1
|
Wilks LR, Joshi G, Rychener N, Gill HS. Generation of Broad Protection against Influenza with Di-Tyrosine-Cross-Linked M2e Nanoclusters. ACS Infect Dis 2024; 10:1552-1560. [PMID: 38623820 DOI: 10.1021/acsinfecdis.3c00429] [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: 04/17/2024]
Abstract
Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.
Collapse
Affiliation(s)
- Logan R Wilks
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Natalie Rychener
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Eighth Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
2
|
Gomes KB, Allotey-Babington GL, D'Sa S, Kang SM, D'Souza MJ. Dendritic cell activation by a micro particulate based system containing the influenza matrix-2 protein virus-like particle (M2e VLP). Int J Pharm 2022; 622:121667. [PMID: 35304243 DOI: 10.1016/j.ijpharm.2022.121667] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 01/01/2023]
Abstract
M2e VLP was previously described as a vaccine that incorporates the extracellular region of the matrix 2 protein (M2e), which is highly conserved amongst all the strains of influenza. In this study, we analyzed activation status of dendritic cells (DCs) after exposure to M2e VLP, stimulating DCs with M2e VLP and co-culturing the stimulated DCs with T cells to observe innate and adaptive immune responses. The M2e VLP microparticle was prepared by encapsulating into a polymer matrix using the one-step spray drying method. Adjuvants Alhydrogel®, MPL-A® or AddavaxTM were used to enhance the DC stimulatory effects by the M2e VLP microparticle. The M2e VLP microparticle yield was found to be 92% and the encapsulation yield was around 84% with a size of approximately 2.78 μm. There was no short-term cytotoxicity found in DCs and macrophages with concentrations up to 1500 μg/mL of M2e VLP microparticle, however long-term exposure resulted in 25% decrease in viability of cells with concentrations more than or equal to 500 μg/mL. The M2e VLP microparticle vaccine with Alhydrogel® and MPL-A® induced high levels of TNFα in both DCs and macrophages. The high levels of MHC I, II, CD28, B7-1, ICAM-1, LFA-1 expression and IL-12 release in the M2e VLP microparticle group with Alhydrogel® suggests that the M2e VLP vaccine with this adjuvant activated T cells via the Th2 pathway. The increased expression of MHC I, II, CD40, CD154, ICAM-1 and LFA-1 on DCs and the release of IL-12 in the M2e VLP microparticle culture of DCs with MPL-A® demonstrated that the M2e VLP vaccine with this adjuvant activated T cells via the Th1 pathway. The decrease in fluorescence in the Alhydrogel® and MPL-A® group illustrates the proliferation of T cells took place following exposure of DCs to the M2e VLP microparticle with these adjuvants. The M2e VLP microparticle exhibited higher stimulatory responses of DCs than the M2e VLP in suspension. Furthermore, the presence of Alhydrogel® and MPL-A® enhanced the stimulatory effects of DCs by the M2e VLP microparticle (MP) vaccine.
Collapse
Affiliation(s)
- Kimberly Braz Gomes
- Mercer University, Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, Atlanta, GA 30341, USA.
| | | | - Sucheta D'Sa
- Mercer University, Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, Atlanta, GA 30341, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Martin J D'Souza
- Mercer University, Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, Atlanta, GA 30341, USA
| |
Collapse
|
3
|
Calzas C, Mao M, Turpaud M, Viboud Q, Mettier J, Figueroa T, Bessière P, Mangin A, Sedano L, Hervé PL, Volmer R, Ducatez MF, Bourgault S, Archambault D, Le Goffic R, Chevalier C. Immunogenicity and Protective Potential of Mucosal Vaccine Formulations Based on Conserved Epitopes of Influenza A Viruses Fused to an Innovative Ring Nanoplatform in Mice and Chickens. Front Immunol 2021; 12:772550. [PMID: 34868036 PMCID: PMC8632632 DOI: 10.3389/fimmu.2021.772550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Current inactivated vaccines against influenza A viruses (IAV) mainly induce immune responses against highly variable epitopes across strains and are mostly delivered parenterally, limiting the development of an effective mucosal immunity. In this study, we evaluated the potential of intranasal formulations incorporating conserved IAV epitopes, namely the long alpha helix (LAH) of the stalk domain of hemagglutinin and three tandem repeats of the ectodomain of the matrix protein 2 (3M2e), as universal mucosal anti-IAV vaccines in mice and chickens. The IAV epitopes were grafted to nanorings, a novel platform technology for mucosal vaccination formed by the nucleoprotein (N) of the respiratory syncytial virus, in fusion or not with the C-terminal end of the P97 protein (P97c), a recently identified Toll-like receptor 5 agonist. Fusion of LAH to nanorings boosted the generation of LAH-specific systemic and local antibody responses as well as cellular immunity in mice, whereas the carrier effect of nanorings was less pronounced towards 3M2e. Mice vaccinated with chimeric nanorings bearing IAV epitopes in fusion with P97c presented modest LAH- or M2e-specific IgG titers in serum and were unable to generate a mucosal humoral response. In contrast, N-3M2e or N-LAH nanorings admixed with Montanide™ gel (MG) triggered strong specific humoral responses, composed of serum type 1/type 2 IgG and mucosal IgG and IgA, as well as cellular responses dominated by type 1/type 17 cytokine profiles. All mice vaccinated with the [N-3M2e + N-LAH + MG] formulation survived an H1N1 challenge and the combination of both N-3M2e and N-LAH nanorings with MG enhanced the clinical and/or virological protective potential of the preparation in comparison to individual nanorings. Chickens vaccinated parenterally or mucosally with N-LAH and N-3M2e nanorings admixed with Montanide™ adjuvants developed a specific systemic humoral response, which nonetheless failed to confer protection against heterosubtypic challenge with a highly pathogenic H5N8 strain. Thus, while the combination of N-LAH and N-3M2e nanorings with Montanide™ adjuvants shows promise as a universal mucosal anti-IAV vaccine in the mouse model, further experiments have to be conducted to extend its efficacy to poultry.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Chickens
- Cytokines/immunology
- Cytokines/metabolism
- Epitopes/immunology
- Female
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunity, Mucosal/drug effects
- Immunity, Mucosal/immunology
- Immunogenicity, Vaccine/immunology
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/physiology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/chemistry
- Influenza Vaccines/immunology
- Influenza in Birds/immunology
- Influenza in Birds/prevention & control
- Influenza in Birds/virology
- Mice, Inbred BALB C
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Protective Agents/administration & dosage
- Survival Analysis
- Vaccination/methods
- Mice
Collapse
Affiliation(s)
- Cynthia Calzas
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Molida Mao
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Mathilde Turpaud
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Quentin Viboud
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Joelle Mettier
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Thomas Figueroa
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Pierre Bessière
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Antoine Mangin
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
- Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Laura Sedano
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Pierre-Louis Hervé
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
- Chemistry Department, Université du Québec à Montréal, Montreal, QC, Canada
| | - Romain Volmer
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Mariette F. Ducatez
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Unité Mixte de Recherche (UMR1225), Interactions Hótes-Agents Pathogénes-Ecole Nationale Vétérinaire de Toulouse (IHAP-ENVT)-University of Toulouse, Toulouse, France
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, Montreal, QC, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Ronan Le Goffic
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| | - Christophe Chevalier
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) Molecular and Virology Unit VIM-Unité Mixte de Recherche (UMR) 892, University Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
4
|
Rapaka RR, Cross AS, McArthur MA. Using Adjuvants to Drive T Cell Responses for Next-Generation Infectious Disease Vaccines. Vaccines (Basel) 2021; 9:vaccines9080820. [PMID: 34451945 PMCID: PMC8402546 DOI: 10.3390/vaccines9080820] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Using adjuvants to drive features of T cell responses to vaccine antigens is an important technological challenge in the design of new and improved vaccines against infections. Properties such as T helper cell function, T cell memory, and CD8+ T cell cytotoxicity may play critical roles in optimal and long-lived immunity through vaccination. Directly manipulating specific immune activation or antigen delivery pathways with adjuvants may selectively augment desired T cell responses in vaccination and may improve the effectiveness and durability of vaccine responses in humans. In this review we outline recently studied adjuvants in their potential for antigen presenting cell and T cell programming during vaccination, with an emphasis on what has been observed in studies in humans as available.
Collapse
|
5
|
Jung YJ, Kim KH, Ko EJ, Lee Y, Kim MC, Lee YT, Kim CH, Jeeva S, Park BR, Kang SM. Adjuvant effects of killed Lactobacillus casei DK128 on enhancing T helper type 1 immune responses and the efficacy of influenza vaccination in normal and CD4-deficient mice. Vaccine 2020; 38:5783-5792. [PMID: 32674907 PMCID: PMC7453881 DOI: 10.1016/j.vaccine.2020.06.075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 11/23/2022]
Abstract
Lactic acid bacteria Lactobacillus casei DK128 isolated from fermented vegetable foods was suggested to stimulate innate immune responses. Here, we investigated whether heat-killed DK128 would exhibit adjuvant effects on enhancing the efficacy of influenza vaccination. Immunization of mice with split influenza virus vaccine in the presence of heat-killed DK128 induced significantly higher levels of both IgG1 and IgG2c isotype antibodies than those by vaccine only. A single dose DK128-adjuvanted influenza vaccination conferred higher efficacy of protection, as evidenced by intact lung function, less weight loss, enhanced clearance of lung viral loads, and lower levels of inflammatory cytokines and infiltrates. Immunization of CD4 T cell-knockout (CD4KO) mice with influenza vaccine and DK128, but not with vaccine alone, induced isotype-switched IgG antibodies and protection against lethal challenge in CD4KO mice. The results in this study suggest heat-killed DK128 as a potential vaccine adjuvant, promoting the induction of IgG isotype switching in CD4-deficient condition and enhancing protective efficacy of split influenza vaccination in immunocompromised and immune-competent subjects.
Collapse
Affiliation(s)
- Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; College of Veterinary Medicine, Jeju National University, Jeju, Jeju-do, Republic of Korea
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; CARESIDE Co., Ltd., Seongnam, Gyeonggi-do, Republic of Korea
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Cheol-Hyun Kim
- Department of Animal Resource Science, Dankook University, Cheonan, Chungnam, Republic of Korea
| | - Subbiah Jeeva
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
6
|
Lee J, Arun Kumar S, Jhan YY, Bishop CJ. Engineering DNA vaccines against infectious diseases. Acta Biomater 2018; 80:31-47. [PMID: 30172933 PMCID: PMC7105045 DOI: 10.1016/j.actbio.2018.08.033] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 08/14/2018] [Accepted: 08/23/2018] [Indexed: 12/30/2022]
Abstract
Engineering vaccine-based therapeutics for infectious diseases is highly challenging, as trial formulations are often found to be nonspecific, ineffective, thermally or hydrolytically unstable, and/or toxic. Vaccines have greatly improved the therapeutic landscape for treating infectious diseases and have significantly reduced the threat by therapeutic and preventative approaches. Furthermore, the advent of recombinant technologies has greatly facilitated growth within the vaccine realm by mitigating risks such as virulence reversion despite making the production processes more cumbersome. In addition, seroconversion can also be enhanced by recombinant technology through kinetic and nonkinetic approaches, which are discussed herein. Recombinant technologies have greatly improved both amino acid-based vaccines and DNA-based vaccines. A plateau of interest has been reached between 2001 and 2010 for the scientific community with regard to DNA vaccine endeavors. The decrease in interest may likely be attributed to difficulties in improving immunogenic properties associated with DNA vaccines, although there has been research demonstrating improvement and optimization to this end. Despite improvement, to the extent of our knowledge, there are currently no regulatory body-approved DNA vaccines for human use (four vaccines approved for animal use). This article discusses engineering DNA vaccines against infectious diseases while discussing advantages and disadvantages of each, with an emphasis on applications of these DNA vaccines. Statement of Significance This review paper summarizes the state of the engineered/recombinant DNA vaccine field, with a scope entailing “Engineering DNA vaccines against infectious diseases”. We endeavor to emphasize recent advances, recapitulating the current state of the field. In addition to discussing DNA therapeutics that have already been clinically translated, this review also examines current research developments, and the challenges thwarting further progression. Our review covers: recombinant DNA-based subunit vaccines; internalization and processing; enhancing immune protection via adjuvants; manufacturing and engineering DNA; the safety, stability and delivery of DNA vaccines or plasmids; controlling gene expression using plasmid engineering and gene circuits; overcoming immunogenic issues; and commercial successes. We hope that this review will inspire further research in DNA vaccine development.
Collapse
|
7
|
Virus-Like Particles Are a Superior Platform for Presenting M2e Epitopes to Prime Humoral and Cellular Immunity against Influenza Virus. Vaccines (Basel) 2018; 6:vaccines6040066. [PMID: 30241300 PMCID: PMC6313937 DOI: 10.3390/vaccines6040066] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/06/2018] [Accepted: 09/14/2018] [Indexed: 11/17/2022] Open
Abstract
Influenza virus M2 protein has a highly conserved ectodomain (M2e) as a cross-protective antigenic target. We investigated the antigenic and immunogenic properties of tandem repeat M2e (5xM2e) proteins and virus-like particles (5xM2e VLP) to better understand how VLP and protein platform vaccines induce innate and protective adaptive immune responses. Despite the high antigenic properties of 5xM2e proteins, the 5xM2e VLP was superior to 5xM2e proteins in inducing IgG2a isotype antibodies, T cell responses, plasma cells and germinal center B cells as well as in conferring cross protection. Mice primed with 5xM2e VLP were found to be highly responsive to 5xM2e protein boost, overcoming the low immunogenicity and protective efficacy of 5xM2e proteins. Immunogenic differences between VLPs and proteins in priming immune responses might be due to an intrinsic ability of 5xM2e VLP to stimulate dendritic cells secreting T helper type 1 (Th1) cytokines. We also found that 5xM2e VLP was effective in inducing inflammatory cytokines and chemokines, and in recruiting macrophages, monocytes, neutrophils, and CD11b+ dendritic cells at the injection site. Therefore, this study provides evidence that 5xM2e VLP is an effective vaccine platform, inducing cross-protection by stimulating innate and adaptive immune responses.
Collapse
|
8
|
Schepens B, De Vlieger D, Saelens X. Vaccine options for influenza: thinking small. Curr Opin Immunol 2018; 53:22-29. [PMID: 29631195 DOI: 10.1016/j.coi.2018.03.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/22/2018] [Accepted: 03/26/2018] [Indexed: 11/16/2022]
|
9
|
Qi M, Zhang XE, Sun X, Zhang X, Yao Y, Liu S, Chen Z, Li W, Zhang Z, Chen J, Cui Z. Intranasal Nanovaccine Confers Homo- and Hetero-Subtypic Influenza Protection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703207. [PMID: 29430819 DOI: 10.1002/smll.201703207] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 01/03/2018] [Indexed: 05/21/2023]
Abstract
Cross-protective and non-invasively administered vaccines are attractive and highly desired for the control of influenza. Self-assembling nanotechnology provides an opportunity for the development of vaccines with superior performance. In this study, an intranasal nanovaccine is developed targeting the conserved ectodomain of influenza matrix protein 2(M2e). 3-sequential repeats of M2e (3M2e) is presented on the self-assembling recombinant human heavy chain ferritin (rHF) cage to form the 3M2e-rHF nanoparticle. Intranasal vaccination with 3M2e-rHF nanoparticles in the absence of an adjuvant induces robust immune responses, including high titers of sera M2e-specific IgG antibodies, T-cell immune responses, and mucosal secretory-IgA antibodies in mice. The 3M2e-rHF nanoparticles also confer complete protection against a lethal infection of homo-subtypic H1N1 and hetero-subtypic H9N2 virus. An analysis of the mechanism of protection underlying the intranasal immunization with the 3M2e-rHF nanoparticle indicates that M2e-specific mucosal secretory-IgA and T-cell immune responses may play critical roles in the prevention of infection. The results suggest that the 3M2e-rHF nanoparticle is a promising, needle-free, intranasally administered, cross-protective influenza vaccine. The use of self-assembling nanovaccines could be an ideal strategy for developing vaccines with characteristics such as high immunogenicity, cross-protection, and convenient administration, as well as being economical and suitable for large-scale production.
Collapse
Affiliation(s)
- Mi Qi
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian-En Zhang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xianxun Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yanfeng Yao
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science and Technology, Wuhan, 430208, China
| | - Siling Liu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ze Chen
- College of Life Sciences, Hunan Normal University, Changsha, 410013, China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhiping Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jianjun Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, University of Chinese Academy of Sciences, Wuhan, 430071, China
| |
Collapse
|
10
|
Intranasal vaccination with M2e5x virus-like particles induces humoral and cellular immune responses conferring cross-protection against heterosubtypic influenza viruses. PLoS One 2018; 13:e0190868. [PMID: 29324805 PMCID: PMC5764335 DOI: 10.1371/journal.pone.0190868] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/21/2017] [Indexed: 11/19/2022] Open
Abstract
Current influenza vaccines do not provide broad cross-protection. Here, we report that intranasal vaccination with virus-like particles containing the highly conserved multiple ectodomains of matrix protein 2 (M2e5x VLP) of influenza virus induces broad cross-protection by M2-specific humoral and cellular immune responses. M2e5x VLP intranasal vaccination prevented severe weight loss, attenuated inflammatory cytokines and cellular infiltrates, and lowered viral loads, and induced germinal center phenotypic B and plasma cells. In addition, depletion studies demonstrate the protective roles of CD4 and CD8 T cells induced by M2e5x VLP intranasal vaccination. Thus, this study provides evidence that mucosal delivery of M2e5x VLP vaccine provides cross-protection by inducing humoral and cellular immune responses.
Collapse
|
11
|
Kim YJ, Lee YT, Kim MC, Lee YN, Kim KH, Ko EJ, Song JM, Kang SM. Cross-Protective Efficacy of Influenza Virus M2e Containing Virus-Like Particles Is Superior to Hemagglutinin Vaccines and Variable Depending on the Genetic Backgrounds of Mice. Front Immunol 2017; 8:1730. [PMID: 29276514 PMCID: PMC5727122 DOI: 10.3389/fimmu.2017.01730] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/23/2017] [Indexed: 12/30/2022] Open
Abstract
Influenza virus M2 extracellular domain (M2e) has been a target for developing cross-protective vaccines. However, the efficacy and immune correlates of M2e vaccination are poorly understood in the different host genetic backgrounds in comparison with influenza vaccines. We previously reported the cross-protective efficacy of virus-like particle (M2e5x VLP) vaccines containing heterologous tandem M2e repeats (M2e5x) derived from human, swine, and avian influenza viruses. In this study to gain better understanding of cross-protective influenza vaccines, we compared immunogenicity and efficacy of M2e5x VLP, H5 hemagglutinin VLP (HA VLP), and inactivated H3N2 virus (H3N2i) in wild-type strains of BALB/c and C57BL/6 mice, and CD4 and CD8 knockout (KO) mice. M2e5x VLP was superior to HA VLP in conferring cross-protection whereas H3N2i inactivated virus vaccine provided high efficacy of homologous protection. After M2e5x VLP vaccination and challenge, BALB/c mice induced higher IgG responses, lower lung viral loads, and less body weight loss when compared with those in C57BL/6 mice. M2e5x VLP but not H3N2i immune mice after primary challenges developed strong immunity against a secondary heterosubtypic virus as a model of future pandemics. M2e5x VLP and HA VLP vaccines were able to raise IgG isotypes in CD4 KO mice. T cells were found to contribute to cross-protection by playing a role in reducing lung viral loads. In conclusion, M2e5x VLP vaccination induced better cross-protection than HA VLP, and its efficacy varied depending on the genetic backgrounds of mice, supporting the important roles of T cells.
Collapse
Affiliation(s)
- Yu-Jin Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Young-Tae Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Min-Chul Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States.,Animal and Plant Quarantine Agency, Gimcheon, South Korea
| | - Yu-Na Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States.,Animal and Plant Quarantine Agency, Gimcheon, South Korea
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Eun-Ju Ko
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| | - Jae-Min Song
- Department of Global Medical Science, Sungshin Women's University, Seoul, South Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
12
|
Virus-like particle vaccine primes immune responses preventing inactivated-virus vaccine-enhanced disease against respiratory syncytial virus. Virology 2017; 511:142-151. [DOI: 10.1016/j.virol.2017.08.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/08/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
|
13
|
A boosting skin vaccination with dissolving microneedle patch encapsulating M2e vaccine broadens the protective efficacy of conventional influenza vaccines. J Control Release 2017. [PMID: 28642154 DOI: 10.1016/j.jconrel.2017.06.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The biodegradable microneedle patch (MNP) is a novel technology for vaccine delivery that could improve the immunogenicity of vaccines. To broaden the protective efficiency of conventional influenza vaccines, a new 4M2e-tFliC fusion protein construct containing M2e sequences from different subtypes was generated. Purified fusion protein was encapsulate into MNPs with a biocompatible polymer for use as a boosting vaccine. The results demonstrated that mice receiving a conventional inactivated vaccine followed by a skin-applied dissolving 4M2e-tFliC MNP boost could better maintain the humoral antibody response than that by the conventional vaccine-prime alone. Compared with an intramuscular injection boost, mice receiving the MNP boost showed significantly enhanced cellular immune responses, hemagglutination-inhibition (HAI) titers, and neutralization titers. Increased frequency of antigen-specific plasma cells and long-lived bone marrow plasma cells was detected in the MNP boosted group as well, indicating that skin vaccination with 4M2e-tFliC facilitated a long-term antibody-mediated immunity. The 4M2e-tFliC MNP-boosted group also possessed enhanced protection against high lethal dose challenges against homologous A/PR/8/34 and A/Aichi/2/68 viruses and protection for a majority of immunized mice against a heterologous A/California/07/2009 H1N1 virus. High levels of M2e specific immune responses were observed in the 4M2e-tFliC MNP-boosted group as well. These results demonstrate that a skin-applied 4M2e-tFliC MNP boosting immunization to seasonal vaccine recipients may be a rapid approach for increasing the protective efficacy of seasonal vaccines in response to a significant drift seen in circulating viruses. The results also provide a new perspective for future exploration of universal influenza vaccines.
Collapse
|
14
|
Watkins HC, Rappazzo CG, Higgins JS, Sun X, Brock N, Chau A, Misra A, Cannizzo JPB, King MR, Maines TR, Leifer CA, Whittaker GR, DeLisa MP, Putnam D. Safe Recombinant Outer Membrane Vesicles that Display M2e Elicit Heterologous Influenza Protection. Mol Ther 2017; 25:989-1002. [PMID: 28215994 DOI: 10.1016/j.ymthe.2017.01.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 01/02/2017] [Accepted: 01/06/2017] [Indexed: 01/10/2023] Open
Abstract
Recombinant, Escherichia coli-derived outer membrane vesicles (rOMVs), which display heterologous protein subunits, have potential as a vaccine adjuvant platform. One drawback to rOMVs is their lipopolysaccharide (LPS) content, limiting their translatability to the clinic due to potential adverse effects. Here, we explore a unique rOMV construct with structurally remodeled lipids containing only the lipid IVa portion of LPS, which does not stimulate human TLR4. The rOMVs are derived from a genetically engineered B strain of E. coli, ClearColi, which produces lipid IVa, and which was further engineered in our laboratory to hypervesiculate and make rOMVs. We report that rOMVs derived from this lipid IVa strain have substantially attenuated pyrogenicity yet retain high levels of immunogenicity, promote dendritic cell maturation, and generate a balanced Th1/Th2 humoral response. Additionally, an influenza A virus matrix 2 protein-based antigen displayed on these rOMVs resulted in 100% survival against a lethal challenge with two influenza A virus strains (H1N1 and H3N2) in mice with different genetic backgrounds (BALB/c, C57BL/6, and DBA/2J). Additionally, a two-log reduction of lung viral titer was achieved in a ferret model of influenza infection with human pandemic H1N1. The rOMVs reported herein represent a potentially safe and simple subunit vaccine delivery platform.
Collapse
Affiliation(s)
- Hannah C Watkins
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - C Garrett Rappazzo
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jaclyn S Higgins
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Xiangjie Sun
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Nicole Brock
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Annie Chau
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Aditya Misra
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Joseph P B Cannizzo
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Michael R King
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Cynthia A Leifer
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Matthew P DeLisa
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - David Putnam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
15
|
Ko EJ, Lee YT, Kim KH, Lee Y, Jung YJ, Kim MC, Lee YN, Kang T, Kang SM. Roles of Aluminum Hydroxide and Monophosphoryl Lipid A Adjuvants in Overcoming CD4+ T Cell Deficiency To Induce Isotype-Switched IgG Antibody Responses and Protection by T-Dependent Influenza Vaccine. THE JOURNAL OF IMMUNOLOGY 2016; 198:279-291. [PMID: 27881702 DOI: 10.4049/jimmunol.1600173] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/24/2016] [Indexed: 01/09/2023]
Abstract
Vaccine adjuvant effects in the CD4-deficient condition largely remain unknown. We investigated the roles of combined monophosphoryl lipid A (MPL) and aluminum hydroxide (Alum) adjuvant (MPL+Alum) in inducing immunity after immunization of CD4 knockout (CD4KO) and wild-type (WT) mice with T-dependent influenza vaccine. MPL+Alum adjuvant mediated IgG isotype-switched Abs, IgG-secreting cell responses, and protection in CD4KO mice, which were comparable to those in WT mice. In contrast, Alum adjuvant effects were dependent on CD4+ T cells. MPL+Alum adjuvant was effective in recruiting monocytes and neutrophils as well as in protecting macrophages from Alum-mediated cell loss at the injection site in CD4KO mice. MPL+Alum appeared to attenuate MPL-induced inflammatory responses in WT mice, likely improving the safety. Additional studies in CD4-depleted WT mice and MHC class II KO mice suggest that MHC class II+ APCs contribute to providing alternative B cell help in the CD4-deficient condition in the context of MPL+Alum-adjuvanted vaccination.
Collapse
Affiliation(s)
- Eun-Ju Ko
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Young-Tae Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Youri Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Yu-Jin Jung
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Min-Chul Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and.,Animal and Plant Quarantine Agency, Gimcheon 39660, Republic of Korea
| | - Yu-Na Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and.,Animal and Plant Quarantine Agency, Gimcheon 39660, Republic of Korea
| | - Taeuk Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303; and
| |
Collapse
|
16
|
Kolpe A, Schepens B, Fiers W, Saelens X. M2-based influenza vaccines: recent advances and clinical potential. Expert Rev Vaccines 2016; 16:123-136. [DOI: 10.1080/14760584.2017.1240041] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Annasaheb Kolpe
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bert Schepens
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Walter Fiers
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- Medical Biotechnology Center, VIB, Ghent, B-9052, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
17
|
Rappazzo CG, Watkins HC, Guarino CM, Chau A, Lopez JL, DeLisa MP, Leifer CA, Whittaker GR, Putnam D. Recombinant M2e outer membrane vesicle vaccines protect against lethal influenza A challenge in BALB/c mice. Vaccine 2016; 34:1252-8. [PMID: 26827663 DOI: 10.1016/j.vaccine.2016.01.028] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/01/2015] [Accepted: 01/17/2016] [Indexed: 12/20/2022]
Abstract
Currently approved influenza vaccines predominantly protect through antibodies directed against the highly variable glycoprotein hemagglutinin (HA), necessitating annual redesign and formulation based on epidemiological prediction of predominant circulating strains. More conserved influenza protein sequences, such as the ectodomain of the influenza M2 protein, or M2e, show promise as a component of a universal influenza A vaccine, but require a Th1-biased immune response for activity. Recently, recombinant, bacterially derived outer membrane vesicles (OMVs) demonstrated potential as a platform to promote a Th1-biased immune response to subunit antigens. Here, we engineer three M2e-OMV vaccines and show that all elicit strong IgG titers, with high IgG2a:IgG1 ratios, in BALB/c mice. Additionally, the administration of one M2e-OMV construct containing tandem heterologous M2e peptides (M2e4xHet-OMV) resulted in 100% survival against lethal doses of the mouse-adapted H1N1 influenza strain PR8. Passive transfer of antibodies from M2e4xHet-OMV vaccinated mice to unvaccinated mice also resulted in 100% survival to challenge, indicating that protection is driven largely via antibody-mediated immunity. The potential mechanism through which M2e-OMVs initiated the immune response was explored and it was found that the constructs triggered TLR1/2, TLR4, and TLR5. Our data indicate that OMVs have potential as a platform for influenza A vaccine development due to their unique adjuvant profile and intrinsic pathogen-mimetic nature.
Collapse
Affiliation(s)
- C Garrett Rappazzo
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Hannah C Watkins
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Cassandra M Guarino
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Annie Chau
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jody L Lopez
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Matthew P DeLisa
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Cynthia A Leifer
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - David Putnam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
18
|
Abstract
The development of vaccines that could provide broad protection against antigenically variant influenza viruses has long been the ultimate prize in influenza research. Recent developments have pushed us closer to this goal, and such vaccines may now be within reach. This brief review outlines the current approaches to broadly protective vaccines, and the probable hurdles and roadblocks to achieving this goal.
Collapse
Affiliation(s)
- John Jay Treanor
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Box 689, 601 Elmwood Avenue, Rochester, NY 14642, United States.
| |
Collapse
|
19
|
Prospects for broadly protective influenza vaccines. Vaccine 2015; 33 Suppl 4:D39-45. [DOI: 10.1016/j.vaccine.2015.08.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/02/2015] [Accepted: 08/03/2015] [Indexed: 12/14/2022]
|
20
|
The application of virus-like particles as vaccines and biological vehicles. Appl Microbiol Biotechnol 2015; 99:10415-32. [PMID: 26454868 PMCID: PMC7080154 DOI: 10.1007/s00253-015-7000-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/01/2015] [Accepted: 09/04/2015] [Indexed: 01/04/2023]
Abstract
Virus-like particles (VLPs) can be spontaneously self-assembled by viral structural proteins under appropriate conditions in vitro while excluding the genetic material and potential replication probability. In addition, VLPs possess several features including can be rapidly produced in large quantities through existing expression systems, highly resembling native viruses in terms of conformation and appearance, and displaying repeated cluster of epitopes. Their capsids can be modified via genetic insertion or chemical conjugation which facilitating the multivalent display of a homologous or heterogeneous epitope antigen. Therefore, VLPs are considered as a safe and effective candidate of prophylactic and therapeutic vaccines. VLPs, with a diameter of approximately 20 to 150 nm, also have the characteristics of nanometer materials, such as large surface area, surface-accessible amino acids with reactive moieties (e.g., lysine and glutamic acid residues), inerratic spatial structure, and good biocompatibility. Therefore, assembled VLPs have great potential as a delivery system for specifically carrying a variety of materials. This review summarized recent researches on VLP development as vaccines and biological vehicles, which demonstrated the advantages and potential of VLPs in disease control and prevention and diagnosis. Then, the prospect of VLP biology application in the future is discussed as well.
Collapse
|
21
|
Beljanski V, Chiang C, Kirchenbaum GA, Olagnier D, Bloom CE, Wong T, Haddad EK, Trautmann L, Ross TM, Hiscott J. Enhanced Influenza Virus-Like Particle Vaccination with a Structurally Optimized RIG-I Agonist as Adjuvant. J Virol 2015; 89:10612-24. [PMID: 26269188 PMCID: PMC4580177 DOI: 10.1128/jvi.01526-15] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The molecular interaction between viral RNA and the cytosolic sensor RIG-I represents the initial trigger in the development of an effective immune response against infection with RNA viruses, resulting in innate immune activation and subsequent induction of adaptive responses. In the present study, the adjuvant properties of a sequence-optimized 5'-triphosphate-containing RNA (5'pppRNA) RIG-I agonist (termed M8) were examined in combination with influenza virus-like particles (VLP) (M8-VLP) expressing H5N1 influenza virus hemagglutinin (HA) and neuraminidase (NA) as immunogens. In combination with VLP, M8 increased the antibody response to VLP immunization, provided VLP antigen sparing, and protected mice from a lethal challenge with H5N1 influenza virus. M8-VLP immunization also led to long-term protective responses against influenza virus infection in mice. M8 adjuvantation of VLP increased endpoint and antibody titers and inhibited influenza virus replication in lungs compared with approved or experimental adjuvants alum, AddaVax, and poly(I·C). Uniquely, immunization with M8-VLP stimulated a TH1-biased CD4 T cell response, as determined by increased TH1 cytokine levels in CD4 T cells and increased IgG2 levels in sera. Collectively, these data demonstrate that a sequence-optimized, RIG-I-specific agonist is a potent adjuvant that can be utilized to increase the efficacy of influenza VLP vaccination and dramatically improve humoral and cellular mediated protective responses against influenza virus challenge. IMPORTANCE The development of novel adjuvants to increase vaccine immunogenicity is an important goal that seeks to improve vaccine efficacy and ultimately prevent infections that endanger human health. This proof-of-principle study investigated the adjuvant properties of a sequence-optimized 5'pppRNA agonist (M8) with enhanced capacity to stimulate antiviral and inflammatory gene networks using influenza virus-like particles (VLP) expressing HA and NA as immunogens. Vaccination with VLP in combination with M8 increased anti-influenza virus antibody titers and protected animals from lethal influenza virus challenge, highlighting the potential clinical use of M8 as an adjuvant in vaccine development. Altogether, the results describe a novel immunostimulatory agonist targeted to the cytosolic RIG-I sensor as an attractive vaccine adjuvant candidate that can be used to increase vaccine efficacy, a pressing issue in children and the elderly population.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Animals
- Antibodies, Viral/biosynthesis
- DEAD Box Protein 58
- DEAD-box RNA Helicases/chemistry
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/immunology
- Dendritic Cells/immunology
- Dendritic Cells/virology
- Female
- HEK293 Cells
- Hemagglutinins, Viral/chemistry
- Hemagglutinins, Viral/genetics
- Hemagglutinins, Viral/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunization
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice
- Mice, Inbred BALB C
- Neuraminidase/chemistry
- Neuraminidase/genetics
- Neuraminidase/immunology
- Oligoribonucleotides/administration & dosage
- Oligoribonucleotides/genetics
- Oligoribonucleotides/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/mortality
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Primary Cell Culture
- Receptors, Immunologic
- Survival Analysis
- Th1-Th2 Balance/drug effects
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Vladimir Beljanski
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Cindy Chiang
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Greg A Kirchenbaum
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - David Olagnier
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Chalise E Bloom
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Terianne Wong
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Elias K Haddad
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Lydie Trautmann
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Ted M Ross
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - John Hiscott
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| |
Collapse
|
22
|
Lee YN, Kim MC, Lee YT, Hwang HS, Lee J, Kim C, Kang SM. Cross Protection against Influenza A Virus by Yeast-Expressed Heterologous Tandem Repeat M2 Extracellular Proteins. PLoS One 2015; 10:e0137822. [PMID: 26366729 PMCID: PMC4569442 DOI: 10.1371/journal.pone.0137822] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 08/24/2015] [Indexed: 01/08/2023] Open
Abstract
The influenza M2 ectodomain (M2e) is well conserved across human influenza A subtypes, but there are few residue changes among avian and swine origin influenza A viruses. We expressed a tandem repeat construct of heterologous M2e sequences (M2e5x) derived from human, swine, and avian origin influenza A viruses using the yeast expression system. Intramuscular immunization of mice with AS04-adjuvanted M2e5x protein vaccines was effective in inducing M2e-specific antibodies reactive to M2e peptide and native M2 proteins on the infected cells with human, swine, or avian influenza virus, mucosal and systemic memory cellular immune responses, and cross-protection against H3N2 virus. Importantly, M2e5x immune sera were found to confer protection against different subtypes of H1N1 and H5N1 influenza A viruses in naïve mice. Also, M2e5x-immune complexes of virus-infected cells stimulated macrophages to secrete cytokines via Fc receptors, indicating a possible mechanism of protection. The present study provides evidence that M2e5x proteins produced in yeast cells could be developed as a potential universal influenza vaccine.
Collapse
Affiliation(s)
- Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, United States of America
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, United States of America
- Animal and Plant Quarantine Agency, Anyang, Gyeonggi-do, South Korea
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, United States of America
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, United States of America
| | - Jongsang Lee
- BEAMS Biotechnology Co. Ltd., Seongnam, Gyeonggi-do, South Korea
| | - Cheol Kim
- BEAMS Biotechnology Co. Ltd., Seongnam, Gyeonggi-do, South Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, United States of America
- * E-mail:
| |
Collapse
|
23
|
Lee YN, Kim MC, Lee YT, Kim YJ, Lee J, Kim C, Ha SH, Kang SM. Co-immunization with tandem repeat heterologous M2 extracellular proteins overcomes strain-specific protection of split vaccine against influenza A virus. Antiviral Res 2015; 122:82-90. [PMID: 26248203 DOI: 10.1016/j.antiviral.2015.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 01/28/2023]
Abstract
Current influenza vaccines are less efficacious against antigenically different influenza A viruses. This study presents an approach to overcome strain-specific protection, using a strategy of co-immunization with seasonal H3N2 split vaccine and yeast-expressed soluble proteins of a tandem repeat containing heterologous influenza M2 ectodomains (M2e5x). Co-immunization with both vaccines in mice was superior to either vaccine alone in inducing cross protection against heterologous H3N2 virus by raising M2e-specific humoral and cellular immune responses toward a T-helper type 1 profile inducing IgG2a isotype antibodies as well as interferon-γ-producing cells in systemic and mucosal sites. In addition, co-immunization sera were found to confer cross-protection against different subtypes of H1N1 and H5N1 influenza A viruses in naïve mice. A mechanistic study provides evidence that activation of dendritic cells by co-stimulation with M2e5x and split vaccine was associated with the proliferation of CD4(+) T cells. Our results suggest that a strategy of co-immunization with seasonal split and M2e5x protein vaccines could be a promising approach for overcoming the limitation of strain-specific protection by current influenza vaccination.
Collapse
Affiliation(s)
- Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Animal and Plant Quarantine Agency, 175 Anyangro, Anyang, Gyeonggi-do 430-757, Republic of Korea
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Jongsang Lee
- BEAMS Biotechnology Co. Ltd., Seongnam, Gyeonggi-do, Republic of Korea
| | - Cheol Kim
- BEAMS Biotechnology Co. Ltd., Seongnam, Gyeonggi-do, Republic of Korea
| | - Suk-Hoon Ha
- Mogam Biotechnology Research Institute, Yongin, Gyeonggi-do, Republic of Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
24
|
Abstract
Emerflu is an inactivated, split-virion pandemic preparedness vaccine, containing 30 μg of hemagglutinin (HA) and 600 μg of aluminum hydroxide adjuvant. It is administered in two doses, 3 weeks apart. Only moderate immunogenicity was evident from clinical studies with the vaccine in adults, and HA antibody responses were below the criteria established by the EMA and US FDA for licensure. With the exception of Australia, the vaccine remains unlicensed. Further clinical development appears to have been suspended, and newer adjuvants such as MF59 and AS03 have since demonstrated safety and superior immunogenicity with lower HA doses. Emerflu is symbolic of the failure of aluminum salts as an adjuvant for influenza vaccines. Reasons for this failure are unclear, and may reflect problems with the adjuvant-antigen complex or interference in the immune response by heterosubtypic immunity.
Collapse
Affiliation(s)
- Barnaby E Young
- Communicable Diseases Centre, Institute of Infectious Diseases and Epidemiology, Communicable Diseases Centre, 144 Moulmein Road, Singapore, Singapore
| | | | | |
Collapse
|
25
|
Prospects of HA-based universal influenza vaccine. BIOMED RESEARCH INTERNATIONAL 2015; 2015:414637. [PMID: 25785268 PMCID: PMC4345066 DOI: 10.1155/2015/414637] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 12/23/2014] [Indexed: 12/02/2022]
Abstract
Current influenza vaccines afford substantial protection in humans by inducing strain-specific neutralizing antibodies (Abs). Most of these Abs target highly variable immunodominant epitopes in the globular domain of the viral hemagglutinin (HA). Therefore, current vaccines may not be able to induce heterosubtypic immunity against the divergent influenza subtypes. The identification of broadly neutralizing Abs (BnAbs) against influenza HA using recent technological advancements in antibody libraries, hybridoma, and isolation of single Ab-secreting plasma cells has increased the interest in developing a universal influenza vaccine as it could provide life-long protection. While these BnAbs can serve as a source for passive immunotherapy, their identification represents an important step towards the design of such a universal vaccine. This review describes the recent advances and approaches used in the development of universal influenza vaccine based on highly conserved HA regions identified by BnAbs.
Collapse
|
26
|
Deng L, Cho KJ, Fiers W, Saelens X. M2e-Based Universal Influenza A Vaccines. Vaccines (Basel) 2015; 3:105-36. [PMID: 26344949 PMCID: PMC4494237 DOI: 10.3390/vaccines3010105] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 12/23/2014] [Accepted: 01/30/2015] [Indexed: 12/13/2022] Open
Abstract
The successful isolation of a human influenza virus in 1933 was soon followed by the first attempts to develop an influenza vaccine. Nowadays, vaccination is still the most effective method to prevent human influenza disease. However, licensed influenza vaccines offer protection against antigenically matching viruses, and the composition of these vaccines needs to be updated nearly every year. Vaccines that target conserved epitopes of influenza viruses would in principle not require such updating and would probably have a considerable positive impact on global human health in case of a pandemic outbreak. The extracellular domain of Matrix 2 (M2e) protein is an evolutionarily conserved region in influenza A viruses and a promising epitope for designing a universal influenza vaccine. Here we review the seminal and recent studies that focused on M2e as a vaccine antigen. We address the mechanism of action and the clinical development of M2e-vaccines. Finally, we try to foresee how M2e-based vaccines could be implemented clinically in the future.
Collapse
Affiliation(s)
- Lei Deng
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Ki Joon Cho
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Walter Fiers
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Xavier Saelens
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| |
Collapse
|
27
|
Sun X, Wang Y, Dong C, Hu J, Yang L. High copy numbers and N terminal insertion position of influenza A M2E fused with hepatitis B core antigen enhanced immunogenicity. Biosci Trends 2015; 9:221-7. [DOI: 10.5582/bst.2015.01060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Xincheng Sun
- Basic Medical School of Zhengzhou University
- College of Food and Biological Engineering, Zhengzhou University of Light Industry
| | - Yunlong Wang
- Basic Medical School of Zhengzhou University
- Bioengineering Research Center of Henan Province
- Henan Biotechnology Research Centre
| | - Caiwen Dong
- College of Food and Biological Engineering, Zhengzhou University of Light Industry
| | - Jinqiang Hu
- College of Food and Biological Engineering, Zhengzhou University of Light Industry
| | - Liping Yang
- Basic Medical School of Henan University of Traditional Chinese Medicine
| |
Collapse
|
28
|
Hadifar F, Ignjatovic J, Tarigan S, Indriani R, Ebrahimie E, Hasan NH, McWhorter A, Putland S, Ownagh A, Hemmatzadeh F. Multimeric recombinant M2e protein-based ELISA: a significant improvement in differentiating avian influenza infected chickens from vaccinated ones. PLoS One 2014; 9:e108420. [PMID: 25330391 PMCID: PMC4199618 DOI: 10.1371/journal.pone.0108420] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 08/28/2014] [Indexed: 02/07/2023] Open
Abstract
Killed avian influenza virus (AIV) vaccines have been used to control H5N1 infections in countries where the virus is endemic. Distinguishing vaccinated from naturally infected birds (DIVA) in such situations however, has become a major challenge. Recently, we introduced the recombinant ectodomain of the M2 protein (M2e) of H5N1 subtype as a novel tool for an ELISA based DIVA test. Despite being antigenic in natural infection the monomer form of the M2e used in ELISA had limited antigenicity and consequently poor diagnostic capability. To address this shortcoming, we evaluated the use of four tandem copies of M2e (tM2e) for increased efficiency of M2e antibody detection. The tM2e gene of H5N1 strain from Indonesia (A/Indonesia/CDC540/2006) was cloned into a pMAL- p4x expression vector and expressed in E.coli as a recombinant tM2e-MBP or M2e-MBP proteins. Both of these, M2e and tM2e antigens reacted with sera obtained from chickens following live H5N1 infection but not with sera from vaccinated birds. A significantly stronger M2e antibody reaction was observed with the tM2e compared to M2e antigen. Western blotting also supported the superiority of tM2e over M2e in detection of specific M2e antibodies against live H5N1 infection. Results from this study demonstrate that M2e tetramer is a better antigen than single M2e and could be more suitable for an ELISA based DIVA test.
Collapse
Affiliation(s)
- Farshid Hadifar
- School of Animal and Veterinary Sciences, The University of Adelaide, South Australia, Adelaide, Australia
| | - Jagoda Ignjatovic
- School of Veterinary Science, The University of Melbourne, Melbourne, Victoria, Australia
| | - Simson Tarigan
- Indonesian Research Centre for Veterinary Science, Bogor, West Java, Indonesia
| | - Risa Indriani
- Indonesian Research Centre for Veterinary Science, Bogor, West Java, Indonesia
| | - Esmaeil Ebrahimie
- School of Molecular and Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Noor Haliza Hasan
- School of Animal and Veterinary Sciences, The University of Adelaide, South Australia, Adelaide, Australia
| | - Andrea McWhorter
- School of Animal and Veterinary Sciences, The University of Adelaide, South Australia, Adelaide, Australia
| | - Sophie Putland
- School of Animal and Veterinary Sciences, The University of Adelaide, South Australia, Adelaide, Australia
| | | | - Farhid Hemmatzadeh
- School of Animal and Veterinary Sciences, The University of Adelaide, South Australia, Adelaide, Australia
- * E-mail:
| |
Collapse
|
29
|
Kim MC, Lee YN, Hwang HS, Lee YT, Ko EJ, Jung YJ, Cho MK, Kim YJ, Lee JS, Ha SH, Kang SM. Influenza M2 virus-like particles confer a broader range of cross protection to the strain-specific pre-existing immunity. Vaccine 2014; 32:5824-31. [PMID: 25171841 DOI: 10.1016/j.vaccine.2014.08.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/05/2014] [Accepted: 08/15/2014] [Indexed: 01/30/2023]
Abstract
Immunity in humans with annual vaccination does not provide effective protection against antigenically distinct strains. As an approach to improve cross-protection in the presence of pre-existing strain-specific immunity, we investigated the efficacy of heterologous and heterosubtypic protection in previously vaccinated mice at earlier times after subsequent immunization with conserved-antigenic target influenza M2 ectodomain (M2e) virus-like particle vaccine (M2e5× VLP). Immunization of mice with H1N1 split vaccine induced virus specific antibodies to homologous influenza virus but did not provide heterosubtypic hemagglutination inhibiting antibody responses and cross-protection. However, subsequent M2e5× VLP immunization induced an M2e specific antibody response as well as interferon-γ (IFN-γ) producing cells in systemic and mucosal sites. Upon lethal challenge with H3N2 or H5N1 subtype influenza viruses, subsequently immunized mice with M2e5× VLP were well protected against heterosubtypic influenza viruses. These results provide evidence that non-seasonal immunization with M2e5× VLP, an experimental candidate for universal vaccine, is a promising approach for broadening the cross-protection even in the presence of strain-specific immunity.
Collapse
Affiliation(s)
- Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Min Kyoung Cho
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Jong Seok Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Suk-Hoon Ha
- Mogam Biotechnology Research Institute, Yongin-si, Gyeonggi-do, Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|