1
|
Hassanin O, Abdallah F, Mohamed MH, Ahmed MS, Al-Rasheed M, Rashad EM. The kinetics of gene expression related to innate and adaptive immunity in the lung and spleen following Newcastle disease virus (NDV) infection in vaccinated broiler chickens employing different vaccination regimes. Vet Microbiol 2025; 305:110525. [PMID: 40262238 DOI: 10.1016/j.vetmic.2025.110525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
Newcastle disease virus (NDV) classified as an avian paramyxovirus-1 poses a significant risk to the global poultry sector. This study explored the kinetics of the innate and adaptive immune responses in chickens exposed to virulent Newcastle Disease Virus (vNDV) genotype VII. A challenge experiment was carried out with both non-vaccinated and NDV-vaccinated chickens utilizing different vaccination strategies commonly used in the industry. The four groups of vaccinated birds were administered either two doses of live NDV vaccines, live vaccines boosted with an inactivated vaccine, the rHVT-NDV-IBDV vaccine alone, or the rHVT-NDV-IBDV vaccine in conjunction with a live vaccine booster. To assess seven cytokines linked to antiviral and proinflammatory innate responses quantitative real-time polymerase chain reaction (qRT-PCR) analyses were implemented. The analyses revealed robust innate immune responses in all cytokines measured in lung and spleen tissues of the group that received both live and inactivated vaccines. Notably these increases were found to correlate with the humoral immune response within that same group. Significant transcriptional activity in the lung and spleen tissues of non-vaccinated chickens at 24 hour post-infection (pi). The most significant upregulations were identified in toll-like receptor 7 (TLR7), TLR5, interleukin 6 (IL6), chicken interferon-alpha (chIFN-α), and myxovirus resistance protein 1 (Mx1), which later exhibited a regression. The administration of both live and inactivated vaccines has proven effective in restoring the suppression or inhibition of vNDV infection across a wide range of cytokines, including TLR7, TLR5, chIFN-α, Mx1, IL6, and MHC-1. Various vaccination strategies have been shown to either mitigate or prevent cytokine storms within 24 hour pi in the lungs, spleen, or both, in certain cytokines such as chIFN-α, Mx1, and IL6. Collectively these results suggest that different vaccination strategies modify the kinetics and pathophysiological responses associated with vNDV infection in chickens.
Collapse
Affiliation(s)
- Ola Hassanin
- Department of Avian and Rabbit Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| | - Fatma Abdallah
- Department of Virology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Mahmoud Ha Mohamed
- Department of Avian and Rabbit Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; Department of Clinical Studies, Collage of Veterinary Medicine, King Faisal University, 31982, Saudi Arabia
| | - Mohamed S Ahmed
- Department of Clinical Studies, Collage of Veterinary Medicine, King Faisal University, 31982, Saudi Arabia; Department of Poultry Diseases, Faculty of Veterinary Medicine, South Valley University, Egypt
| | - Mohammed Al-Rasheed
- Department of Clinical Studies, Collage of Veterinary Medicine, King Faisal University, 31982, Saudi Arabia
| | - Eman M Rashad
- Animal Health Research Institute (Domietta Branch), Agriculture Research Centre, Egypt
| |
Collapse
|
2
|
Zhu Z, Lou G, Luo Y, Yihunie K, Hoar J, Daniel JA, Evers BM, Yao C, Wu T. Aging Compromises Terminal Differentiation Program of Cytotoxic Effector Lineage and Promotes Exhaustion in CD8 + T Cells Responding to Coronavirus Infection. Aging Cell 2025:e70109. [PMID: 40396260 DOI: 10.1111/acel.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/16/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025] Open
Abstract
T cell aging increases the risk of viral infection-related morbidity and mortality and reduces vaccine efficacy in the elderly. A major hallmark of T cell aging is the loss of quiescence and shift toward terminal differentiation during homeostasis. However, how aging impacts the differentiation program of virus-specific T cells during infection is unclear. Here, in a murine coronavirus (MHV) infection model with age-associated increased mortality, we demonstrate that aging impairs, instead of promoting, the terminal differentiation program of virus-specific CD8+ T cells. Upon infection, CD8+ and CD4+ T cells in old mice showed marked reduction in clonal expansion and upregulation of immune checkpoints associated with T cell exhaustion. Bulk and single-cell transcriptomics showed that aging upregulated the T cell exhaustion transcriptional program associated with TOX in virus-specific CD8+ T cells and shifted the myeloid compartment from immunostimulatory to immunosuppressive phenotype. In addition, aging downregulated the transcriptional program of terminally differentiated effector CD8+ T cells and diminished the CX3CR1+ cytotoxic effector lineage. Mechanistically, virus-specific CD8+ T cells from infected aged mice displayed defects in inducing transcription factors ZEB2 and KLF2, which were required for terminal differentiation of effector CD8+ T cells. Together, our study shows that aging impairs terminal differentiation and promotes exhaustion of virus-specific CD8+ T cells responding to coronavirus infection through dysregulating expression of lineage-defining transcription factors.
Collapse
Affiliation(s)
- Ziang Zhu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Immunology Ph.D. Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guohua Lou
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ying Luo
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kiddist Yihunie
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Ph.D. Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan Hoar
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Juan A Daniel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chen Yao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tuoqi Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
3
|
Post CM, McDonough C, Lawrence BP. Binary and quaternary mixtures of perfluoroalkyl substances (PFAS) differentially affect the immune response to influenza A virus infection. J Immunotoxicol 2024; 21:2340495. [PMID: 38946256 PMCID: PMC11219007 DOI: 10.1080/1547691x.2024.2340495] [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: 01/05/2024] [Accepted: 04/03/2024] [Indexed: 07/02/2024] Open
Abstract
Per- and polyfluoroalkyl substances (PFAS) are anthropogenic organofluorine compounds that persist indefinitely in the environment and bioaccumulate throughout all trophic levels. Biomonitoring efforts have detected multiple PFAS in the serum of most people. Immune suppression has been among the most consistent effects of exposure to PFAS. PFAS often co-occur as mixtures in the environment, however, few studies have examined immunosuppression of PFAS mixtures or determined whether PFAS exposure affects immune function in the context of infection. In this study, mixtures containing two or four different PFAS and a mouse model of infection with influenza A virus (IAV) were used to assess immunotoxicity of PFAS mixtures. PFAS were administered via the drinking water as either a binary mixture of perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) or quaternary mixture of PFOS, PFOA, perfluorohexane sulfonate (PFHxS), and perfluorononanoic acid (PFNA). The results indicated that the binary mixture affected the T-cell response, while the quaternary mixture affected the B-cell response to infection. These findings indicate that the immunomodulatory effects of PFAS mixtures are not simply additive, and that the sensitivity of immune responses to PFAS varies by cell type and mixture. The study also demonstrates the importance of studying adverse health effects of PFAS mixtures.
Collapse
Affiliation(s)
- Christina M. Post
- University of Rochester School of Medicine & Dentistry, Rochester NY
| | | | - B. Paige Lawrence
- University of Rochester School of Medicine & Dentistry, Rochester NY
| |
Collapse
|
4
|
Hamza H, Ghosh M, Löffler MW, Rammensee HG, Planz O. Identification and relative abundance of naturally presented and cross-reactive influenza A virus MHC class I-restricted T cell epitopes. Emerg Microbes Infect 2024; 13:2306959. [PMID: 38240239 PMCID: PMC10854457 DOI: 10.1080/22221751.2024.2306959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/14/2024] [Indexed: 02/10/2024]
Abstract
Cytotoxic T lymphocytes are key for controlling viral infection. Unravelling CD8+ T cell-mediated immunity to distinct influenza virus strains and subtypes across prominent HLA types is relevant for combating seasonal infections and emerging new variants. Using an immunopeptidomics approach, naturally presented influenza A virus-derived ligands restricted to HLA-A*24:02, HLA-A*68:01, HLA-B*07:02, and HLA-B*51:01 molecules were identified. Functional characterization revealed multifunctional memory CD8+ T cell responses for nine out of sixteen peptides. Peptide presentation kinetics was optimal around 12 h post infection and presentation of immunodominant epitopes shortly after infection was not always persistent. Assessment of immunogenic epitopes revealed that they are highly conserved across the major zoonotic reservoirs and may contain a single substitution in the vicinity of the anchor residues. These findings demonstrate how the identified epitopes promote T cell pools, possibly cross-protective in individuals and can be potential targets for vaccination.
Collapse
Affiliation(s)
- Hazem Hamza
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Virology Laboratory, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Michael Ghosh
- Institute for Immunology, University of Tübingen, Tübingen, Germany
| | - Markus W Löffler
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, Tübingen, Germany
- Centre for Clinical Transfusion Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Cluster of Excellence CMFI (EXC2124) "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Oliver Planz
- Institute for Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
Ballow M, Ortiz-de-Lejarazu R, Quinti I, Miller MS, Warnatz K. Contribution of immunoglobulin products in influencing seasonal influenza infection and severity in antibody immune deficiency patients receiving immunoglobulin replacement therapy. Front Immunol 2024; 15:1452106. [PMID: 39502688 PMCID: PMC11534824 DOI: 10.3389/fimmu.2024.1452106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Seasonal and pandemic influenza infection present a potential threat to patients with antibody deficiency. The acceptance and effect of the current recommendation for annual vaccination against influenza for patients with antibody deficiency is not well investigated and due to antigenic drift or shift the protective capacity of regular IgG replacement therapy (IgRT) is considered low. This narrative review considers the effect of influenza vaccination in immunodeficient patients and discusses available information on the effect of immunoglobulin products on seasonal influenza infectivity and severity in antibody deficiency patients receiving IgRT. The humoral immune response to seasonal influenza vaccination is reduced in patients with antibody immune deficiency. However, there is no evidence that the proportion of patients with primary antibody deficiency who develop influenza illness, and the severity of such illness, is increased when compared with the general population. The IgRT that patients receive has been shown to contain neutralizing antibodies as a consequence of past flu infections against both the hemagglutinin and neuraminidase surface proteins and other viral internal proteins of different influenza A virus strains. Studies have demonstrated not only significant levels of specific but also cross-reactive antibodies against seasonal influenza virus strains. Thus, despite the yearly changes in influenza viral antigenicity that occur, IgRT could potentially contribute to the protection of patients against seasonal influenza. Currently, only limited clinical data are available confirming a preventative effect of IgRT with respect to seasonal influenza infection. In conclusion, there is some evidence that IgRT could contribute to protection against seasonal influenza in patients with antibody-related immunodeficiency. However, additional clinical data are needed to confirm the extent and relevance of this protection and identify the main responsible virus targets of that protection.
Collapse
Affiliation(s)
- Mark Ballow
- Division of Allergy & Immunology, Morsani College of Medicine, University of South Florida, at Johns Hopkins All Children’s Hospital, St. Petersburg Florida, FL, United States
| | - Raúl Ortiz-de-Lejarazu
- Professor of Microbiology, Scientific Advisor & Emeritus Director, National Influenza Center, Valladolid, Spain
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Head of the Primary Immunodeficiency Unit, Rome, Italy
| | - Matthew S. Miller
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center – University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Freiburg, Germany
- Department of Clinical Immunology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Zhang T, Tian Y, Zhang X, Wang W, He Y, Ge C, Jia F, Wang Z, Jiang Y. Improved cellular immune response induced by intranasal boost immunization with chitosan coated DNA vaccine against H9N2 influenza virus challenge. Microb Pathog 2024; 195:106871. [PMID: 39163919 DOI: 10.1016/j.micpath.2024.106871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
The H9N2 avian influenza virus (AIV) is spreading worldwide. Presence of H9N2 virus tends to increase the chances of infection with other pathogens which can lead to more serious economic losses. In a previous study, a regulated delayed lysis Salmonella vector was used to deliver a DNA vaccine named pYL233 encoding M1 protein, mosaic HA protein and chicken GM-CSF adjuvant. To further increase its efficiency, chitosan as a natural adjuvant was applied in this study. The purified plasmid pYL233 was coated with chitosan to form a DNA containing nanoparticles (named CS233) by ionic gel method and immunized by intranasal boost immunization in birds primed by oral administration with Salmonella strain. The CS233 DNA nanoparticle has a particle size of about 150 nm, with an encapsulation efficiency of 93.2 ± 0.12 % which protected the DNA plasmid from DNase I digestion and could be stable for a period of time at 37°. After intranasal boost immunization, the CS233 immunized chickens elicited higher antibody response, elevated CD4+ T cells and CD8+ T cells activation and increased T-lymphocyte proliferation, as well as increased productions of IL-4 and IFN-γ. After challenge, chickens immunized with CS233 resulted in the lowest levels of pulmonary virus titer and viral shedding as compared to the other challenge groups. The results showed that the combination of intranasal immunization with chitosan-coated DNA vaccine and oral immunization with regulatory delayed lytic Salmonella strain could enhance the immune response and able to provide protection against H9N2 challenge.
Collapse
MESH Headings
- Animals
- Influenza A Virus, H9N2 Subtype/immunology
- Influenza A Virus, H9N2 Subtype/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Chitosan
- Administration, Intranasal
- Influenza in Birds/prevention & control
- Influenza in Birds/immunology
- Chickens/immunology
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Antibodies, Viral/blood
- Immunity, Cellular
- Virus Shedding
- Plasmids/genetics
- Nanoparticles
- Immunization, Secondary
- CD8-Positive T-Lymphocytes/immunology
- Adjuvants, Immunologic/administration & dosage
- Interferon-gamma
- Interleukin-4
- Adjuvants, Vaccine
- Poultry Diseases/prevention & control
- Poultry Diseases/immunology
- Poultry Diseases/virology
- CD4-Positive T-Lymphocytes/immunology
- Salmonella/immunology
- Salmonella/genetics
Collapse
Affiliation(s)
- Tongyu Zhang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yawen Tian
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Xiao Zhang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Wenfeng Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yingkai He
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Chongbo Ge
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Futing Jia
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Zhannan Wang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Yanlong Jiang
- College of Animal Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
7
|
Leddy O, Yuki Y, Carrington M, Bryson BD, White FM. PathMHC: a workflow to selectively target pathogen-derived MHC peptides in discovery immunopeptidomics experiments for vaccine target identification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612454. [PMID: 39314426 PMCID: PMC11419027 DOI: 10.1101/2024.09.11.612454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Vaccine-elicited T cell responses can contribute to immune protection against emerging infectious disease risks such as antimicrobials-resistant (AMR) microbial pathogens and viruses with pandemic potential, but rapidly identifying appropriate targets for T cell priming vaccines remains challenging. Mass spectrometry (MS) analysis of peptides presented on major histocompatibility complexes (MHCs) can identify potential targets for protective T cell responses in a proteome-wide manner. However, pathogen-derived peptides are outnumbered by self peptides in the MHC repertoire and may be missed in untargeted MS analyses. Here we present a novel approach, termed PathMHC, that uses computational analysis of untargeted MS data followed by targeted MS to discover novel pathogen-derived MHC peptides more efficiently than untargeted methods alone. We applied this workflow to identify MHC peptides derived from multiple microbes, including potential vaccine targets presented on MHC-I by human dendritic cells infected with Mycobacterium tuberculosis . PathMHC will facilitate antigen discovery campaigns for vaccine development.
Collapse
|
8
|
Jang S, Hong W, Moon Y. Obesity-compromised immunity in post-COVID-19 condition: a critical control point of chronicity. Front Immunol 2024; 15:1433531. [PMID: 39188722 PMCID: PMC11345197 DOI: 10.3389/fimmu.2024.1433531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Post-COVID-19 condition is recognized as a multifactorial disorder, with persistent presence of viral antigens, discordant immunity, delayed viral clearance, and chronic inflammation. Obesity has emerged as an independent risk factor for both SARS-CoV-2 infection and its subsequent sequelae. In this study, we aimed to predict the molecular mechanisms linking obesity and post-COVID-19 distress. Viral antigen-exposed adipose tissues display remarkable levels of viral receptors, facilitating viral entry, deposition, and chronic release of inflammatory mediators and cells in patients. Subsequently, obesity-associated inflammatory insults are predicted to disturb cellular and humoral immunity by triggering abnormal cell differentiation and lymphocyte exhaustion. In particular, the decline in SARS-CoV-2 antibody titers and T-cell exhaustion due to chronic inflammation may account for delayed virus clearance and persistent activation of inflammatory responses. Taken together, obesity-associated defective immunity is a critical control point of intervention against post-COVID-19 progression, particularly in subjects with chronic metabolic distress.
Collapse
Affiliation(s)
- Soonwoo Jang
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
| | - Wooyoung Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
9
|
Wang X, Geng Z, Bao Y, Zhong J, Ma J, Cui X, Shi Y. Shufeng Jiedu capsule alleviates influenza A (H1N1) virus induced acute lung injury by regulating the lung inflammatory microenvironment. Heliyon 2024; 10:e33237. [PMID: 39021925 PMCID: PMC11252743 DOI: 10.1016/j.heliyon.2024.e33237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/13/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Background Death caused by respiratory tract infection is one of the leading causes of death in the world today. Shufeng Jiedu Capsule (SFJDC) is a traditional Chinese medicine that has been widely used clinically for coronavirus disease 2019 (COVID-19), H1N1 influenza virus pneumonia and other diseases. Its pharmacological effect is to inhibit inflammation and improve the body's ability to clear viruses. However, the mechanism of SFJDC in the treatment of viral pneumonia, especially its effect on the inflammatory-immune microenvironment of lung tissue remains unclear. Methods Mice with H1N1 influenza virus pneumonia were used as a model to verify the efficacy of SFJDC through death protection, lung index, viral load, and HE staining of lung tissue. The levels of inflammatory cytokines and chemokines in lung tissue were investigated by multi-analyte immunoassay. The number and proportion of cells in peripheral blood were detected by blood routine. The percentage of infiltrating immune cells in lung tissue was detected by flow cytometry and immunofluorescence. Results SFJDC (2.2 g/kg·d-1 and 1.1 g/kg·d-1) increased survival rate (P<0.01, P<0.05), prolonged the survival period of mice, and alleviated the histopathological damage in lung (P<0.01). SFJDC (2.2 g/kg·d-1, 1.1 g/kg·d-1 and 0.055 g/kg·d-1) increased body weight(P<0.01, P<0.05), improved activity status, reduced the lung index (P<0.01, P<0.05) and viral load (P<0.01). SFJDC (2.2 g/kg·d-1 and 1.1 g/kg·d-1) reduced interleukin-1β (IL-1β), interleukin-18(IL-18), tumour necrosis factor α (TNF-α), monocyte chemoattractant protein (MCP), chemokine (C-X-C motif) ligand 1 (CXCL1) (P<0.01, P<0.05), and SFJDC (2.2 g/kg·d-1) increased IL-10 levels (P<0.05) to regulate inflammation. SFJDC (2.2 g/kg·d-1) increased the percentages of CD4+ T cells (P<0.01), CD8+ T cells (P<0.05), and B cells(P<0.05), and decreased F4/80+ macrophages (P<0.05). Conclusion Our findings indicated that SFJDC could inhibit inflammation and lung injury while maintaining the function of the adaptive immune response mediated by T and B cells, and promote the clearance of the virus, thereby treating influenza A (H1N1) virus-induced pneumonia.
Collapse
Affiliation(s)
- Xiaorui Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Zihan Geng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Yanyan Bao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Juying Zhong
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Jing Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Xiaolan Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| | - Yujing Shi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100025, China
| |
Collapse
|
10
|
Finn CM, McKinstry KK. Ex Pluribus Unum: The CD4 T Cell Response against Influenza A Virus. Cells 2024; 13:639. [PMID: 38607077 PMCID: PMC11012043 DOI: 10.3390/cells13070639] [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: 02/26/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Current Influenza A virus (IAV) vaccines, which primarily aim to generate neutralizing antibodies against the major surface proteins of specific IAV strains predicted to circulate during the annual 'flu' season, are suboptimal and are characterized by relatively low annual vaccine efficacy. One approach to improve protection is for vaccines to also target the priming of virus-specific T cells that can protect against IAV even in the absence of preexisting neutralizing antibodies. CD4 T cells represent a particularly attractive target as they help to promote responses by other innate and adaptive lymphocyte populations and can also directly mediate potent effector functions. Studies in murine models of IAV infection have been instrumental in moving this goal forward. Here, we will review these findings, focusing on distinct subsets of CD4 T cell effectors that have been shown to impact outcomes. This body of work suggests that a major challenge for next-generation vaccines will be to prime a CD4 T cell population with the same spectrum of functional diversity generated by IAV infection. This goal is encapsulated well by the motto 'ex pluribus unum': that an optimal CD4 T cell response comprises many individual specialized subsets responding together.
Collapse
Affiliation(s)
| | - K. Kai McKinstry
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
11
|
Mao J, Eom GD, Yoon KW, Kim MJ, Chu KB, Kang HJ, Quan FS. Crossprotection induced by virus-like particles containing influenza dual-hemagglutinin and M2 ectodomain. Nanomedicine (Lond) 2024; 19:741-754. [PMID: 38390688 DOI: 10.2217/nnm-2023-0353] [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] [Indexed: 02/24/2024] Open
Abstract
Aims: To develop an effective universal vaccine against antigenically different influenza viruses. Materials & methods: We generated influenza virus-like particles (VLPs) expressing the H1 and H3 antigens with or without M2e5x. VLP-induced immune responses and crossprotection against H1N1, H3N2 or H5N1 viruses were assessed to evaluate their protective efficacy. Results: H1H3M2e5x immunization elicited higher crossreactive IgG antibodies than H1H3 VLPs. Upon challenge, both VLPs enhanced lung IgG, IgA and germinal center B-cell responses compared with control. While these VLPs conferred protection, H1H3M2e5x showed greater lung viral load reduction than H1H3 VLPs with minimal body weight loss. Conclusion: Utilizing VLPs containing dual-hemagglutinin, along with M2e5x, can be a vaccination strategy for inducing crossprotection against influenza A viruses.
Collapse
Affiliation(s)
- Jie Mao
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Gi-Deok Eom
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hae-Ji Kang
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA 30303, USA
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
12
|
Tian J, Fu W, Xie Z, Wang X, Miao M, Shan F, Yu X. Methionine enkephalin(MENK) upregulated memory T cells in anti-influenza response. BMC Immunol 2023; 24:38. [PMID: 37828468 PMCID: PMC10571428 DOI: 10.1186/s12865-023-00573-0] [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: 03/30/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
Novel prophylactic drugs and vaccination strategies for protection against influenza virus should induce specific effector T-cell immune responses in pulmonary airways and peripheral lymphoid organs. Designing approaches that promote T-cell-mediated responses and memory T-cell differentiation would strengthen host resistance to respiratory infectious diseases. The results of this study showed that pulmonary delivery of MENK via intranasal administration reduced viral titres, upregulated opioid receptor MOR and DOR, increased the proportions of T-cell subsets including CD8+ T cells, CD8+ TEM cells, NP/PA-effector CD8+ TEM cells in bronchoalveolar lavage fluid and lungs, and CD4+/CD8+ TCM cells in lymph nodes to protect mice against influenza viral challenge. Furthermore, we demonstrated that, on the 10th day of infection, the proportions of CD4+ TM and CD8+ TM cells were significantly increased, which meant that a stable TCM and TEM lineage was established in the early stage of influenza infection. Collectively, our data suggested that MENK administered intranasally, similar to the route of natural infection by influenza A virus, could exert antiviral activity through upregulating T-cell-mediated adaptive immune responses against influenza virus.
Collapse
Affiliation(s)
- Jing Tian
- Department of Immunology, School of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, China
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, 110122, China
| | - Wenrui Fu
- Department of Immunology, School of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, China
| | - Zifeng Xie
- Department of Immunology, School of Basic Medical Science, Jinzhou Medical University, Jinzhou, 121001, China
| | - Xiaonan Wang
- Biostax Inc., 1317 Edgewater Dr., Ste 4882, Orlando, FL, 32804, USA
| | - Miao Miao
- Biostax Inc., 1317 Edgewater Dr., Ste 4882, Orlando, FL, 32804, USA
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, 110122, China.
| | - Xiaodong Yu
- Department of Nursing, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
13
|
Tan Y, Wang Y, Souza-Moreira L, Wang C, Murray ABP, Salkhordeh M, Florian M, McIntyre L, Stewart DJ, Mei SHJ. Mesenchymal stem cells induce dynamic immunomodulation of airway and systemic immune cells in vivo but do not improve survival for mice with H1N1 virus-induced acute lung injury. Front Bioeng Biotechnol 2023; 11:1203387. [PMID: 37362215 PMCID: PMC10285296 DOI: 10.3389/fbioe.2023.1203387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction: Influenza A virus (IAV)-induced acute lung injury (ALI) is characterized by pronounced proinflammatory activation and respiratory lung dysfunction. In this study, we performed deep immune profiling on airway and circulating immune cells to examine the effect of immunomodulation and therapeutic outcomes of mesenchymal stem cells (MSCs) therapy in mice with IAV-induced ALI. Methods: Animals were inoculated intranasally with H1N1 IAV, followed by intravenous administration of vehicle, or human clinical-grade, bone marrow-derived MSCs 24-h later, and monitored for six days to evaluate the survival. In another set of animals, bronchoalveolar lavage (BAL) fluid and whole blood were collected three days after infection for flow or mass cytometry (CyTOF) immune profiling analysis. Results: Immune cell population and phenotypic shifts in blood were mapped by CyTOF. Increases were observed in granulocytes and myeloid-derived cells in blood from vehicle-treated animals. While MSC treatment accentuated changes in these populations, naïve B, antibody-secreting B cells, and T cells were decreased in MSC-treated animals at day 3. Compared to sham animals, IAV infection induced a significant 5.5-fold increase in BAL total cell counts, including CD4+ and CD8+ T cells, CD19+ B cells, CD11b + Ly6G + neutrophils, and CD11b + Ly6C + monocytes. MSC treatment significantly decreased BAL total cell counts in IAV-infected mice, specifically the number of infiltrating CD4+ T cells and CD11b + Ly6G + neutrophils. In contrast, there were increases in CD8+ T cells, B cells, and monocytes in the alveolar space in MSC-treated animals. Phenotypic immune cell profiling of blood and BAL revealed a significantly higher proportion of the monocyte population with the M2 phenotype (CD206) in MSC-treated animals; however, this failed to confer protective effects in the survival of infected mice or reduce viral titer in the lung. Further investigation revealed that MSCs were susceptible to IAV infection, leading to increased cell death and potentially affecting their efficacy. Conclusion: These findings provided in vivo evidence that MSCs promote the selective recruitment of immune cells to the site of infection during IAV infection, with reductions in proinflammatory phenotypes. However, MSCs offered no survival benefit in IAV-infected animals, possibly due to MSCs' H1N1 IAV susceptibility and subsequent cell death.
Collapse
Affiliation(s)
- Yuan Tan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Yan Wang
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Luciana Souza-Moreira
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Chi Wang
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Aidan B. P. Murray
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mahmoud Salkhordeh
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Maria Florian
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Lauralyn McIntyre
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Duncan J. Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Shirley H. J. Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
14
|
Cui X, Guo Y, Liu Q. Qingfei Jiedu Granules fight influenza by regulating inflammation, immunity, metabolism, and gut microbiota. J Tradit Complement Med 2023; 13:170-182. [PMID: 36970461 PMCID: PMC10037062 DOI: 10.1016/j.jtcme.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 10/14/2022] Open
Abstract
Background and aim Qingfei Jiedu Granules (QFJD) are a new Traditional Chinese Medicine (TCM) which has been clinically used against coronavirus pneumonia in China. In this study, the therapeutic effect and the underlying mechanisms of QFJD against influenza were investigated. Experimental procedure Pneumonia mice were induced by influenza A virus. Survival rate, weight loss, lung index and lung pathology were measured to evaluate the therapeutic effect of QFJD. The expression of inflammatory factors and lymphocytes was used to assess anti-inflammatory and immunomodulatory effect of QFJD. Gut microbiome analysis was performed to decipher the potential effect of QFJD on intestinal microbiota. Metabolomics approach was conducted to explore the overall metabolic regulation of QFJD. Result and conclusion QFJD shows a significant therapeutic effect on the treatment of influenza and the expression of many pro-inflammatory cytokines were obviously inhibited. QFJD also markedly modulates the level of T and B lymphocytes. The high-dose QFJD has shown similar therapeutic efficiency compared to positive drugs. QFJD profoundly enriched Verrucomicrobia and maintained the balance between Bacteroides and Firmicutes. QFJD associated with 12 signaling pathways in metabolomics study, 9 of which were the same as the model group and were closely related to citrate cycle and amino acid metabolism.To sum up, QFJD is a novel and promising drug against influenza. It can regulate inflammation, immunity, metabolism, and gut microbiota to fight influenza. Verrucomicrobia shows great potential to improve influenza infection and may be an important target.
Collapse
Affiliation(s)
- Xuran Cui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, 100010, China
| | - Yuhong Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, 100010, China
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
- Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, 100010, China
| |
Collapse
|
15
|
Post CM, Myers JR, Winans B, Lawrence BP. Postnatal administration of S-adenosylmethionine restores developmental AHR activation-induced deficits in CD8+ T cell function during influenza A virus infection. Toxicol Sci 2023; 192:kfad019. [PMID: 36847456 PMCID: PMC10109536 DOI: 10.1093/toxsci/kfad019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Developmental exposures can influence life-long health; yet, counteracting negative consequences is challenging due to poor understanding of cellular mechanisms. The aryl hydrocarbon receptor (AHR) binds many small molecules, including numerous pollutants. Developmental exposure to the signature environmental AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) significantly dampens adaptive immune responses to influenza A virus (IAV) in adult offspring. CD8+ cytotoxic T lymphocytes (CTL) are crucial for successful infection resolution, which depends on the number generated and the complexity of their functionality. Prior studies showed developmental AHR activation significantly reduced the number of virus-specific CD8+ T cells, but impact on their functions is less clear. Other studies showed developmental exposure was associated with differences in DNA methylation in CD8+ T cells. Yet, empirical evidence that differences in DNA methylation are causally related to altered CD8+ T cell function is lacking. The two objectives were to ascertain whether developmental AHR activation affects CTL function, and whether differences in methylation contribute to reduced CD8+ T cell responses to infection. Developmental AHR triggering significantly reduced CTL polyfunctionality, and modified the transcriptional program of CD8+ T cells. S-adenosylmethionine (SAM), which increases DNA methylation, but not Zebularine, which diminishes DNA methylation, restored polyfunctionality and boosted the number of virus-specific CD8+ T cells. These findings suggest that diminished methylation, initiated by developmental exposure to an AHR-binding chemical, contributes to durable changes in antiviral CD8+ CTL functions later in life. Thus, deleterious consequence of development exposure to environmental chemicals are not permanently fixed, opening the door for interventional strategies to improve health.
Collapse
Affiliation(s)
- Christina M Post
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jason R Myers
- Genomics Research Center, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Bethany Winans
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
16
|
Escudero-Pérez B, Lawrence P, Castillo-Olivares J. Immune correlates of protection for SARS-CoV-2, Ebola and Nipah virus infection. Front Immunol 2023; 14:1156758. [PMID: 37153606 PMCID: PMC10158532 DOI: 10.3389/fimmu.2023.1156758] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Correlates of protection (CoP) are biological parameters that predict a certain level of protection against an infectious disease. Well-established correlates of protection facilitate the development and licensing of vaccines by assessing protective efficacy without the need to expose clinical trial participants to the infectious agent against which the vaccine aims to protect. Despite the fact that viruses have many features in common, correlates of protection can vary considerably amongst the same virus family and even amongst a same virus depending on the infection phase that is under consideration. Moreover, the complex interplay between the various immune cell populations that interact during infection and the high degree of genetic variation of certain pathogens, renders the identification of immune correlates of protection difficult. Some emerging and re-emerging viruses of high consequence for public health such as SARS-CoV-2, Nipah virus (NiV) and Ebola virus (EBOV) are especially challenging with regards to the identification of CoP since these pathogens have been shown to dysregulate the immune response during infection. Whereas, virus neutralising antibodies and polyfunctional T-cell responses have been shown to correlate with certain levels of protection against SARS-CoV-2, EBOV and NiV, other effector mechanisms of immunity play important roles in shaping the immune response against these pathogens, which in turn might serve as alternative correlates of protection. This review describes the different components of the adaptive and innate immune system that are activated during SARS-CoV-2, EBOV and NiV infections and that may contribute to protection and virus clearance. Overall, we highlight the immune signatures that are associated with protection against these pathogens in humans and could be used as CoP.
Collapse
Affiliation(s)
- Beatriz Escudero-Pérez
- WHO Collaborating Centre for Arbovirus and Haemorrhagic Fever Reference and Research, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Braunschweig, Germany
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| | - Philip Lawrence
- CONFLUENCE: Sciences et Humanités (EA 1598), Université Catholique de Lyon (UCLy), Lyon, France
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| |
Collapse
|
17
|
Influenza Virus Infection during Pregnancy as a Trigger of Acute and Chronic Complications. Viruses 2022; 14:v14122729. [PMID: 36560733 PMCID: PMC9786233 DOI: 10.3390/v14122729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Influenza A virus (IAV) infection during pregnancy disrupts maternal and fetal health through biological mechanisms, which are to date poorly characterised. During pregnancy, the viral clearance mechanisms from the lung are sub-optimal and involve hyperactive innate and adaptive immune responses that generate wide-spread inflammation. Pregnancy-related adaptations of the immune and the cardiovascular systems appear to result in delayed recovery post-viral infection, which in turn promotes a prolonged inflammatory phenotype, increasing disease severity, and causing maternal and fetal health problems. This has immediate and long-term consequences for the mother and fetus, with complications including acute cardiopulmonary distress syndrome in the mother that lead to perinatal complications such as intrauterine growth restriction (IUGR), and birth defects; cleft lip, cleft palate, neural tube defects and congenital heart defects. In addition, an increased risk of long-term neurological disorders including schizophrenia in the offspring is reported. In this review we discuss the pathophysiology of IAV infection during pregnancy and its striking similarity to other well-established complications of pregnancy such as preeclampsia. We discuss general features of vascular disease with a focus on vascular inflammation and define the "Vascular Storm" that is triggered by influenza infection during pregnancy, as a pivotal disease mechanism for short and long term cardiovascular complications.
Collapse
|
18
|
Wang YH, Noyer L, Kahlfuss S, Raphael D, Tao AY, Kaufmann U, Zhu J, Mitchell-Flack M, Sidhu I, Zhou F, Vaeth M, Thomas PG, Saunders SP, Stauderman K, Curotto de Lafaille MA, Feske S. Distinct roles of ORAI1 in T cell-mediated allergic airway inflammation and immunity to influenza A virus infection. SCIENCE ADVANCES 2022; 8:eabn6552. [PMID: 36206339 PMCID: PMC9544339 DOI: 10.1126/sciadv.abn6552] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 08/22/2022] [Indexed: 06/16/2023]
Abstract
T cell activation and function depend on Ca2+ signals mediated by store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI1 proteins. We here investigated how SOCE controls T cell function in pulmonary inflammation during a T helper 1 (TH1) cell-mediated response to influenza A virus (IAV) infection and TH2 cell-mediated allergic airway inflammation. T cell-specific deletion of Orai1 did not exacerbate pulmonary inflammation and viral burdens following IAV infection but protected mice from house dust mite-induced allergic airway inflammation. ORAI1 controlled the expression of genes including p53 and E2F transcription factors that regulate the cell cycle in TH2 cells in response to allergen stimulation and the expression of transcription factors and cytokines that regulate TH2 cell function. Systemic application of a CRAC channel blocker suppressed allergic airway inflammation without compromising immunity to IAV infection, suggesting that inhibition of SOCE is a potential treatment for allergic airway disease.
Collapse
Affiliation(s)
- Yin-Hu Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Lucile Noyer
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sascha Kahlfuss
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Dimitrius Raphael
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony Y. Tao
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ulrike Kaufmann
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jingjie Zhu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Marisa Mitchell-Flack
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ikjot Sidhu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Fang Zhou
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Martin Vaeth
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Paul G. Thomas
- St. Jude’s Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sean P. Saunders
- Division of Pulmonary, Critical Care and Sleep Medicine, Departments of Medicine and Cell Biology, New York University Grossman School of Medicine, NY 10016, USA
| | | | - Maria A. Curotto de Lafaille
- Division of Pulmonary, Critical Care and Sleep Medicine, Departments of Medicine and Cell Biology, New York University Grossman School of Medicine, NY 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
19
|
Zhou X, Ye G, Lv Y, Guo Y, Pan X, Li Y, Shen G, He Y, Lei P. IL-6 drives T cell death to participate in lymphopenia in COVID-19. Int Immunopharmacol 2022; 111:109132. [PMID: 35964413 PMCID: PMC9359506 DOI: 10.1016/j.intimp.2022.109132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 01/08/2023]
Abstract
Lymphopenia is a common observation in patients with COVID-19. To explore the cause of T cell lymphopenia in the disease, laboratory results of 64 hospitalized COVID-19 patients were retrospectively analyzed and six patients were randomly selected to trace their changes of T lymphocytes and plasma concentration of IL-6 for the course of disease. Results confirmed that the T-cell lymphopenia, especially CD4+ T cell reduction in COVID-19 patients, was a reliable indicator of severity and hospitalization in infected patients. And CD4+ T cell count below 200 cells/μL predicts critical illness in COVID-19 patients. In vitro assay supported that exposure to key contributors (IL-1β, IL-6, TNF-α and IFN-γ) of COVID-19 cytokine storm caused substantial death of activated T cells. Among these contributors, IL-6 level was found to probably reversely correlate with T cell counts in patients. And IL-6 alone was potent to induce T cell reduction by gasderminE-mediated pyroptosis, inferring IL-6 took a part in affecting the function and status of T cells in COVID-19 patients. Intervention of IL-6 mediated T cell pryprotosis may effectively delay disease progression, maintain normal immune status at an early stage of infection.
Collapse
Affiliation(s)
- Xiaoqi Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Nuclear Medicine and PET Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guangming Ye
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yibing Lv
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanyan Guo
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingfei Pan
- Department of Infectious Diseases, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yirong Li
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong He
- Department of Nuclear Medicine and PET Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Mao J, Eom GD, Yoon KW, Kang HJ, Chu KB, Quan FS. Sublingual Vaccination with Live Influenza Virus Induces Better Protection Than Oral Immunization in Mice. Life (Basel) 2022; 12:life12070975. [PMID: 35888065 PMCID: PMC9321673 DOI: 10.3390/life12070975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
Both sublingual (SL) and oral vaccine administration modalities are convenient, easy, and safe. Here, we have investigated the differences in vaccine efficacy that are induced by oral and sublingual immunization with live influenza virus (A/Hong Kong/1/1968, H3N2) in mice. Intranasally administering a lethal dose of the influenza virus resulted in the deaths of the mice, whereas viral replication in the lungs did not occur upon SL or oral administration. At 30 days post-immunization through the SL or oral route, the mice were intranasally challenge-infected with the lethal dose of the homologous influenza virus. Both SL and oral immunizations with the influenza virus elicited significantly higher levels of virus-specific IgG and IgA antibody responses, as well as HAI titers in the sera. Upon challenge infection, the SL immunization elicited higher levels of pulmonary IgG antibody and CD8+ T cell responses than the oral immunization. Enhanced splenic germinal center B (GC B) and B cell proliferation were also detected from the SL immunization, both of which were significantly greater than those of the oral immunization. Importantly, compared to oral immunization, significantly lessened lung viral loads and bodyweight reductions were observed from the SL immunization and these parameters contributed to prolonging the survival of the immunized mice. These results indicate that both SL and oral administration could be effective routes in inducing protective immunity against influenza virus infection, with SL immunization being the better of the two delivery routes.
Collapse
Affiliation(s)
- Jie Mao
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.M.); (G.-D.E.); (K.-W.Y.); (H.-J.K.)
| | - Gi-Deok Eom
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.M.); (G.-D.E.); (K.-W.Y.); (H.-J.K.)
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.M.); (G.-D.E.); (K.-W.Y.); (H.-J.K.)
| | - Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.M.); (G.-D.E.); (K.-W.Y.); (H.-J.K.)
| | - Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea;
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
21
|
Tsybalova LM, Stepanova LA, Ramsay ES, Vasin AV. Influenza B: Prospects for the Development of Cross-Protective Vaccines. Viruses 2022; 14:1323. [PMID: 35746794 PMCID: PMC9228933 DOI: 10.3390/v14061323] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 01/04/2023] Open
Abstract
In this review, we analyze the epidemiological and ecological features of influenza B, one of the most common and severe respiratory infections. The review presents various strategies for cross-protective influenza B vaccine development, including recombinant viruses, virus-like particles, and recombinant proteins. We provide an overview of viral proteins as cross-protective vaccine targets, along with other updated broadly protective vaccine strategies. The importance of developing such vaccines lies not only in influenza B prevention, but also in the very attractive prospect of eradicating the influenza B virus in the human population.
Collapse
Affiliation(s)
- Liudmila M. Tsybalova
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
| | - Liudmila A. Stepanova
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
| | - Edward S. Ramsay
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
| | - Andrey V. Vasin
- Smorodintsev Research Institute of Influenza, Prof. Popova Str., 15/17, 197376 St. Petersburg, Russia; (L.A.S.); (E.S.R.); or (A.V.V.)
- Research Institute of Influenza named after A.A. Smorodintsev, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251 St. Petersburg, Russia
| |
Collapse
|
22
|
Martins de Camargo M, Caetano AR, Ferreira de Miranda Santos IK. Evolutionary pressures rendered by animal husbandry practices for avian influenza viruses to adapt to humans. iScience 2022; 25:104005. [PMID: 35313691 PMCID: PMC8933668 DOI: 10.1016/j.isci.2022.104005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Commercial poultry operations produce and crowd billions of birds every year, which is a source of inexpensive animal protein. Commercial poultry is intensely bred for desirable production traits, and currently presents very low variability at the major histocompatibility complex. This situation dampens the advantages conferred by the MHC’s high genetic variability, and crowding generates immunosuppressive stress. We address the proteins of influenza A viruses directly and indirectly involved in host specificities. We discuss how mutants with increased virulence and/or altered host specificity may arise if few class I alleles are the sole selective pressure on avian viruses circulating in immunocompromised poultry. This hypothesis is testable with peptidomics of MHC ligands. Breeding strategies for commercial poultry can easily and inexpensively include high variability of MHC as a trait of interest, to help save billions of dollars as a disease burden caused by influenza and decrease the risk of selecting highly virulent strains.
Collapse
|
23
|
Dong C, Wang BZ. Engineered Nanoparticulate Vaccines to Combat Recurring and Pandemic Influenza Threats. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100122. [PMID: 35754779 PMCID: PMC9231845 DOI: 10.1002/anbr.202100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Reoccurring seasonal flu epidemics and occasional pandemics are among the most severe threats to public health. Current seasonal influenza vaccines provide limited protection against drifted circulating strains and no protection against influenza pandemics. Next-generation influenza vaccines, designated as universal influenza vaccines, should be safe, affordable, and elicit long-lasting cross-protective influenza immunity. Nanotechnology plays a critical role in the development of such novel vaccines. Engineered nanoparticles can incorporate multiple advantageous properties into the same nanoparticulate platforms to improve vaccine potency and breadth. These immunological properties include virus-like biomimicry, high antigen-load, controlled antigen release, targeted delivery, and induction of innate signaling pathways. Many nanoparticle influenza vaccines have shown promising results in generating potent and broadly protective immune responses. This review will summarize the necessity and characteristics of next-generation influenza vaccines and the immunological correlates of broad influenza immunity and focus on how cutting-edge nanoparticle technology contributes to such vaccine development. The review will give new insights into the rational design of nanoparticle universal vaccines to combat influenza epidemics and pandemics.
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia 30303, USA
| |
Collapse
|
24
|
Viral PB1-F2 and host IFN-γ guide ILC2 and T cell activity during influenza virus infection. Proc Natl Acad Sci U S A 2022; 119:2118535119. [PMID: 35169077 PMCID: PMC8872759 DOI: 10.1073/pnas.2118535119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2022] [Indexed: 12/28/2022] Open
Abstract
The regulation of functional immune cell plasticity is poorly understood. Host environmental cues are critical, but the possible influence of pathogen-derived virulence factors has not been described. We have used reverse-engineered influenza A viruses that differ in PB1-F2 activity to analyze influenza in mice in the presence or absence of host interferon (IFN)-γ. In the absence of functional PB1-F2 and IFN-γ, lung ILC2s initiated robust IL-5 responses following viral challenge, which led to improved tissue integrity and survival. Conversely, functional PB1-F2 suppressed IL-5+ ILC2 responses and induced a dominant IL-13+ CD8 T cell response regardless of host IFN-γ. These findings demonstrate the critical interplay between the viral virulence factors and host cytokines in regulating protective pulmonary immunity during influenza virus infection. Functional plasticity of innate lymphoid cells (ILCs) and T cells is regulated by host environmental cues, but the influence of pathogen-derived virulence factors has not been described. We now report the interplay between host interferon (IFN)-γ and viral PB1-F2 virulence protein in regulating the functions of ILC2s and T cells that lead to recovery from influenza virus infection of mice. In the absence of IFN-γ, lung ILC2s from mice challenged with the A/California/04/2009 (CA04) H1N1 virus, containing nonfunctional viral PB1-F2, initiated a robust IL-5 response, which also led to improved tissue integrity and increased survival. Conversely, challenge with Puerto Rico/8/1934 (PR8) H1N1 virus expressing fully functional PB1-F2, suppressed IL-5+ ILC2 responses, and induced a dominant IL-13+ CD8 T cell response, regardless of host IFN-γ expression. IFN-γ–deficient mice had increased survival and improved tissue integrity following challenge with lethal doses of CA04, but not PR8 virus, and increased resistance was dependent on the presence of IFN-γR+ ILC2s. Reverse-engineered influenza viruses differing in functional PB1-F2 activity induced ILC2 and T cell phenotypes similar to the PB1-F2 donor strains, demonstrating the potent role of viral PB1-F2 in host resistance. These results show the ability of a pathogen virulence factor together with host IFN-γ to regulate protective pulmonary immunity during influenza infection.
Collapse
|
25
|
Dong C, Wang Y, Zhu W, Ma Y, Kim J, Wei L, Gonzalez GX, Wang BZ. Polycationic HA/CpG Nanoparticles Induce Cross-Protective Influenza Immunity in Mice. ACS APPLIED MATERIALS & INTERFACES 2022; 14:6331-6342. [PMID: 35084819 PMCID: PMC8832387 DOI: 10.1021/acsami.1c19192] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 05/28/2023]
Abstract
The intranasal (i.n.) route is an ideal vaccination approach for infectious respiratory diseases like influenza. Polycationic polyethylenimine (PEI) could form nanoscale complexes with negatively charged viral glycoproteins. Here we fabricated PEI-hemagglutinin (HA) and PEI-HA/CpG nanoparticles and investigated their immune responses and protective efficacies with an i.n. vaccination regimen in mice. Our results revealed that the nanoparticles significantly enhanced HA immunogenicity, providing heterologous cross-protection. The conserved HA stalk region induced substantial antibodies in the nanoparticle immunization groups. In contrast to the Th2-biased, IgG1-dominant antibody response generated by PEI-HA nanoparticles, PEI-HA/CpG nanoparticles generated more robust and balanced IgG1/IgG2a antibody responses with augmented neutralization activity and Fc-mediated antibody-dependent cellular cytotoxicity (ADCC). PEI-HA/CpG nanoparticles also induced enhanced local and systemic cellular immune responses. These immune responses did not decay over six months of observation postimmunization. PEI and CpG synergized these comprehensive immune responses. Thus, the PEI-HA/CpG nanoparticle is a potential cross-protective influenza vaccine candidate. Polycationic PEI nanoplatforms merit future development into mucosal vaccine systems.
Collapse
|
26
|
Hendy DA, Amouzougan EA, Young IC, Bachelder EM, Ainslie KM. Nano/microparticle Formulations for Universal Influenza Vaccines. AAPS J 2022; 24:24. [PMID: 34997352 PMCID: PMC8741137 DOI: 10.1208/s12248-021-00676-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/17/2021] [Indexed: 11/30/2022] Open
Abstract
Influenza affects millions of people worldwide and can result in severe sickness and even death. The best method of prevention is vaccination; however, the seasonal influenza vaccine often suffers from low efficacy and requires yearly vaccination due to changes in strain and viral mutations. More conserved universal influenza antigens like M2 ectodomain (M2e) and the stalk region of hemagglutinin (HA stalk) have been used clinically but often suffer from low antigenicity. To increase antigenicity, universal antigens have been formulated using nano/microparticles as vaccine carriers against influenza. Utilizing polymers, liposomes, metal, and protein-based particles, indicators of immunity and protection in mouse, pig, ferrets, and chicken models of influenza have been shown. In this review, seasonal and universal influenza vaccine formulations comprised of these materials including their physiochemical properties, fabrication, characterization, and biologic responses in vivo are highlighted. The review is concluded with future perspectives for nano/microparticles as carrier systems and other considerations within the universal influenza vaccine delivery landscape. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Dylan A Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Eva A Amouzougan
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Isabella C Young
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 4012 Marsico Hall, 125 Mason Farm Road, Chapel Hill, North Carolina, 27599, USA. .,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA. .,Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
27
|
Kozlovski S, Regev O, Sapoznikov A, Kizner M, Achdout H, Petrovich-Kopitman E, Elkahal J, Addadi Y, Silva Castanheira FVE, Feigelson SW, Kubes P, Erez N, Garbi N, Alon R. ICAMs are dispensable for influenza clearance and anti-viral humoral and cellular immunity. Front Immunol 2022; 13:1041552. [PMID: 36895258 PMCID: PMC9988921 DOI: 10.3389/fimmu.2022.1041552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 12/12/2022] [Indexed: 02/25/2023] Open
Abstract
αLβ2 (LFA-1) mediated interactions with ICAM-1 and ICAM-2 predominate leukocyte-vascular interactions, but their functions in extravascular cell-cell communications is still debated. The roles of these two ligands in leukocyte trafficking, lymphocyte differentiation, and immunity to influenza infections were dissected in the present study. Surprisingly, double ICAM-1 and ICAM-2 knock out mice (herein ICAM-1/2-/- mice) infected with a lab adapted H1N1 influenza A virus fully recovered from infection, elicited potent humoral immunity, and generated normal long lasting anti-viral CD8+ T cell memory. Furthermore, lung capillary ICAMs were dispensable for both NK and neutrophil entry to virus infected lungs. Mediastinal lymph nodes (MedLNs) of ICAM-1/2-/- mice poorly recruited naïve T cells and B lymphocytes but elicited normal humoral immunity critical for viral clearance and effective CD8+ differentiation into IFN-γ producing T cells. Furthermore, whereas reduced numbers of virus specific effector CD8+ T cells accumulated inside infected ICAM-1/2-/- lungs, normal virus-specific TRM CD8+ cells were generated inside these lungs and fully protected ICAM-1/2-/- mice from secondary heterosubtypic infections. B lymphocyte entry to the MedLNs and differentiation into extrafollicular plasmablasts, producing high affinity anti-influenza IgG2a antibodies, were also ICAM-1 and ICAM-2 independent. A potent antiviral humoral response was associated with accumulation of hyper-stimulated cDC2s in ICAM null MedLNs and higher numbers of virus-specific T follicular helper (Tfh) cells generated following lung infection. Mice selectively depleted of cDC ICAM-1 expression supported, however, normal CTL and Tfh differentiation following influenza infection, ruling out essential co-stimulatory functions of DC ICAM-1 in CD8+ and CD4+ T cell differentiation. Collectively our findings suggest that lung ICAMs are dispensable for innate leukocyte trafficking to influenza infected lungs, for the generation of peri-epithelial TRM CD8+ cells, and long term anti-viral cellular immunity. In lung draining LNs, although ICAMs promote lymphocyte homing, these key integrin ligands are not required for influenza-specific humoral immunity or generation of IFN-γ effector CD8+ T cells. In conclusion, our findings suggest unexpected compensatory mechanisms that orchestrate protective anti-influenza immunity in the absence of vascular and extravascular ICAMs.
Collapse
Affiliation(s)
- Stav Kozlovski
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofer Regev
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Marina Kizner
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Achdout
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | | | - Jacob Elkahal
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sara W Feigelson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Paul Kubes
- Department of Pharmacology and Physiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Noam Erez
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Natalio Garbi
- Department of Cellular Immunology, Institute of Experimental Immunology Medical Faculty, University of Bonn, Bonn, Germany
| | - Ronen Alon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
28
|
Omokanye A, Ong LC, Lebrero-Fernandez C, Bernasconi V, Schön K, Strömberg A, Bemark M, Saelens X, Czarnewski P, Lycke N. Clonotypic analysis of protective influenza M2e-specific lung resident Th17 memory cells reveals extensive functional diversity. Mucosal Immunol 2022; 15:717-729. [PMID: 35260804 PMCID: PMC8903128 DOI: 10.1038/s41385-022-00497-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/31/2022] [Accepted: 02/06/2022] [Indexed: 02/06/2023]
Abstract
The fate of tissue-resident memory CD4 T cells (Trm) has been incompletely investigated. Here we show that intranasal, but not parenteral, immunization with CTA1-3M2e-DD stimulated M2e-specific Th17 Trm cells, which conferred strong protection against influenza virus infection in the lung. These cells rapidly expanded upon infection and effectively restricted virus replication as determined by CD4 T cell depletion studies. Single-cell RNAseq transcriptomic and TCR VDJ-analysis of M2e-tetramer-sorted CD4 T cells on day 3 and 8 post infection revealed complete Th17-lineage dominance (no Th1 or Tregs) with extensive functional diversity and expression of gene markers signifying mature resident Trm cells (Cd69, Nfkbid, Brd2, FosB). Unexpectedly, the same TCR clonotype hosted cells with different Th17 subcluster functions (IL-17, IL-22), regulatory and cytotoxic cells, suggesting a tissue and context-dependent differentiation of reactivated Th17 Trm cells. A gene set enrichment analysis demonstrated up-regulation of regulatory genes (Lag3, Tigit, Ctla4, Pdcd1) in M2e-specific Trm cells on day 8, indicating a tissue damage preventing function. Thus, contrary to current thinking, lung M2e-specific Th17 Trm cells are sufficient for controlling infection and for protecting against tissue injury. These findings will have strong implications for vaccine development against respiratory virus infections and influenza virus infections, in particular.
Collapse
Affiliation(s)
- Ajibola Omokanye
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Li Ching Ong
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Cristina Lebrero-Fernandez
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Valentina Bernasconi
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anneli Strömberg
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mats Bemark
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Xavier Saelens
- grid.5342.00000 0001 2069 7798VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paulo Czarnewski
- grid.10548.380000 0004 1936 9377Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Nils Lycke
- grid.8761.80000 0000 9919 9582Mucosal Immunobiology and Vaccine Center (MIVAC), Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
29
|
Rozario C, Martínez-Sobrido L, McSorley HJ, Chauché C. Could Interleukin-33 (IL-33) Govern the Outcome of an Equine Influenza Virus Infection? Learning from Other Species. Viruses 2021; 13:2519. [PMID: 34960788 PMCID: PMC8704309 DOI: 10.3390/v13122519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/04/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
Influenza A viruses (IAVs) are important respiratory pathogens of horses and humans. Infected individuals develop typical respiratory disorders associated with the death of airway epithelial cells (AECs) in infected areas. Virulence and risk of secondary bacterial infections vary among IAV strains. The IAV non-structural proteins, NS1, PB1-F2, and PA-X are important virulence factors controlling AEC death and host immune responses to viral and bacterial infection. Polymorphism in these proteins impacts their function. Evidence from human and mouse studies indicates that upon IAV infection, the manner of AEC death impacts disease severity. Indeed, while apoptosis is considered anti-inflammatory, necrosis is thought to cause pulmonary damage with the release of damage-associated molecular patterns (DAMPs), such as interleukin-33 (IL-33). IL-33 is a potent inflammatory mediator released by necrotic cells, playing a crucial role in anti-viral and anti-bacterial immunity. Here, we discuss studies in human and murine models which investigate how viral determinants and host immune responses control AEC death and subsequent lung IL-33 release, impacting IAV disease severity. Confirming such data in horses and improving our understanding of early immunologic responses initiated by AEC death during IAV infection will better inform the development of novel therapeutic or vaccine strategies designed to protect life-long lung health in horses and humans, following a One Health approach.
Collapse
Affiliation(s)
- Christoforos Rozario
- Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, UK;
| | | | - Henry J. McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Wellcome Trust Building, Dow Street, Dundee DD1 5EH, UK;
| | - Caroline Chauché
- Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, UK;
| |
Collapse
|
30
|
Hao X, Zhang F, Yang Y, Shang S. The Evaluation of Cellular Immunity to Avian Viral Diseases: Methods, Applications, and Challenges. Front Microbiol 2021; 12:794514. [PMID: 34950125 PMCID: PMC8689181 DOI: 10.3389/fmicb.2021.794514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022] Open
Abstract
Cellular immune responses play critical roles in the control of viral infection. However, the immune protection against avian viral diseases (AVDs), a major challenge to poultry industry, is yet mainly evaluated by measuring humoral immune response though antibody-independent immune protection was increasingly evident in the development of vaccines against some of these diseases. The evaluation of cellular immune response to avian viral infection has long been neglected due to limited reagents and methods. Recently, with the availability of more immunological reagents and validated approaches, the evaluation of cellular immunity has become feasible and necessary for AVD. Herein, we reviewed the methods used for evaluating T cell immunity in chickens following infection or vaccination, which are involved in the definition of different cellular subset, the analysis of T cell activation, proliferation and cytokine secretion, and in vitro culture of antigen-presenting cells (APC) and T cells. The pros and cons of each method were discussed, and potential future directions to enhance the studies of avian cellular immunity were suggested. The methodological improvement and standardization in analyzing cellular immune response in birds after viral infection or vaccination would facilitate the dissection of mechanism of immune protection and the development of novel vaccines and therapeutics against AVD.
Collapse
Affiliation(s)
- Xiaoli Hao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Fan Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yi Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Shaobin Shang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- International Corporation Laboratory of Agriculture and Agricultural Products Safety, Yangzhou University, Yangzhou, China
| |
Collapse
|
31
|
Meldgaard TS, Blengio F, Maffione D, Sammicheli C, Tavarini S, Nuti S, Kratzer R, Medini D, Siena E, Bertholet S. Single-Cell Analysis of Antigen-Specific CD8+ T-Cell Transcripts Reveals Profiles Specific to mRNA or Adjuvanted Protein Vaccines. Front Immunol 2021; 12:757151. [PMID: 34777370 PMCID: PMC8586650 DOI: 10.3389/fimmu.2021.757151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/05/2021] [Indexed: 12/29/2022] Open
Abstract
CD8+ T cells play a key role in mediating protective immunity after immune challenges such as infection or vaccination. Several subsets of differentiated CD8+ T cells have been identified, however, a deeper understanding of the molecular mechanism that underlies T-cell differentiation is lacking. Conventional approaches to the study of immune responses are typically limited to the analysis of bulk groups of cells that mask the cells' heterogeneity (RNA-seq, microarray) and to the assessment of a relatively limited number of biomarkers that can be evaluated simultaneously at the population level (flow and mass cytometry). Single-cell analysis, on the other hand, represents a possible alternative that enables a deeper characterization of the underlying cellular heterogeneity. In this study, a murine model was used to characterize immunodominant hemagglutinin (HA533-541)-specific CD8+ T-cell responses to nucleic- and protein-based influenza vaccine candidates, using single-cell sorting followed by transcriptomic analysis. Investigation of single-cell gene expression profiles enabled the discovery of unique subsets of CD8+ T cells that co-expressed cytotoxic genes after vaccination. Moreover, this method enabled the characterization of antigen specific CD8+ T cells that were previously undetected. Single-cell transcriptome profiling has the potential to allow for qualitative discrimination of cells, which could lead to novel insights on biological pathways involved in cellular responses. This approach could be further validated and allow for more informed decision making in preclinical and clinical settings.
Collapse
Affiliation(s)
- Trine Sundebo Meldgaard
- Research & Development, GSK, Siena, Italy
- Biochemistry & Molecular Biology, University of Siena, Siena, Italy
| | - Fabiola Blengio
- Chemical & Biological Sciences, University of Torino, Torino, Italy
| | - Denise Maffione
- Chemical & Biological Sciences, University of Torino, Torino, Italy
| | | | | | - Sandra Nuti
- Research & Development, GSK, Siena, Italy
- Research & Development, GSK, Rockville, MD, United States
| | | | | | | | - Sylvie Bertholet
- Research & Development, GSK, Siena, Italy
- Research & Development, GSK, Rockville, MD, United States
| |
Collapse
|
32
|
Saito T, Sako Y, Sato-Kaneko F, Hosoya T, Yao S, Lao FS, Shpigelman J, Messer K, Pu M, Shukla NM, Chan M, Chu PJ, Cottam HB, Hayashi T, Carson DA, Corr M. Small Molecule Potentiator of Adjuvant Activity Enhancing Survival to Influenza Viral Challenge. Front Immunol 2021; 12:701445. [PMID: 34650551 PMCID: PMC8505803 DOI: 10.3389/fimmu.2021.701445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/30/2021] [Indexed: 01/02/2023] Open
Abstract
As viruses continue to mutate the need for rapid high titer neutralizing antibody responses has been highlighted. To meet these emerging threats, agents that enhance vaccine adjuvant activity are needed that are safe with minimal local or systemic side effects. To respond to this demand, we sought small molecules that would sustain and improve the protective effect of a currently approved adjuvant, monophosphoryl lipid A (MPLA), a Toll-like receptor 4 (TLR4) agonist. A lead molecule from a high-throughput screen, (N-(4-(2,5-dimethylphenyl)thiazol-2-yl)-4-(piperidin-1-ylsulfonyl)benzamide, was identified as a hit compound that sustained NF-κB activation by a TLR4 ligand, lipopolysaccharide (LPS), after an extended incubation (16 h). In vitro, the resynthesized compound (2D216) enhanced TLR4 ligand-induced innate immune activation and antigen presenting function in primary murine bone marrow-derived dendritic cells without direct activation of T cells. In vivo murine vaccination studies demonstrated that compound 2D216 acted as a potent co-adjuvant when used in combination with MPLA that enhanced antigen-specific IgG equivalent to that of AS01B. The combination adjuvant MPLA/2D216 produced Th1 dominant immune responses and importantly protected mice from lethal influenza virus challenge. 2D216 alone or 2D216/MPLA demonstrated minimal local reactogenicity and no systemic inflammatory response. In summary, 2D216 augmented the beneficial protective immune responses of MPLA as a co-adjuvant and showed an excellent safety profile.
Collapse
Affiliation(s)
- Tetsuya Saito
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States.,Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yukiya Sako
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Fumi Sato-Kaneko
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Tadashi Hosoya
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States.,Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shiyin Yao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Fitzgerald S Lao
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Jonathan Shpigelman
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Karen Messer
- The Herbert Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, CA, United States
| | - Minya Pu
- The Herbert Wertheim School of Public Health and Longevity, University of California San Diego, La Jolla, CA, United States
| | - Nikunj M Shukla
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Michael Chan
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Paul J Chu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Howard B Cottam
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Tomoko Hayashi
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Dennis A Carson
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
33
|
Neuraminidase in Virus-like Particles Contributes to the Protection against High Dose of Avian Influenza Virus Challenge Infection. Pathogens 2021; 10:pathogens10101291. [PMID: 34684240 PMCID: PMC8537550 DOI: 10.3390/pathogens10101291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 11/22/2022] Open
Abstract
Neuraminidase is an important target for influenza vaccination. In this study, we generated avian influenza VLPs, expressing hemagglutinin (HA), neuraminidase (NA), HA and NA co-expressed (HANA), to evaluate the protective role of NA against a high (10LD50) and low (2LD50) dose of avian influenza virus challenge infections. A single immunization with HANA VLPs elicited the highest level of virus-specific IgG, IgG1, and IgG2a responses from the sera post-vaccination and the lungs post-challenge-infection. Potent antibody-secreting cell responses were observed from the spleens and lungs of HANA-VLP-immunized mice post-challenge-infection. HANA VLPs induced the highest CD4+ T cell, CD8+ T cell, and germinal center B cells, while strongly limiting inflammatory cytokine production in the lungs compared to other VLP immunization groups. In correlation with these findings, the lowest bodyweight losses and lung virus titers were observed from HANA VLP immunization, and all of the immunized mice survived irrespective of the challenge dose. Contrastingly, VLPs expressing either HA or NA alone failed to elicit complete protection. These results indicated that NA in VLPs played a critical role in inducing protection against a high dose of the challenge infection.
Collapse
|
34
|
Li C, Revote J, Ramarathinam SH, Chung SZ, Croft NP, Scull KE, Huang Z, Ayala R, Braun A, Mifsud NA, Illing PT, Faridi P, Purcell AW. Resourcing, annotating, and analysing synthetic peptides of SARS-CoV-2 for immunopeptidomics and other immunological studies. Proteomics 2021; 21:e2100036. [PMID: 33811468 PMCID: PMC8250278 DOI: 10.1002/pmic.202100036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022]
Abstract
SARS-CoV-2 has caused a significant ongoing pandemic worldwide. A number of studies have examined the T cell mediated immune responses against SARS-CoV-2, identifying potential T cell epitopes derived from the SARS-CoV-2 proteome. Such studies will aid in identifying targets for vaccination and immune monitoring. In this study, we applied tandem mass spectrometry and proteomic techniques to a library of ∼40,000 synthetic peptides, in order to generate a large dataset of SARS-CoV-2 derived peptide MS/MS spectra. On this basis, we built an online knowledgebase, termed virusMS (https://virusms.erc.monash.edu/), to document, annotate and analyse these synthetic peptides and their spectral information. VirusMS incorporates a user-friendly interface to facilitate searching, browsing and downloading the database content. Detailed annotations of the peptides, including experimental information, peptide modifications, predicted peptide-HLA (human leukocyte antigen) binding affinities, and peptide MS/MS spectral data, are provided in virusMS.
Collapse
Affiliation(s)
- Chen Li
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Jerico Revote
- Monash Bioinformatics PlatformMonash UniversityMelbourneVictoriaAustralia
| | - Sri H. Ramarathinam
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Shan Zou Chung
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Nathan P. Croft
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Katherine E. Scull
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Ziyi Huang
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Rochelle Ayala
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Asolina Braun
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Nicole A. Mifsud
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Patricia T. Illing
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Pouya Faridi
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology and Infection and Immunity ProgramMonash Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
35
|
Youngs J, Provine NM, Lim N, Sharpe HR, Amini A, Chen YL, Luo J, Edmans MD, Zacharopoulou P, Chen W, Sampson O, Paton R, Hurt WJ, Duncan DA, McNaughton AL, Miao VN, Leaver S, Wyncoll DLA, Ball J, Hopkins P, Oxford Immunology Network Covid-19 response T cell Consortium, Oxford Protective T cell Immunology for COVID-19 (OPTIC) Clinical team, Skelly DT, Barnes E, Dunachie S, Ogg G, Lambe T, Pavord I, Shalek AK, Thompson CP, Xue L, Macallan DC, Goulder P, Klenerman P, Bicanic T. Identification of immune correlates of fatal outcomes in critically ill COVID-19 patients. PLoS Pathog 2021; 17:e1009804. [PMID: 34529726 PMCID: PMC8445447 DOI: 10.1371/journal.ppat.1009804] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022] Open
Abstract
Prior studies have demonstrated that immunologic dysfunction underpins severe illness in COVID-19 patients, but have lacked an in-depth analysis of the immunologic drivers of death in the most critically ill patients. We performed immunophenotyping of viral antigen-specific and unconventional T cell responses, neutralizing antibodies, and serum proteins in critically ill patients with SARS-CoV-2 infection, using influenza infection, SARS-CoV-2-convalescent health care workers, and healthy adults as controls. We identify mucosal-associated invariant T (MAIT) cell activation as an independent and significant predictor of death in COVID-19 (HR = 5.92, 95% CI = 2.49-14.1). MAIT cell activation correlates with several other mortality-associated immunologic measures including broad activation of CD8+ T cells and non-Vδ2 γδT cells, and elevated levels of cytokines and chemokines, including GM-CSF, CXCL10, CCL2, and IL-6. MAIT cell activation is also a predictor of disease severity in influenza (ECMO/death HR = 4.43, 95% CI = 1.08-18.2). Single-cell RNA-sequencing reveals a shift from focused IFNα-driven signals in COVID-19 ICU patients who survive to broad pro-inflammatory responses in fatal COVID-19 -a feature not observed in severe influenza. We conclude that fatal COVID-19 infection is driven by uncoordinated inflammatory responses that drive a hierarchy of T cell activation, elements of which can serve as prognostic indicators and potential targets for immune intervention.
Collapse
Affiliation(s)
- Jonathan Youngs
- Institute for Infection & Immunity, St. George’s University of London, London, United Kingdom
- Clinical Academic Group in Infection and Immunity, St. George’s Hospital NHS Trust, London, United Kingdom
| | - Nicholas M. Provine
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicholas Lim
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Ali Amini
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Yi-Ling Chen
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Jian Luo
- Respiratory Medicine Unit, and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Matthew D. Edmans
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Panagiota Zacharopoulou
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Wentao Chen
- Respiratory Medicine Unit, and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Oliver Sampson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Robert Paton
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - William J. Hurt
- Institute for Infection & Immunity, St. George’s University of London, London, United Kingdom
- Clinical Academic Group in Infection and Immunity, St. George’s Hospital NHS Trust, London, United Kingdom
| | - David A. Duncan
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, United Kingdom
| | - Anna L. McNaughton
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Vincent N. Miao
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Susannah Leaver
- Intensive Care Medicine, St George’s University Hospital NHS Foundation Trust, London, United Kingdom
| | - Duncan L. A. Wyncoll
- Intensive Care Medicine, Guy’s and St Thomas’ Hospital NHS Foundation Trust, London, United Kingdom
| | - Jonathan Ball
- Intensive Care Medicine, St George’s University Hospital NHS Foundation Trust, London, United Kingdom
| | - Philip Hopkins
- Centre for Human & Applied Physiological Sciences, School of Basic & Medical Biosciences, Faculty of Life Sciences, & Medicine, King’s College, London, United Kingdom
| | | | | | - Donal T. Skelly
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Graham Ogg
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Teresa Lambe
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Ian Pavord
- Respiratory Medicine Unit, and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Alex K. Shalek
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Craig P. Thompson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Luzheng Xue
- Respiratory Medicine Unit, and Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Derek C. Macallan
- Institute for Infection & Immunity, St. George’s University of London, London, United Kingdom
- Clinical Academic Group in Infection and Immunity, St. George’s Hospital NHS Trust, London, United Kingdom
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tihana Bicanic
- Institute for Infection & Immunity, St. George’s University of London, London, United Kingdom
- Clinical Academic Group in Infection and Immunity, St. George’s Hospital NHS Trust, London, United Kingdom
| |
Collapse
|
36
|
Schoeman D, Fielding BC. Human Coronaviruses: Counteracting the Damage by Storm. Viruses 2021; 13:1457. [PMID: 34452323 PMCID: PMC8402835 DOI: 10.3390/v13081457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/15/2022] Open
Abstract
Over the past 18 years, three highly pathogenic human (h) coronaviruses (CoVs) have caused severe outbreaks, the most recent causative agent, SARS-CoV-2, being the first to cause a pandemic. Although much progress has been made since the COVID-19 pandemic started, much about SARS-CoV-2 and its disease, COVID-19, is still poorly understood. The highly pathogenic hCoVs differ in some respects, but also share some similarities in clinical presentation, the risk factors associated with severe disease, and the characteristic immunopathology associated with the progression to severe disease. This review aims to highlight these overlapping aspects of the highly pathogenic hCoVs-SARS-CoV, MERS-CoV, and SARS-CoV-2-briefly discussing the importance of an appropriately regulated immune response; how the immune response to these highly pathogenic hCoVs might be dysregulated through interferon (IFN) inhibition, antibody-dependent enhancement (ADE), and long non-coding RNA (lncRNA); and how these could link to the ensuing cytokine storm. The treatment approaches to highly pathogenic hCoV infections are discussed and it is suggested that a greater focus be placed on T-cell vaccines that elicit a cell-mediated immune response, using rapamycin as a potential agent to improve vaccine responses in the elderly and obese, and the potential of stapled peptides as antiviral agents.
Collapse
Affiliation(s)
| | - Burtram C. Fielding
- Molecular Biology and Virology Research Laboratory, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa;
| |
Collapse
|
37
|
Myers MA, Smith AP, Lane LC, Moquin DJ, Aogo R, Woolard S, Thomas P, Vogel P, Smith AM. Dynamically linking influenza virus infection kinetics, lung injury, inflammation, and disease severity. eLife 2021; 10:68864. [PMID: 34282728 PMCID: PMC8370774 DOI: 10.7554/elife.68864] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Influenza viruses cause a significant amount of morbidity and mortality. Understanding host immune control efficacy and how different factors influence lung injury and disease severity are critical. We established and validated dynamical connections between viral loads, infected cells, CD8+ T cells, lung injury, inflammation, and disease severity using an integrative mathematical model-experiment exchange. Our results showed that the dynamics of inflammation and virus-inflicted lung injury are distinct and nonlinearly related to disease severity, and that these two pathologic measurements can be independently predicted using the model-derived infected cell dynamics. Our findings further indicated that the relative CD8+ T cell dynamics paralleled the percent of the lung that had resolved with the rate of CD8+ T cell-mediated clearance rapidly accelerating by over 48,000 times in 2 days. This complimented our analyses showing a negative correlation between the efficacy of innate and adaptive immune-mediated infected cell clearance, and that infection duration was driven by CD8+ T cell magnitude rather than efficacy and could be significantly prolonged if the ratio of CD8+ T cells to infected cells was sufficiently low. These links between important pathogen kinetics and host pathology enhance our ability to forecast disease progression, potential complications, and therapeutic efficacy.
Collapse
Affiliation(s)
- Margaret A Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - Amanda P Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - Lindey C Lane
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - David J Moquin
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Rosemary Aogo
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - Stacie Woolard
- Flow Cytometry Core, St. Jude Children's Research Hospital, Memphis, United States
| | - Paul Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, United States
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, United States
| | - Amber M Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| |
Collapse
|
38
|
Adam L, Rosenbaum P, Quentric P, Parizot C, Bonduelle O, Guillou N, Corneau A, Dorgham K, Miyara M, Luyt CE, Guihot A, Gorochov G, Combadière C, Combadière B. Nucleocapsid-specific and PD-L1+CXCR3+ CD8 polyfunctional T-cell abundances are associated with survival of critical SARS-CoV2-infected patients. JCI Insight 2021; 6:e151571. [PMID: 34283810 PMCID: PMC8492305 DOI: 10.1172/jci.insight.151571] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/14/2021] [Indexed: 11/30/2022] Open
Abstract
The importance of the adaptive T cell response in the control and resolution of viral infection has been well established. However, the nature of T cell–mediated viral control mechanisms in life-threatening stages of COVID-19 has yet to be determined. The aim of the present study was to determine the function and phenotype of T cell populations associated with survival or death of patients with COVID-19 in intensive care as a result of phenotypic and functional profiling by mass cytometry. Increased frequencies of circulating, polyfunctional CD4+CXCR5+HLA-DR+ stem cell memory T cells (Tscms) and decreased proportions of granzyme B–expressing and perforin-expressing effector memory T cells were detected in recovered and deceased patients, respectively. The higher abundance of polyfunctional PD-L1+CXCR3+CD8+ effector T cells (Teffs), CXCR5+HLA-DR+ Tscms, and anti-nucleocapsid (anti-NC) cytokine-producing T cells permitted us to differentiate between recovered and deceased patients. The results from a principal component analysis show an imbalance in the T cell compartment that allowed for the separation of recovered and deceased patients. The paucity of circulating PD-L1+CXCR3+CD8+ Teffs and NC-specific CD8+ T cells accurately forecasts fatal disease outcome. This study provides insight into the nature of the T cell populations involved in the control of COVID-19 and therefore might impact T cell–based vaccine designs for this infectious disease.
Collapse
Affiliation(s)
- Lucille Adam
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| | - Pierre Rosenbaum
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| | - Paul Quentric
- Département d'Immunologie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christophe Parizot
- Département d'Immunologie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Olivia Bonduelle
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| | - Noëlline Guillou
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| | - Aurelien Corneau
- Plateforme de cytométrie de la Pitié-Salpêtrière CyPS, UPMC/CNRS/INSERM, Paris, France
| | - Karim Dorgham
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| | - Makoto Miyara
- Département d'Immunologie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Charles-Edouard Luyt
- Service de Médecine Intensive-Réanimation et Pneumologie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Amélie Guihot
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| | - Guy Gorochov
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| | | | - Behazine Combadière
- Centre d'Immunologie et des Maladies Infectieuses, INSERM UMR 1135, Paris, France
| |
Collapse
|
39
|
Vandoorn E, Leroux-Roels I, Leroux-Roels G, Parys A, Vincent A, Van Reeth K. Detection of H1 Swine Influenza A Virus Antibodies in Human Serum Samples by Age Group 1. Emerg Infect Dis 2021; 26:2118-2128. [PMID: 32818398 PMCID: PMC7454048 DOI: 10.3201/eid2609.191796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Most H1 influenza A viruses (IAVs) of swine are derived from past human viruses. As human population immunity against these IAVs gradually decreases, the risk of reintroduction to humans increases. We examined 549 serum samples from persons 0-97 years of age collected in Belgium during 2017-2018 for hemagglutination inhibiting and virus neutralizing antibodies against 7 major H1 swine IAV (swIAV) clades and 3 human progenitor IAVs. Seroprevalence (titers >40) rates were >50% for classical swine and European human-like swIAVs, >24% for North American human-like δ1a and Asian avian-like swIAVs, and <10% for North American human-like δ1b and European avian-like swIAVs, but rates were age-dependent. Antibody titers against human-like swIAVs and supposed human precursor IAVs correlated with correlation coefficients of 0.30-0.86. Our serologic findings suggest that European avian-like, clade 1C.2.1, and North American human-like δ1b, clade 1B.2.2.2, H1 swIAVs pose the highest pandemic risk.
Collapse
|
40
|
Postoak JL, Yang G, Wu L, Van Kaer L. Cellular self-cannibalism helps immune cells fight the flu. FEBS J 2021; 288:3154-3158. [PMID: 33411418 PMCID: PMC8131204 DOI: 10.1111/febs.15641] [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: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 11/30/2022]
Abstract
CD4 T cells are critical for generating protective immune responses to infection with influenza virus. Although most CD4 T cells react with peptides from extracellular sources, many react with peptides from viral particles synthesized inside cells; however, the pathways employed for processing the latter antigens remain uncertain. Deng et al. provide evidence for a role of autophagy, a cellular self-eating process, in this unconventional antigen processing route, with potential implications for the development of influenza virus vaccines.
Collapse
Affiliation(s)
- Joshua Luke Postoak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Guan Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
41
|
Thompson EA, Cascino K, Ordonez AA, Zhou W, Vaghasia A, Hamacher-Brady A, Brady NR, Sun IH, Wang R, Rosenberg AZ, Delannoy M, Rothman R, Fenstermacher K, Sauer L, Shaw-Saliba K, Bloch EM, Redd AD, Tobian AAR, Horton M, Smith K, Pekosz A, D'Alessio FR, Yegnasubramanian S, Ji H, Cox AL, Powell JD. Metabolic programs define dysfunctional immune responses in severe COVID-19 patients. Cell Rep 2021; 34:108863. [PMID: 33691089 PMCID: PMC7908880 DOI: 10.1016/j.celrep.2021.108863] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/17/2020] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
It is unclear why some SARS-CoV-2 patients readily resolve infection while others develop severe disease. By interrogating metabolic programs of immune cells in severe and recovered coronavirus disease 2019 (COVID-19) patients compared with other viral infections, we identify a unique population of T cells. These T cells express increased Voltage-Dependent Anion Channel 1 (VDAC1), accompanied by gene programs and functional characteristics linked to mitochondrial dysfunction and apoptosis. The percentage of these cells increases in elderly patients and correlates with lymphopenia. Importantly, T cell apoptosis is inhibited in vitro by targeting the oligomerization of VDAC1 or blocking caspase activity. We also observe an expansion of myeloid-derived suppressor cells with unique metabolic phenotypes specific to COVID-19, and their presence distinguishes severe from mild disease. Overall, the identification of these metabolic phenotypes provides insight into the dysfunctional immune response in acutely ill COVID-19 patients and provides a means to predict and track disease severity and/or design metabolic therapeutic regimens.
Collapse
Affiliation(s)
- Elizabeth A Thompson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Katherine Cascino
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alvaro A Ordonez
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Weiqiang Zhou
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Ajay Vaghasia
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Anne Hamacher-Brady
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Nathan R Brady
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Im-Hong Sun
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Rulin Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael Delannoy
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Richard Rothman
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Katherine Fenstermacher
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lauren Sauer
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kathyrn Shaw-Saliba
- Department of Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andrew D Redd
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Baltimore, MD 21205, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Maureen Horton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kellie Smith
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Franco R D'Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Srinivasan Yegnasubramanian
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Andrea L Cox
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA.
| | - Jonathan D Powell
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
42
|
Wu J, Abraham SN. The Roles of T cells in Bladder Pathologies. Trends Immunol 2021; 42:248-260. [PMID: 33536141 PMCID: PMC7914211 DOI: 10.1016/j.it.2021.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/26/2022]
Abstract
T lymphocytes play important roles in the skin and mucosal surfaces such as the gut and lung. Until recently the contributions of T cells to mammalian bladder immunity were largely unknown. With newer techniques, including single-cell RNA sequencing and reporter mice, an understanding is emerging of T cell roles in bladder diseases (bacterial infections, bladder cancer, chronic inflammation). In these pathologies, many bladder T cell responses can be harmful to the host through suboptimal clearance of bacteria or cancer cells, or by modulating autoinflammation. Recent findings suggest that T cell behavior might be influenced by resident T cell interactions with the bladder microbiota and other immunostimulants. Thus, regulating bladder T cell functions could emerge as a putative immunotherapy to treat some bladder diseases.
Collapse
Affiliation(s)
- Jianxuan Wu
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Soman N Abraham
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore.
| |
Collapse
|
43
|
Topham DJ, DeDiego ML, Nogales A, Sangster MY, Sant A. Immunity to Influenza Infection in Humans. Cold Spring Harb Perspect Med 2021; 11:a038729. [PMID: 31871226 PMCID: PMC7919402 DOI: 10.1101/cshperspect.a038729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review discusses the human immune responses to influenza infection with some insights from studies using animal models, such as experimental infection of mice. Recent technological advances in the study of human immune responses have greatly added to our knowledge of the infection and immune responses, and therefore much of the focus is on recent studies that have moved the field forward. We consider the complexity of the adaptive response generated by many sequential encounters through infection and vaccination.
Collapse
Affiliation(s)
- David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Marta L DeDiego
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Cientificas, 28049 Madrid, Spain
| | - Aitor Nogales
- Instituto Nacional de Investigación y Tecnologia Agraria y Ailmentaria, 28040 Madrid, Spain
| | - Mark Y Sangster
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | - Andrea Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, USA
| |
Collapse
|
44
|
Mohn KGI, Brokstad KA, Islam S, Oftung F, Tøndel C, Aarstad HJ, Cox RJ. Early Induction of Cross-Reactive CD8+ T-Cell Responses in Tonsils After Live-Attenuated Influenza Vaccination in Children. J Infect Dis 2021; 221:1528-1537. [PMID: 32255493 PMCID: PMC7137893 DOI: 10.1093/infdis/jiz583] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/10/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Live-attenuated influenza vaccine (LAIV) was licensed for prophylaxis of children 2-17 years old in Europe in 2012 and is administered as a nasal spray. Live-attenuated influenza vaccine induces both mucosal and systemic antibodies and systemic T-cell responses. Tonsils are the lymph nodes serving the upper respiratory tract, acting as both induction and effector site for mucosal immunity. METHODS Here, we have studied the early tonsillar T-cell responses induced in children after LAIV. Thirty-nine children were immunized with trivalent LAIV (containing A/H1N1, A/H3N2, and B viruses) at days 3, 7, and 14 before tonsillectomy. Nonvaccinated controls were included for comparison. Tonsils and peripheral blood (pre- and postvaccination) were collected to study T-cell responses. RESULTS Tonsillar and systemic T-cell responses differed between influenza strains, and both were found against H3N2 and B viruses, whereas only systemic responses were observed against A/H1N1. A significant increase in cross-reactive tonsillar CD8+ T cells recognizing conserved epitopes from a broad range of seasonal and pandemic viruses occurred at day 14. Tonsillar T cells showed significant cytokine responses (Th1, Th2, and granulocyte-macrophage colony-stimulating factor). CONCLUSIONS Our findings support the use of LAIV in children to elicit broadly cross-reactive T cells, which are not induced by traditional inactivated influenza vaccines and may provide protection to novel virus strains.
Collapse
Affiliation(s)
- K G-I Mohn
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Emergency Care Clinic, Haukeland University Hospital, Bergen, Norway
| | - K A Brokstad
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - S Islam
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway
| | - F Oftung
- Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - C Tøndel
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - H J Aarstad
- Department of Otorhinolaryngology/Head and Neck Surgery, Haukeland University Hospital, Bergen, Norway
| | - R J Cox
- Influenza Centre, University of Bergen, Bergen, Norway.,K. G. Jebsen Center for Influenza Vaccines, University of Bergen, Bergen, Norway.,Department of Research and Development, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
45
|
NMP4 regulates the innate immune response to influenza A virus infection. Mucosal Immunol 2021; 14:209-218. [PMID: 32152414 PMCID: PMC7483155 DOI: 10.1038/s41385-020-0280-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 02/05/2020] [Accepted: 02/25/2020] [Indexed: 02/04/2023]
Abstract
Severe influenza A virus infection typically triggers excessive and detrimental lung inflammation with massive cell infiltration and hyper-production of cytokines and chemokines. We identified a novel function for nuclear matrix protein 4 (NMP4), a zinc-finger-containing transcription factor playing roles in bone formation and spermatogenesis, in regulating antiviral immune response and immunopathology. Nmp4-deficient mice are protected from H1N1 influenza infection, losing only 5% body weight compared to a 20% weight loss in wild type mice. While having no effects on viral clearance or CD8/CD4 T cell or humoral responses, deficiency of Nmp4 in either lung structural cells or hematopoietic cells significantly reduces the recruitment of monocytes and neutrophils to the lungs. Consistent with fewer innate cells in the airways, influenza-infected Nmp4-deficient mice have significantly decreased expression of chemokine genes Ccl2, Ccl7 and Cxcl1 as well as pro-inflammatory cytokine genes Il1b and Il6. Furthermore, NMP4 binds to the promoters and/or conserved non-coding sequences of the chemokine genes and regulates their expression in mouse lung epithelial cells and macrophages. Our data suggest that NMP4 functions to promote monocyte- and neutrophil-attracting chemokine expression upon influenza A infection, resulting in exaggerated innate inflammation and lung tissue damage.
Collapse
|
46
|
Asha K, Khanna M, Kumar B. Current Insights into the Host Immune Response to Respiratory Viral Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:59-83. [PMID: 34661891 DOI: 10.1007/978-3-030-67452-6_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Respiratory viral infections often lead to severe illnesses varying from mild or asymptomatic upper respiratory tract infections to severe bronchiolitis and pneumonia or/and chronic obstructive pulmonary disease. Common viral infections, including but not limited to influenza virus, respiratory syncytial virus, rhinovirus and coronavirus, are often the leading cause of morbidity and mortality. Since the lungs are continuously exposed to foreign particles, including respiratory pathogens, it is also well equipped for recognition and antiviral defense utilizing the complex network of innate and adaptive immune cells. Immediately upon infection, a range of proinflammatory cytokines, chemokines and an interferon response is generated, thereby making the immune response a two edged sword, on one hand it is required to eliminate viral pathogens while on other hand it's prolonged response can lead to chronic infection and significant pulmonary damage. Since vaccines to all respiratory viruses are not available, a better understanding of the virus-host interactions, leading to the development of immune response, is critically needed to design effective therapies to limit the severity of inflammatory damage, enhance viral clearance and to compliment the current strategies targeting the virus. In this chapter, we discuss the host responses to common respiratory viral infections, the key players of adaptive and innate immunity and the fine balance that exists between the viral clearance and immune-mediated damage.
Collapse
Affiliation(s)
- Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Madhu Khanna
- Department of Virology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Binod Kumar
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
47
|
Host factors involved in influenza virus infection. Emerg Top Life Sci 2020; 4:389-398. [PMID: 33210707 DOI: 10.1042/etls20200232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022]
Abstract
Influenza virus causes an acute febrile respiratory disease in humans that is commonly known as 'flu'. Influenza virus has been around for centuries and is one of the most successful, and consequently most studied human viruses. This has generated tremendous amount of data and information, thus it is pertinent to summarise these for, particularly interdisciplinary readers. Viruses are acellular organisms and exist at the interface of living and non-living. Due to this unique characteristic, viruses require another organism, i.e. host to survive. Viruses multiply inside the host cell and are obligate intracellular pathogens, because their relationship with the host is almost always harmful to host. In mammalian cells, the life cycle of a virus, including influenza is divided into five main steps: attachment, entry, synthesis, assembly and release. To complete these steps, some viruses, e.g. influenza utilise all three parts - plasma membrane, cytoplasm and nucleus, of the cell; whereas others, e.g. SARS-CoV-2 utilise only plasma membrane and cytoplasm. Hence, viruses interact with numerous host factors to complete their life cycle, and these interactions are either exploitative or antagonistic in nature. The host factors involved in the life cycle of a virus could be divided in two broad categories - proviral and antiviral. This perspective has endeavoured to assimilate the information about the host factors which promote and suppress influenza virus infection. Furthermore, an insight into host factors that play a dual role during infection or contribute to influenza virus-host adaptation and disease severity has also been provided.
Collapse
|
48
|
Toy R, Keenum MC, Pradhan P, Phang K, Chen P, Chukwu C, Nguyen LAH, Liu J, Jain S, Kozlowski G, Hosten J, Suthar MS, Roy K. TLR7 and RIG-I dual-adjuvant loaded nanoparticles drive broadened and synergistic responses in dendritic cells in vitro and generate unique cellular immune responses in influenza vaccination. J Control Release 2020; 330:866-877. [PMID: 33160004 DOI: 10.1016/j.jconrel.2020.10.060] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 01/08/2023]
Abstract
Although the existing flu vaccines elicit strong antigen-specific antibody responses, they fail to provide effective, long term protection - partly due to the absence of robust cellular memory immunity. We hypothesized that co-administration of combination adjuvants, mirroring the flu-virus related innate signaling pathways, could elicit strong cellular immunity. Here, we show that the small molecule adjuvant R848 and the RNA adjuvant PUUC, targeting endosomal TLR7s and cytoplasmic RLRs respectively, when delivered together in polymer nanoparticles (NP), elicits a broadened immune responses in mouse bone marrow-derived dendritic cells (mBMDCs) and a synergistic response in both mouse and human plasmacytoid dendritic cells (pDCs). In mBMDCs, NP-R848-PUUC induced both NF-κB and interferon signaling. Interferon responses to co-delivered R848 and PUUC were additive in human peripheral blood mononuclear cells (PBMCs) and synergistic in human FLT3-differentiated mBMDCs and CAL-1 pDCs. Vaccination with NPs loaded with H1N1 Flu antigen, R848, and PUUC increased percentage of CD8+ T-cells in the lungs, percentage of antigen-specific CD4-T-cells in the spleen, and enhanced overall cytokine-secreting T cell percentages upon antigen restimulation. Also, in the spleen, T lymphopenia, especially after in vitro restimulation with dual adjuvants, was observed, indicating highly antigen-reactive T cells. Our results demonstrate that simultaneous engagement of TLR7 and RIG-I pathways using particulate carriers is a potential approach to improve cellular immunity in flu vaccination.
Collapse
Affiliation(s)
- Randall Toy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - M Cole Keenum
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Pallab Pradhan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Katelynn Phang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Patrick Chen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Chinwendu Chukwu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Lily Anh H Nguyen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Jiaying Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sambhav Jain
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Gabrielle Kozlowski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Justin Hosten
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
49
|
Tang S, Zhu W, Wang BZ. Influenza Vaccines toward Universality through Nanoplatforms and Given by Microneedle Patches. Viruses 2020; 12:E1212. [PMID: 33114336 PMCID: PMC7690886 DOI: 10.3390/v12111212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/25/2022] Open
Abstract
Influenza is one of the top threats to public health. The best strategy to prevent influenza is vaccination. Because of the antigenic changes in the major surface antigens of influenza viruses, current seasonal influenza vaccines need to be updated every year to match the circulating strains and are suboptimal for protection. Furthermore, seasonal vaccines do not protect against potential influenza pandemics. A universal influenza vaccine will eliminate the threat of both influenza epidemics and pandemics. Due to the massive challenge in realizing influenza vaccine universality, a single vaccine strategy cannot meet the need. A comprehensive approach that integrates advances in immunogen designs, vaccine and adjuvant nanoplatforms, and vaccine delivery and controlled release has the potential to achieve an effective universal influenza vaccine. This review will summarize the advances in the research and development of an affordable universal influenza vaccine.
Collapse
Affiliation(s)
| | | | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA; (S.T.); (W.Z.)
| |
Collapse
|
50
|
Moise L, Gutiérrez AH, Khan S, Tan S, Ardito M, Martin WD, De Groot AS. New Immunoinformatics Tools for Swine: Designing Epitope-Driven Vaccines, Predicting Vaccine Efficacy, and Making Vaccines on Demand. Front Immunol 2020; 11:563362. [PMID: 33123135 PMCID: PMC7571332 DOI: 10.3389/fimmu.2020.563362] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022] Open
Abstract
Novel computational tools for swine vaccine development can expand the range of immunization approaches available to prevent economically devastating swine diseases and spillover events between pigs and humans. PigMatrix and EpiCC are two new tools for swine T cell epitope identification and vaccine efficacy analysis that have been integrated into an existing computational vaccine design platform named iVAX. The iVAX platform is already in use for the development of human vaccines, thus integration of these tools into iVAX improves and expands the utility of the platform overall by making previously validated immunoinformatics tools, developed for humans, available for use in the design and analysis of swine vaccines. PigMatrix predicts T cell epitopes for a broad array of class I and class II swine leukocyte antigen (SLA) using matrices that enable the scoring of sequences for likelihood of binding to SLA. PigMatrix facilitates the prospective selection of T cell epitopes from the sequences of swine pathogens for vaccines and permits the comparison of those predicted epitopes with "self" (the swine proteome) and with sequences from other strains. Use of PigMatrix with additional tools in the iVAX toolkit also enables the computational design of vaccines in silico, for testing in vivo. EpiCC uses PigMatrix to analyze existing or proposed vaccines for their potential to protect, based on a comparison between T cell epitopes in the vaccine and circulating strains of the same pathogen. Performing an analysis of T cell epitope relatedness analysis using EpiCC may facilitate vaccine selection when a novel strain emerges in a herd and also permits analysis of evolutionary drift as a means of immune escape. This review of novel computational immunology tools for swine describes the application of PigMatrix and EpiCC in case studies, such as the design of cross-conserved T cell epitopes for swine influenza vaccine or for African Swine Fever. We also describe the application of EpiCC for determination of the best vaccine strains to use against circulating viral variants of swine influenza, swine rotavirus, and porcine circovirus type 2. The availability of these computational tools accelerates infectious disease research for swine and enable swine vaccine developers to strategically advance their vaccines to market.
Collapse
Affiliation(s)
- Lenny Moise
- EpiVax, Inc., Providence, RI, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | | | | | - Swan Tan
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | | | | | - Anne S De Groot
- EpiVax, Inc., Providence, RI, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| |
Collapse
|