1
|
Calzas C, Alkie TN, Suderman M, Embury-Hyatt C, Khatri V, Le Goffic R, Berhane Y, Bourgault S, Archambault D, Chevalier C. M2e nanovaccines supplemented with recombinant hemagglutinin protect chickens against heterologous HPAI H5N1 challenge. NPJ Vaccines 2024; 9:161. [PMID: 39237609 PMCID: PMC11377767 DOI: 10.1038/s41541-024-00944-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
Current poultry vaccines against influenza A viruses target the globular head region of the hemagglutinin (HA1), providing limited protection against antigenically divergent strains. Experimental subunit vaccines based on the conserved ectodomain of the matrix protein 2 (M2e) induce cross-reactive antibody responses, but fail to fully prevent virus shedding after low pathogenic avian influenza (LPAI) virus challenge, and are ineffective against highly pathogenic avian influenza (HPAI) viruses. This study assessed the benefits of combining nanoparticles bearing three tandem M2e repeats (NR-3M2e nanorings or NF-3M2e nanofilaments) with an HA1 subunit vaccine in protecting chickens against a heterologous HPAI H5N1 virus challenge. Chickens vaccinated with the combined formulations developed M2e and HA1-specific antibodies, were fully protected from clinical disease and mortality, and showed no histopathological lesions or virus shedding, unlike those given only HA1, NR-3M2e, or NF-3M2e. Thus, the combined vaccine formulations provided complete cross-protection against HPAI H5N1 virus, and prevented environmental virus shedding, crucial for controlling avian influenza outbreaks.
Collapse
Affiliation(s)
- Cynthia Calzas
- INRAE, UVSQ, UMR892 VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Tamiru N Alkie
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Matthew Suderman
- Public Health Agency of Canada, National Microbiology Laboratory, Winnipeg, Canada
| | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Vinay Khatri
- Chemistry Department, Université du Québec à Montréal, Montréal, QC, Canada
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, QC, Canada
| | - Ronan Le Goffic
- INRAE, UVSQ, UMR892 VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Yohannes Berhane
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, Canada
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, Montréal, QC, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, QC, Canada.
| | | |
Collapse
|
2
|
Santos‐Beneit F. What is the role of microbial biotechnology and genetic engineering in medicine? Microbiologyopen 2024; 13:e1406. [PMID: 38556942 PMCID: PMC10982607 DOI: 10.1002/mbo3.1406] [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: 01/12/2024] [Revised: 02/26/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
Microbial products are essential for developing various therapeutic agents, including antibiotics, anticancer drugs, vaccines, and therapeutic enzymes. Genetic engineering techniques, functional genomics, and synthetic biology unlock previously uncharacterized natural products. This review highlights major advances in microbial biotechnology, focusing on gene-based technologies for medical applications.
Collapse
Affiliation(s)
- Fernando Santos‐Beneit
- Institute of Sustainable ProcessesValladolidSpain
- Department of Chemical Engineering and Environmental Technology, School of Industrial EngineeringUniversity of ValladolidValladolidSpain
| |
Collapse
|
3
|
Jawalagatti V, Kirthika P, Lee JH. Oral mRNA Vaccines Against Infectious Diseases- A Bacterial Perspective [Invited]. Front Immunol 2022; 13:884862. [PMID: 35592330 PMCID: PMC9110646 DOI: 10.3389/fimmu.2022.884862] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/11/2022] [Indexed: 01/04/2023] Open
Abstract
The mRNA vaccines from Pfizer/BioNTech and Moderna were granted emergency approval in record time in the history of vaccinology and played an instrumental role in limiting the pandemic caused by SARS-CoV-2. The success of these vaccines resulted from over 3 decades of research from many scientists. However, the development of orally administrable mRNA vaccine development is surprisingly underexplored. Our group specializing in Salmonella-based vaccines explored the possibility of oral mRNA vaccine development. Oral delivery was made possible by the exploitation of the Semliki Forest viral replicon and Salmonella vehicle for transgene amplification and gene delivery, respectively. Herein we highlight the prospect of developing oral replicon-based mRNA vaccines against infectious diseases based on our recent primary studies on SARS-CoV-2. Further, we discuss the potential advantages and limitations of bacterial gene delivery.
Collapse
Affiliation(s)
| | | | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
4
|
Alqazlan N, Astill J, Raj S, Sharif S. Strategies for enhancing immunity against avian influenza virus in chickens: A review. Avian Pathol 2022; 51:211-235. [PMID: 35297706 DOI: 10.1080/03079457.2022.2054309] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Poultry infection with avian influenza viruses (AIV) is a continuous source of concern for poultry production and human health. Uncontrolled infection and transmission of AIV in poultry increases the potential for viral mutation and reassortment, possibly resulting in the emergence of zoonotic viruses. To this end, implementing strategies to disrupt the transmission of AIVs in poultry, including a wide array of traditional and novel methods, is much needed. Vaccination of poultry is a targeted approach to reduce clinical signs and shedding in infected birds. Strategies aimed at enhancing the effectiveness of AIV vaccines are multi-pronged and include methods directed towards eliciting immune responses in poultry. Strategies include producing vaccines of greater immunogenicity via vaccine type and adjuvant application and increasing bird responsiveness to vaccines by modification of the gastrointestinal tract (GIT) microbiome and dietary interventions. This review provides an in-depth discussion of recent findings surrounding novel AIV vaccines for poultry, including reverse genetics vaccines, vectors, protein vaccines and virus like particles, highlighting their experimental efficacy among other factors such as safety and potential for use in the field. In addition to the type of vaccine employed, vaccine adjuvants also provide an effective way to enhance AIV vaccine efficacy, therefore, research on different types of vaccine adjuvants and vaccine adjuvant delivery strategies is discussed. Finally, the poultry gastrointestinal microbiome is emerging as an important factor in the effectiveness of prophylactic treatments. In this regard, current findings on the effects of the chicken GIT microbiome on AIV vaccine efficacy are summarized here.
Collapse
Affiliation(s)
- Nadiyah Alqazlan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jake Astill
- Artemis Technologies Inc., Guelph, ON, N1L 1E3, Canada
| | - Sugandha Raj
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
5
|
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
|
6
|
Senevirathne A, Hewawaduge C, Lee JH. Genetic interference exerted by Salmonella-delivered CRISPR/Cas9 significantly reduces the pathological burden caused by Marek's disease virus in chickens. Vet Res 2021; 52:125. [PMID: 34593043 PMCID: PMC8482593 DOI: 10.1186/s13567-021-00995-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/09/2021] [Indexed: 11/29/2022] Open
Abstract
Efficient in vivo delivery of a CRISPR/Cas9 plasmid is of paramount importance for effective therapy. Here, we investigated the usability of Salmonella as a plasmid carrier for in vivo therapy against virus-induced cancer using Marek’s disease virus (MDV) as a model for study in chickens. A green fluorescent protein-expressing CRISPR/Cas9 plasmid encoding the virulence gene pp38 was constructed against Marek’s disease virus. Therapeutic plasmids were transformed into Salmonella carrying lon and sifA gene deletions. The animals in 5 groups were intraperitoneally inoculated with phosphate-buffered saline, vector control, or Salmonella before or after MDV infection, or left uninfected as a naïve control. Therapeutic effectiveness was evaluated by observing disease outcomes and the viral copy number in peripheral blood mononuclear cells. The efficacy of plasmid delivery by Salmonella was 13 ± 1.7% in the spleen and 8.0 ± 1.8% in the liver on the 6th day post-infection. The Salmonella-treated groups showed significant resistance to MDV infection. The maximum effect was observed in the group treated with Salmonella before MDV infection. None of the chickens fully recovered; however, the results suggested that timely delivery of Salmonella could be effective for in vivo CRISPR/Cas9-mediated genetic interference against highly pathogenic MDV. The use of Salmonella in CRISPR systems provides a simpler and more efficient platform for in vivo therapy with CRISPR than the use of conventional in vivo gene delivery methods and warrants further development.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea.
| |
Collapse
|
7
|
Hajam IA, Kirthika P, Hewawaduge C, Jawalagatti V, Park S, Senevirathne A, Lee JH. Oral immunization with an attenuated Salmonella Gallinarum encoding the H9N2 haemagglutinin and M2 ectodomain induces protective immune responses against H9N2 infection in chickens. Avian Pathol 2020; 49:486-495. [PMID: 32483989 DOI: 10.1080/03079457.2020.1775782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
H9N2, a low pathogenic avian influenza virus, causes significant economic losses in the poultry industry worldwide. Herein, we describe the construction of an attenuated Salmonella Gallinarum (SG) strain for expression and delivery of H9N2 haemagglutinin (HA) 1 (SG-HA1), HA2 (SG-HA2) and/or the conserved matrix protein 2 ectodomain (SG-M2e). We demonstrated that recombinant SG strains expressing HA1, HA2 and M2e antigens were immunogenic and safe in a chicken model. Chickens (n = 8) were vaccinated once orally with SG alone, SG-HA1, SG-HA2, SG-M2e, or mixture of SG-HA1, SG-HA2 and SG-M2e, or vaccinated once intramuscularly with an oil-adjuvant inactivated H9N2 vaccine. Our results demonstrated that vaccination with SG mutants encoding influenza antigens, administered individually or as a mixture, elicited significantly (P < 0.05) greater antigen-specific humoral and cell-mediated immune responses in chickens compared with those vaccinated with SG alone. A conventional H9N2 vaccine induced significantly (P < 0.05) greater HA1 and HA2 antibody responses than SG-based H9N2 vaccine strains, but significantly (P < 0.05) less robust M2e-specific responses. Upon challenge with the virulent H9N2 virus on day 28 post-vaccination, chickens vaccinated with either the SG-based H9N2 or conventional H9N2 vaccines exhibited comparable lung inflammation and viral loads, although both were significantly lower (P < 0.05) than in the group vaccinated with SG alone. In conclusion, our results showed that SG-based vaccination stimulated efficient immune responses against virulent H9N2. Further studies are needed to fully develop this approach as a preventive strategy for low pathogenic avian influenza viruses affecting poultry. RESEARCH HIGHLIGHTS S. gallinarum expressing HA1, HA2 and M2e antigens are immunogenic and safe. Salmonella has dual function of acting as a delivery system and as a natural adjuvant. Vaccine constructs elicit specific humoral and cell-mediated immune responses.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| | - Perumalraja Kirthika
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| | | | - SungWoo Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| |
Collapse
|
8
|
Hajam IA, Senevirathne A, Hewawaduge C, Kim J, Lee JH. Intranasally administered protein coated chitosan nanoparticles encapsulating influenza H9N2 HA2 and M2e mRNA molecules elicit protective immunity against avian influenza viruses in chickens. Vet Res 2020; 51:37. [PMID: 32143695 PMCID: PMC7060564 DOI: 10.1186/s13567-020-00762-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 02/17/2020] [Indexed: 12/31/2022] Open
Abstract
Chitosan nanoparticles (CNPs) represent an efficient vaccination tool to deliver immunogenic antigens to the antigen-presenting cells (APCs), which subsequently stimulate protective immune responses against infectious diseases. Herein, we prepared CNPs encapsulating mRNA molecules followed by surface coating with conserved H9N2 HA2 and M2e influenza proteins. We demonstrated that CNPs efficiently delivered mRNA molecules into APCs and had effectively penetrated the mucosal barrier to reach to the immune initiation sites. To investigate the potential of CNPs delivering influenza antigens to stimulate protective immunity, we intranasally vaccinated chickens with empty CNPs, CNPs delivering HA2 and M2e in both mRNA and protein formats (CNPs + RNA + Pr) or CNPs delivering antigens in protein format only (CNPs + Pr). Our results demonstrated that chickens vaccinated with CNPs + RNA + Pr elicited significantly (p < 0.05) higher systemic IgG, mucosal IgA antibody responses and cellular immune responses compared to the CNPs + Pr vaccinated group. Consequently, upon challenge with either H7N9 or H9N2 avian influenza viruses (AIVs), efficient protection, in the context of viral load and lung pathology, was observed in chickens vaccinated with CNPs + RNA + Pr than CNPs + Pr vaccinated group. In conclusion, we show that HA2 and M2e antigens elicited a broad spectrum of protection against AIVs and incorporation of mRNAs in vaccine formulation is an effective strategy to induce superior immune responses.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Chamit Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Jehyoung Kim
- College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, Republic of Korea.
| |
Collapse
|
9
|
Zhao N, Lv J, Lu Y, Jiang Y, Li H, Liu Y, Zhang X, Zhao X. Prolonging and enhancing the protective efficacy of the EtMIC3-C-MAR against eimeria tenella through delivered by attenuated salmonella typhimurium. Vet Parasitol 2020; 279:109061. [PMID: 32143014 DOI: 10.1016/j.vetpar.2020.109061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 01/09/2023]
Abstract
The microneme adhesive repeats (MAR) of Eimeria tenella microneme protein 3 (EtMIC3) are associated with binding to and invasion of host cells. Adhesion and invasion-related proteins or domains are often strongly immunogenic, immune responses mounted against these factors that play a key role in blocking invasion. In the present study, an oral live vaccine consisting of attenuated Salmonella typhimurium X4550 carrying two MAR domains fragment (St-X4550-MAR) was constructed and its protective efficacies were evaluated. The results showed that St-X4550-MAR was more immunogenic and conferred a higher degree of protection than recombinant MAR polypeptide as reflected by increased body weight, decreased oocyst shedding and lesion scores, increased serum IgG and cecal sIgA antibody production, and increasing levels of interferon-γ and interleukin-10. Thus, MAR domains are highly immunogenic and St-X4550-MAR had moderate activity against E. tenella infection by stimulating humoral, mucosal and cellular immunity. Chickens immunized with our constructed live vaccine provided considerable protections as early as at 10 d post-immunization (ACI: 155.17), and maintained higher protection levels at 20 d post-immunization (ACI: 173.66), and at 30 d post-immunization (ACI: 162.4). While the protective efficacy of chickens immunized with the recombinant MAR peptides showed a decreased trend as the post immunization time prolonging. Thus, using live-attenuated S. typhimurium X4550 as a vaccine expression and delivery system can significantly improve the protective efficacy and duration of protective immunity of MAR of EtMIC3.
Collapse
Affiliation(s)
- Ningning Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Junfeng Lv
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Yaru Lu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Yingying Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Hongmei Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China
| | - Yumin Liu
- Shandong Huamutianyuan Agriculture and Animal Husbandry Co., Ltd., 1 Gangxing 3 Road, Jinan, Shandong Province, 250101, China
| | - Xiao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China.
| | - Xiaomin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Taian City, Shandong Province, 271018, China.
| |
Collapse
|
10
|
Kim J, Hajam IA, Lee JH. Human antigen presenting cells stimulated with Salmonella delivered influenza antigens induce cytokine production and proliferation of human CD4 + T cells in vitro. J Immunol Methods 2019; 470:20-26. [PMID: 31028753 DOI: 10.1016/j.jim.2019.04.006] [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: 12/26/2018] [Revised: 03/19/2019] [Accepted: 04/23/2019] [Indexed: 11/15/2022]
Abstract
This study aimed to investigate whether the human antigen presenting cells (APCs) can process and present Salmonella expressing H7N9 hemagglutinin (Sal-HA), neuraminidase (Sal-NA) or M2 ectodomain (Sal-M2e) to T cells and subsequently activate CD4+ T cell responses in vitro. In this study, APCs generated from human peripheral blood mononuclear cells (PBMCs) were first treated with mitomycin-C, followed by stimulation with Sal-HA, Sal-M2e, Sal-NA or Salmonella alone for 24 h. Subsequently, stimulated APCs were coincubated with untreated PBMCs (1:10) of the same individual for 24 or 72 h and then analysed for cytokine induction and T cell proliferations by qRT-PCR assay and flow cytometry, respectively. Our results demonstrated that APCs stimulated with Sal-HA, Sal-M2e or Sal-NA induced significantly (p < .05) higher CD3+CD4+ T cell proliferations compared to the APCs treated with Salmonella alone. Our data further revealved that APCs treated with Sal-HA induced significantly (p < .05) higher CD3+CD4+ T cell responses compared to the APCs treated with either Sal-M2e or Sal-NA, which both induced almost comparable levels. The T cell proliferation responses were further measured by lymphocyte proliferation assay and the results showed that Sal-HA and Sal-M2e stimulated APCs induced significantly (p < .05) higher proliferations in T cells compared to the APCs stimulated with either Sal-NA or Salmonella alone. With respect to cytokine inductions, APCs treated with either Sal-HA or Sal-M2e induced significantly (p < .05) higher mRNA transcription levels of proinflammatory (IL-1β, IL-6, IL-12 and IL-23), Th1 (IFN-γ), Th17 (IL-17 and IL-21) and Th2 (IL-10 and TGF-β) cytokines in T cells compared to Sal-NA or Salmonella alone treated APCs. In conclusion, we show that Salmonella system can efficiently deliver vaccine antigens to APCs and is, thus, capable to elicit heterologous antigen-specific adaptive immunity.
Collapse
Affiliation(s)
- Jehyoung Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Irshad Ahmed Hajam
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| |
Collapse
|
11
|
Gao X, Xu K, Yang G, Shi C, Huang H, Wang J, Yang W, Liu J, Liu Q, Kang Y, Jiang Y, Wang C. Construction of a novel DNA vaccine candidate targeting F gene of genotype VII Newcastle disease virus and chicken IL-18 delivered by Salmonella. J Appl Microbiol 2019; 126:1362-1372. [PMID: 30785663 DOI: 10.1111/jam.14228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/01/2019] [Accepted: 02/14/2019] [Indexed: 11/30/2022]
Abstract
AIMS Genotype VII Newcastle disease (ND) is one of the most epidemic and serious infectious diseases in the poultry industry. A novel vaccine targeting VII Newcastle disease virus (NDV) is still proving elusive. METHODS AND RESULTS In this study, we constructed regulated delayed lysis Salmonella strains expressing either a fusion protein (F) alone under an eukaryotic CMV promoter or together with chicken IL-18 (chIL-18) as a molecular adjuvant under prokaryotic Ptrc promoter, named pYL1 and pYL23 respectively. Oral immunization with recombinant strains induced NDV-specific serum IgG antibodies in both pYL1- and pYL23-immunized chickens. The presence of chIL-18 significantly increased lymphocyte proliferation in immunized chickens, as well as the percentages of CD3+ CD4+ and CD3+ CD8+ T cells in serum, even if a statistically significant difference did not exist. After a virulent challenge, pYL23 immunization provided about 80% protection at day 10 postinfection, compared with 60% of protection offered by pYL1 immunization and 100% protection in the inactivated vaccine group, indicating the enhanced immune response provided by chIL-18, which was also confirmed by histochemical analysis. CONCLUSIONS Recombinant lysis Salmonella-vectored DNA vaccine could provide us a novel potential option for controlling NDV infection. SIGNIFICANCE AND IMPACT OF THE STUDY This study took use of a regulated delayed lysis Salmonella vector for the design of an orally administrated vaccine against NDV.
Collapse
Affiliation(s)
- X Gao
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - K Xu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - G Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - C Shi
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - H Huang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - J Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - W Yang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - J Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Q Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Y Kang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Y Jiang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - C Wang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of animal production and product quality safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
12
|
Hajam IA, Kim J, Lee JH. Intranasally administered polyethylenimine adjuvanted influenza M2 ectodomain induces partial protection against H9N2 influenza A virus infection in chickens. Vet Immunol Immunopathol 2019; 209:78-83. [PMID: 30885310 DOI: 10.1016/j.vetimm.2019.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/11/2019] [Accepted: 02/19/2019] [Indexed: 12/25/2022]
Abstract
This study aimed to investigate whether intranasally coadministered four tandem copies of extracellular domains of M2 (M2e) and polyethyleneimine (PEI), a mucosal adjuvant, can protect chickens against H9N2 influenza A virus infection. Groups of chickens were intranasally vaccinated with M2e plus PEI adjuvant, M2e alone or PEI adjuvant, and antibody (serum IgG and mucosal IgA) and cellular (CD4+ T cells and IFN-γ levels) immune responses were measured post-vaccination. We demonstrated that the chickens vaccinated with M2e plus PEI adjuvant showed significantly (p < 0.05) higher M2e-specific systemic IgG and mucosal IgA responses compared to the chickens that received either M2e alone or PEI adjuvant. The IgA responses measured in lungs were almost comparable to that of the serum IgG levels. Upon restimulation of the vaccinated peripheral blood mononuclear cells (PBMCs) with M2e antigen, significantly (p < 0.05) higher IFN-γ levels were observed only in M2e plus PEI adjuvant vaccinated group. Lymphoproliferative and CD4+ T cell responses, as measured by MTT-based assay and flow cytometry, respectively, were also observed significantly (p < 0.05) higher in M2e plus PEI adjuvant vaccinated chickens. On challenge with the H9N2 virus (104TCID50) at 28th day post-vaccination, M2e plus PEI adjuvant vaccinated group exhibited lower lung inflammation and viral load compared to the chickens treated with either M2e alone or PEI adjuvant. In summary, we show that intranasally coadministered M2e and PEI adjuvant can elicit humoral and cell-mediated immune responses and can reduce viremia levels in chickens post H9N2 infection in chickens.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jehyoung Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea.
| |
Collapse
|
13
|
Attenuated Salmonella secreting Brucella protective antigens confer dual-faceted protection against brucellosis and salmonellosis in a mouse model. Vet Immunol Immunopathol 2019; 209:31-36. [PMID: 30885303 DOI: 10.1016/j.vetimm.2019.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/02/2019] [Accepted: 02/02/2019] [Indexed: 11/21/2022]
Abstract
We demonstrated the use of attenuated Salmonella strains secreting Brucella antigens SodC, Omp19, BLS, and PrpA as live vaccine candidates against Brucella abortus infection and presented their cross-protection against Salmonella infections using a BALB/c mice model. Here, a single immunization with each individual strain was capable of establishing significantly high (p < 0.05) Brucella-specific systemic immunoglobulin (Ig)G and secretory IgA (sIgA) responses compared to control mice. Upon stimulation of the splenocytes harvested from immunized mice with the respective antigens SodC, Omp19, BLS, and PrpA, significant increases in splenocyte proliferative responses against all four antigens versus PBS and vector controls were observed (p < 0.05). Additionally, interferon-γ and interleukin-4 secretion clearly demonstrated an uplift of these cytokines in all four strains upon immunization compared to the control groups. However, a significantly high response was noted in the mice groups immunized with Salmonella secreting SodC and Omp19 only. Upon virulent Brucella abortus 544 challenge, all four antigens presented a significantly high protection index (PI) in the spleen, as follows: 0.85 for SodC; 0.96 for Omp19; 0.6 for BLS; and 0.66 for PrpA. In contrast, in the liver, the same antigens resulted in PI values of 1.37, 1.14, 1.12, and 1.81, respectively. Immunological profiling of immunized mice against Salmonella-specific immune responses also showed significant elicitation of both humoral and cell-mediated immune responses as measured by IgG, sIgA, splenocyte proliferation, and cytokine induction. In addition, full protection against virulent Salmonella challenge was shown with no mortality in immunized mice, whereas 100% (8/8) mortality was observed in control mice over a two-week post-Salmonella challenge. In conclusion, we show that the live attenuated Salmonella delivering Brucella protective antigens may efficiently confer dual protection against both brucellosis and salmonellosis in immunized mice.
Collapse
|
14
|
Intranasally administered anti-Brucella subunit vaccine formulation induces protective immune responses against nasal Brucella challenge. Vet Microbiol 2018; 228:112-118. [PMID: 30593355 DOI: 10.1016/j.vetmic.2018.11.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/25/2022]
Abstract
The present study was aimed to develop a safe and effective anti-Brucella subunit vaccine for mucosal protection against the respiratory exposure of Brucella infection. A chitosan-based Brucella nasal vaccine (BNV) was formulated using well-known Brucella immunogens, sodC, omp19, BLS and PrpA and tested against nasal Brucella challenge in BALB/c mice. The mice were intra-nasally vaccinated with sterile phosphate buffer saline (PBS), BNV or BNV plus Brucella LPS, and humoral (systemic IgG and mucosal IgA) and cell-mediated immune responses were analyzed. Results showed that mice vaccinated with either BNV or BNV plus LPS elicited significantly (p < 0.05) high IgG and IgA responses compared to the PBS control. The IgG responses were significantly (p < 0.05) higher than IgA levels, which showed almost comparable levels observed in either intestines or in lungs. Furthermore, the IgG and IgA responses against each individual component of the BNV formulation indicated that omp19 induced highest levels of both IgG and IgA levels than the other constituents of BNV formulation. Upon re-stimulation of the splenocytes with Brucella whole cell lysate, significantly (p < 0.05) high IFN-γ levels, lymphocyte proliferation, and CD4+ T cell responses were observed in mice vaccinated with BNV or BNV plus LPS. Upon sub-lethal nasal challenge with wild-type Brucella strain, vaccinated mice showed significant reduction of Brucella recovery in lungs and spleen compared to the PBS control. This study indicates that BNV formulation with or without Brucella LPS efficiently induced humoral and cell-mediated immune responses and conferred significant protection against the sub-lethal Brucella challenge.
Collapse
|
15
|
Hajam IA, Kim J, Lee JH. Salmonella Gallinarum delivering M2eCD40L in protein and DNA formats acts as a bivalent vaccine against fowl typhoid and H9N2 infection in chickens. Vet Res 2018; 49:99. [PMID: 30285855 PMCID: PMC6389227 DOI: 10.1186/s13567-018-0593-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 09/17/2018] [Indexed: 02/07/2023] Open
Abstract
Fowl typhoid (FT), a septicemic disease caused by Salmonella Gallinarum (SG), and H9N2 influenza infection are two economically important diseases that affect poultry industry worldwide. Herein, we exploited a live attenuated SG mutant (JOL967) to deliver highly conserved extracellular domains of H9N2 M2 (M2e) to induce protective immunity against both H9N2 infection and FT. To increase the immunogenicity of M2e, we physically linked it with CD40L and cloned the fusion gene into either prokaryotic constitutive expression vector pJHL65 or mammalian expression vector pcDNA3.1+. Then pJHL65-M2eCD40L or pcDNA-M2eCD40L recombinant plasmid was electroporated into JOL967 strain and the resultant clones were designated as JOL2074 and JOL2076, respectively. We demonstrated that the chickens vaccinated once orally with a co-mix of JOL2074 and JOL2076 strains elicited significantly (p < 0.05) higher M2e-specific humoral and cell-mediated immunity compared to JOL2074 alone vaccinated group. However, SG-specific immune responses were comparable in both the vaccination groups. On challenge with the virulent H9N2 virus (105 TCID50) at 28th day post-vaccination, chickens that received a co-mix of JOL2074 plus JOL2076 strains exhibited significantly (p < 0.05) lower lung inflammation and viral load in both lungs and cloacal samples than JOL2074 alone vaccinated group. Against challenge with the lethal wild-type SG, both the vaccination groups exhibited only 12.5% mortality compared to 75% mortality observed in the control group. In conclusion, we show that SG delivering M2eCD40L can act as a bivalent vaccine against FT and H9N2 infection and further studies are warranted to develop this SG-M2eCD40L vaccine as a broadly protective vaccine against avian influenza virus subtypes.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596 Republic of Korea
| | - Jehyoung Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596 Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596 Republic of Korea
| |
Collapse
|
16
|
Hajam IA, Kim J, Lee JH. Oral immunization with a novel attenuated Salmonella Gallinarum encoding infectious bronchitis virus spike protein induces protective immune responses against fowl typhoid and infectious bronchitis in chickens. Vet Res 2018; 49:91. [PMID: 30208963 PMCID: PMC6134591 DOI: 10.1186/s13567-018-0588-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/03/2018] [Indexed: 12/24/2022] Open
Abstract
Fowl typhoid (FT), a septicemic disease caused by Salmonella Gallinarum (SG), and infectious bronchitis (IB) are two economically important avian diseases that affect poultry industry worldwide. Herein, we exploited a live attenuated SG mutant, JOL967, to deliver spike (S) protein 1 of IB virus (V) to elicit protective immunity against both FT and IB in chickens. The codon optimized S1 nucleotide sequence was cloned in-frame into a prokaryotic constitutive expression vector, pJHL65. Subsequently, empty pJHL65 or recombinant pJHL65-S1 plasmid was electroporated into JOL967 and the resultant clones were designated as JOL2068 and JOL2077, respectively. Our results demonstrated that the chickens vaccinated once orally with JOL2077 elicited significantly (p < 0.05) higher IBV-specific humoral and cell-mediated immunity compared to JOL2068 and PBS control groups. Consequently, on challenge with the virulent IBV strain at 28th day post-vaccination, JOL2077 vaccinated birds displayed significantly (p < 0.05) lower inflammatory lesions in virus-targeted tissues compared to control groups. Furthermore, 33.3% (2 of 6) of birds vaccinated with JOL2077 vaccine had shown virus recovery from tracheal tissues compared to 100% (6 of 6) recovery obtained in both the control groups. Against wild-type SG lethal challenge, both JOL2077 and JOL2068 vaccinated groups exhibited only 10% mortality compared to 80% mortality observed in PBS control group. In conclusion, we show that JOL2077 can induce efficient IBV- and carrier-specific protective immunity and can act as a bivalent vaccine against FT and IB. Further studies are warranted to investigate the potential of JOL2077 vaccine in broiler and young layer birds.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - Jehyoung Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea.
| |
Collapse
|
17
|
Schepens B, De Vlieger D, Saelens X. Vaccine options for influenza: thinking small. Curr Opin Immunol 2018; 53:22-29. [DOI: 10.1016/j.coi.2018.03.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [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]
|
18
|
Zhu C, Lin H, Tang L, Chen J, Wu Y, Zhong G. Oral Chlamydia vaccination induces transmucosal protection in the airway. Vaccine 2018; 36:2061-2068. [PMID: 29550196 DOI: 10.1016/j.vaccine.2018.03.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 11/19/2022]
Abstract
Although Chlamydia has been frequently detected in the gastrointestinal tracts of both humans and animals, it is not associated with any gastrointestinal pathology. We have recently shown that gastrointestinal Chlamydiamuridarum is not only non-pathogenic but also induces protective immunity in the genital tract. We now report that the transmucosal immunity induced by a single oral immunization with C.muridarum protected the mouse airway from a subsequent challenge infection. The oral immunization significantly reduced chlamydial burden in the airway as early as day 3 after intranasal challenge. As a result, the airway chlamydial spreading to extra-airway tissues was completely prevented on day 3 and significantly reduced on day 9. The immunized mice were protected from any significant systemic toxicity caused by the intranasal challenge since there was no significant bodyweight drop in the immunized mice. This robust protection correlated well with Chlamydia-specific antibodies that recognize chlamydial organism surface antigens and T cell responses that are dominated with a Th1 phenotype. The immunized mice developed high ratios of IgG2b/c over IgG1 levels and IFNγ-producing over IL-5- or IL-13-producing lymphocytes. Thus, we have demonstrated that oral vaccination with C. muridarum can induce Th1-dominant transmucosal immunity in the airway. Together with previous studies, we propose that non-pathogenic colonization of Chlamydia in the gastrointestinal tract be explored as an oral delivery system for inducing protection against infections and pathologies in extra-gastrointestinal tissues.
Collapse
Affiliation(s)
- Cuiming Zhu
- Department of Medical Microbiology, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan 421001, China; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Hui Lin
- The 2nd Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Lingli Tang
- The 2nd Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jianlin Chen
- The 2nd Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yimou Wu
- Department of Medical Microbiology, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan 421001, China.
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, TX 78229, United States.
| |
Collapse
|