co. KG, Hannover, Germany
| | - Eduardo A. Scodeller
- Laboratory of Virology, Institute of Experimental Medicine and Biology of Cuyo (IMBECU-CCT, CONICET), Mendoza, Argentina
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
27
|
CD4 T cell help is limiting and selective during the primary B cell response to influenza virus infection. J Virol 2013; 88:314-24. [PMID: 24155379 DOI: 10.1128/jvi.02077-13] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Influenza virus vaccination strategies are focused upon the elicitation of protective antibody responses through administration of viral protein through either inactivated virions or live attenuated virus. Often overlooked in this strategy is the CD4 T cell response: how it develops into memory, and how it may support future primary B cell responses to heterologous infection. Through the utilization of a peptide-priming regimen, this study describes a strategy for developing CD4 T cell memory with the capacity to robustly expand in the lung-draining lymph node after live influenza virus infection. Not only were frequencies of antigen-specific CD4 T cells enhanced, but these cells also supported an accelerated primary B cell response to influenza virus-derived protein, evidenced by high anti-nucleoprotein (NP) serum antibody titers early, while there is still active viral replication ongoing in the lung. NP-specific antibody-secreting cells and heightened frequencies of germinal center B cells and follicular T helper cells were also readily detectable in the draining lymph node. Surprisingly, a boosted memory CD4 T cell response was not sufficient to provide intermolecular help for antibody responses. Our study demonstrates that CD4 T cell help is selective and limiting to the primary antibody response to influenza virus infection and that preemptive priming of CD4 T cell help can promote effective and rapid conversion of naive B cells to mature antibody-secreting cells.
Collapse
|