1
|
Song H, Liu X, Gao X, Li J, Shang Y, Gao W, Li Y, Zhang Z. Transcriptome analysis of pre-immune state induced by interferon gamma inhibiting the replication of H 9N 2 avian influenza viruses in chicken embryo fibroblasts. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 103:105332. [PMID: 35811034 DOI: 10.1016/j.meegid.2022.105332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 06/15/2023]
Abstract
Interferon (IFN), a critical antiviral cytokine produced by pathogens-induced cells, plays an important role in host innate immune system. In this study, to investigate the inhibition effect of IFN on avian influenza virus (AIV), Chicken Embryo Fibroblasts (CEFs) was infected by H9N2 AIV. The pre-immune state and transcriptome analysis have been observed and performed. The result showed chicken interferon gamma (chIFN-γ) have the most inhibitory effect on H9N2 virus among three types of chicken interferons (chIFNs). Inhibition of chIFN-γ on H9N2 virus was verified by indirect immunofluorescence, RT-qPCR and western blot. The possible signaling pathways induced by chIFN-γ with or without virus were analyzed by transcriptome. The transcriptome data were compared among H9N2-infected, chIFN-γ-treated, chIFN-γ + H9N2-treated, and Control groups. In summary, RNA-sequencing (RNA-seq) data suggested that H9N2 virus infection resulted in corresponding response of certain defensive, inflammatory and metabolism pathways to the virus replication in CEFs. Furthermore, while CEFs were treated with chIFN-γ, many immune-related signaling pathways in cells are affected and altered. Antiviral genes involved in these immune pathways such as interferon regulatory factors, chemokines, interferon-stimulated genes (ISGs) and transcription factors were significantly up-regulated, and showed significant antiviral responses. Compared with virus infected CEFs alone, pretreatment with IFN induced the expression of antiviral genes and activated related antiviral pathways, inhibited the viral replication as result. Our study provided functional annotations for antiviral genes and the basis for studying the mechanism of chIFN-γ mediated response against H9N2 AIV.
Collapse
Affiliation(s)
- Haozhi Song
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xingjian Liu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xintao Gao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jialei Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yuting Shang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Weisong Gao
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yinü Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Zhifang Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
2
|
Sadria M, Layton AT. Modeling within-Host SARS-CoV-2 Infection Dynamics and Potential Treatments. Viruses 2021; 13:1141. [PMID: 34198530 PMCID: PMC8231802 DOI: 10.3390/v13061141] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/27/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
The goal of this study was to develop a mathematical model to simulate the actions of drugs that target SARS-CoV-2 virus infection. To accomplish that goal, we have developed a mathematical model that describes the control of a SARS-CoV-2 infection by the innate and adaptive immune components. Invasion of the virus triggers the innate immunity, whereby interferon renders some of the target cells resistant to infection, and infected cells are removed by effector cells. The adaptive immune response is represented by plasma cells and virus-specific antibodies. The model is parameterized and then validated against viral load measurements collected in COVID-19 patients. We apply the model to simulate three potential anti-SARS-CoV-2 therapies: (1) Remdesivir, a repurposed drug that has been shown to inhibit the transcription of SARS-CoV-2, (2) an alternative (hypothetical) therapy that inhibits the virus' entry into host cells, and (3) convalescent plasma transfusion therapy. Simulation results point to the importance of early intervention, i.e., for any of the three therapies to be effective, it must be administered sufficiently early, not more than a day or two after the onset of symptoms. The model can serve as a key component in integrative platforms for rapid in silico testing of potential COVID-19 therapies and vaccines.
Collapse
Affiliation(s)
- Mehrshad Sadria
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Anita T. Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
- Departments of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Cheriton School of Computer Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- School of Pharmacy, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
3
|
Kwak C, Nguyen QT, Kim J, Kim TH, Poo H. Influenza Chimeric Protein (3M2e-3HA2-NP) Adjuvanted with PGA/Alum Confers Cross-Protection against Heterologous Influenza A Viruses. J Microbiol Biotechnol 2021; 31:304-316. [PMID: 33263336 PMCID: PMC9705887 DOI: 10.4014/jmb.2011.11029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
Vaccination is the most effective way to prevent influenza virus infections. However, conventional vaccines based on hemagglutinin (HA) have to be annually updated because the HA of influenza viruses constantly mutates. In this study, we produced a 3M2e-3HA2-NP chimeric protein as a vaccine antigen candidate using an Escherichia coli expression system. The vaccination of chimeric protein (15 μg) conferred complete protection against A/Puerto Rico/8/1934 (H1N1; PR8) in mice. It strongly induced influenza virus-specific antibody responses, cytotoxic T lymphocyte activity, and antibody-dependent cellular cytotoxicity. To spare the dose and enhance the cross-reactivity of the chimeric, we used a complex of poly-γ-glutamic acid and alum (PGA/alum) as an adjuvant. PGA/alum-adjuvanted, low-dose chimeric protein (1 or 5 μg) exhibited higher cross-protective effects against influenza A viruses (PR8, CA04, and H3N2) compared with those of chimeric alone or alum-adjuvanted proteins in vaccinated mice. Moreover, the depletion of CD4+ T, CD8+ T, and NK cells reduced the survival rate and efficacy of the PGA/alum-adjuvanted chimeric protein. Collectively, the vaccination of PGA/alum-adjuvanted chimeric protein induced strong protection efficacy against homologous and heterologous influenza viruses in mice, which suggests that it may be a promising universal influenza vaccine candidate.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Alum Compounds/administration & dosage
- Animals
- Antibodies, Viral/immunology
- Cross Reactions
- Female
- Hemagglutinins, Viral
- Humans
- Immunity, Humoral
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nucleocapsid Proteins/administration & dosage
- Nucleocapsid Proteins/genetics
- Nucleocapsid Proteins/immunology
- Polyglutamic Acid/administration & dosage
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Viral Matrix Proteins/administration & dosage
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
Collapse
Affiliation(s)
- Chaewon Kwak
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Quyen Thi Nguyen
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jaemoo Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Tae-Hwan Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
| | - Haryoung Poo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
4
|
Zhao S, Yuan L, Li Y, Liu L, Luo Z, Lv Q, Rong R, Yang Y. Secretory IgA in Mucosa of Pharynx and Larynx Plays an Important Role against Influenza A Virus Infection in Kidney Yang Deficiency Syndrome Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:9316763. [PMID: 32308721 PMCID: PMC7146091 DOI: 10.1155/2020/9316763] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/10/2019] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Influenza virus poses a major threat to human health and has serious morbidity and mortality which commonly occurs in high-risk populations. Pharynx and larynx of the upper respiratory tract mucosa is the first defense line against influenza virus infection. However, the ability of the pharynx and larynx organ to eliminate the influenza pathogen is still not clear under different host conditions. METHODS In this study, a mouse model of kidney yang deficiency syndrome (KYDS) was used to mimic high-risk peoples. Two different methods of influenza A (H1N1) virus infection by nasal dropping or tracheal intubation were applied to these mice, which were divided into four groups: normal intubation (NI) group, normal nasal dropping (ND) group, model intubation (MI) group, and model nasal dropping (MD) group. The normal control (NC) group was used as a negative control. Body weight, rectal temperature, and survival rate were observed every day. Histopathologic changes, visceral index, gene expressions of H1N1, cytokine expressions, secretory IgA (SIgA) antibodies of tracheal lavage fluids in the upper respiratory tract, and bronchoalveolar lavage fluids were analyzed by ELISA. RESULTS The MD group had an earlier serious morbidity and mortality than the others. MI and NI groups became severe only in the 6th to 7th day after infection. The index of the lung increased significantly in NI, MI, and MD groups. Conversely, indices of the thymus and spleen increased significantly in NC and ND groups. H&E staining showed severe tissue lesions in MD, MI, and NI groups. H1N1 gene expressions were higher in the MD group compared with the MI group on the 3rd day; however, the MD group decreased significantly on the 7th day. IL-6 levels increased remarkably, and SIgA expressions decreased significantly in the MD group compared with the NC group. CONCLUSIONS SIgA secretions are influenced directly by different conditions of the host in the pharynx and larynx in the upper respiratory tract mucosa. In the KYDS virus disease mode, SIgA expressions could be inhibited severely, which leads to serious morbidity and mortality after influenza A virus infection. The SIgA expressions of the pharynx and larynx would be an important target in high-risk populations against the influenza A virus for vaccine or antiviral drugs research.
Collapse
Affiliation(s)
- Shaozhe Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Lei Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Yi Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Longchan Liu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Zixin Luo
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Qingtao Lv
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Rong Rong
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
- Shandong Provincial Collaborative Innovation Center for Antiviral Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Yong Yang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
- Shandong Provincial Collaborative Innovation Center for Antiviral Traditional Chinese Medicine, Jinan, Shandong 250355, China
| |
Collapse
|
5
|
Cho HK, Kang YM, Kim HM, Lee CH, Kim DY, Choi SH, Lee MH, Kang HM. Sales and immunogenicity of commercial vaccines to H9N2 low pathogenic avian influenza virus in Korea from 2007 to 2017. Vaccine 2020; 38:3191-3195. [PMID: 32169392 DOI: 10.1016/j.vaccine.2020.02.083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/15/2020] [Accepted: 02/29/2020] [Indexed: 11/18/2022]
Abstract
The present study was conducted to monitor sales activity and immunogenicity of commercial H9N2 vaccines produced in Korea from 2007 to 2017. Recorded sales of H9N2 vaccine were around 671 million doses, with 10 million doses sold in 2007, rising to a peak of 93 million doses in 2016, with a slight fall in 2017. Multivalent combined vaccines made up around 90% of all vaccine sales, and around 30% of all vaccines were distributed by regional governments for free. The regional vaccination rate was the highest in Gyeonggi and Chungnam, respectively with proportional to the population of layer and breeder chickens. There have been no cases of field infection since 2009. The mean antibody titer was 5.82 log2 across the study period. Our results suggest that continuous genetic monitoring of H9N2 viruses circulating in the field and updating the vaccine seed strain periodically are necessary in order to control H9N2 outbreaks.
Collapse
Affiliation(s)
- Hyun-Kyu Cho
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea; College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Yong-Myung Kang
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Hyun-Mi Kim
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Chi-Ho Lee
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Do-Young Kim
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Sang-Hyun Choi
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Myoung-Heon Lee
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea
| | - Hyun-Mi Kang
- Avian Influenza Research & Diagnostic Division, Animal and Plant Quarantine Agency, 177 Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do 39660, Republic of Korea.
| |
Collapse
|
6
|
Warren R, Domm W, Yee M, Campbell A, Malone J, Wright T, Mayer-Pröschel M, O'Reilly MA. Ataxia-telangiectasia mutated is required for the development of protective immune memory after influenza A virus infection. Am J Physiol Lung Cell Mol Physiol 2019; 317:L591-L601. [PMID: 31509427 PMCID: PMC6879906 DOI: 10.1152/ajplung.00031.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 08/21/2019] [Accepted: 09/01/2019] [Indexed: 01/26/2023] Open
Abstract
Ataxia-telangiectasia (A-T), caused by mutations in the A-T mutated (ATM) gene, is a neurodegenerative disorder affecting ∼1 in 40,000-100,000 children. Recurrent respiratory infections are a common and challenging comorbidity, often leading to the development of bronchiectasis in individuals with A-T. The role of ATM in development of immune memory in response to recurrent respiratory viral infections is not well understood. Here, we infect wild-type (WT) and Atm-null mice with influenza A virus (IAV; HKx31, H3N2) and interrogate the immune memory with secondary infections designed to challenge the B cell memory response with homologous infection (HKx31) and the T cell memory response with heterologous infection (PR8, H1N1). Although Atm-null mice survived primary and secondary infections, they lost more weight than WT mice during secondary infections. This enhanced morbidity to secondary infections was not attributed to failure to effectively clear virus during the primary IAV infection. Instead, Atm-null mice developed persistent peribronchial inflammation, characterized in part by clusters of B220+ B cells. Additionally, levels of select serum antibodies to hemagglutinin-specific IAV were significantly lower in Atm-null than WT mice. These findings reveal that Atm is required to mount a proper memory response to a primary IAV infection, implying that vaccination of children with A-T by itself may not be sufficiently protective against respiratory viral infections.
Collapse
Affiliation(s)
- Rachel Warren
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - William Domm
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Min Yee
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Andrew Campbell
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Jane Malone
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Terry Wright
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Margot Mayer-Pröschel
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| |
Collapse
|
7
|
Gianchecchi E, Manenti A, Kistner O, Trombetta C, Manini I, Montomoli E. How to assess the effectiveness of nasal influenza vaccines? Role and measurement of sIgA in mucosal secretions. Influenza Other Respir Viruses 2019; 13:429-437. [PMID: 31225704 PMCID: PMC6692539 DOI: 10.1111/irv.12664] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 01/07/2023] Open
Abstract
Secretory IgAs (sIgA) constitute the principal isotype of antibodies present in nasal and mucosal secretions. They are secreted by plasma cells adjacent to the mucosal epithelial cells, the site where infection occurs, and are the main humoral mediator of mucosal immunity. Mucosally delivered vaccines, such as live attenuated influenza vaccine (LAIV), are able to mimic natural infection without causing disease or virus transmission and mainly elicit a local immune response. The measurement of sIgA concentrations in nasal swab/wash and saliva samples is therefore a valuable tool for evaluating their role in the effectiveness of such vaccines. Here, we describe two standardized assays (enzyme‐linked immunosorbent assay and microneutralization) available for the quantification of sIgA and discuss the advantages and limitations of their use.
Collapse
Affiliation(s)
| | | | | | - Claudia Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- VisMederi Srl, Siena, Italy.,VisMederi Research Srl, Siena, Italy.,Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
8
|
Kaoud H, Hussein H, El-Dahshan A, Kaliefa H, Rohaim M. Co-circulation of avian influenza viruses in commercial farms, backyards and live market birds in Egypt. Int J Vet Sci Med 2019. [DOI: 10.1016/j.ijvsm.2014.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- H.A. Kaoud
- Department of Veterinary Hygiene and Environmental Pollution, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - H.A. Hussein
- Department of Virology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - A.R. El-Dahshan
- Department of Veterinary Hygiene and Environmental Pollution, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - H.S. Kaliefa
- Department of Veterinary Hygiene and Environmental Pollution, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - M.A. Rohaim
- Department of Virology, Faculty of Veterinary Medicine, Cairo University, Egypt
| |
Collapse
|
9
|
Carter DM, Darby CA, Johnson SK, Carlock MA, Kirchenbaum GA, Allen JD, Vogel TU, Delagrave S, DiNapoli J, Kleanthous H, Ross TM. Elicitation of Protective Antibodies against a Broad Panel of H1N1 Viruses in Ferrets Preimmune to Historical H1N1 Influenza Viruses. J Virol 2017; 91:e01283-17. [PMID: 28978709 PMCID: PMC5709581 DOI: 10.1128/jvi.01283-17] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/13/2017] [Indexed: 11/20/2022] Open
Abstract
Most preclinical animal studies test influenza vaccines in immunologically naive animal models, even though the results of vaccination may not accurately reflect the effectiveness of vaccine candidates in humans that have preexisting immunity to influenza. In this study, novel, broadly reactive influenza vaccine candidates were assessed in preimmune ferrets. These animals were infected with different H1N1 isolates before being vaccinated or infected with another influenza virus. Previously, our group has described the design and characterization of computationally optimized broadly reactive hemagglutinin (HA) antigens (COBRA) for H1N1 isolates. Vaccinating ferrets with virus-like particle (VLP) vaccines expressing COBRA HA proteins elicited antibodies with hemagglutination inhibition (HAI) activity against more H1N1 viruses in the panel than VLP vaccines expressing wild-type HA proteins. Specifically, ferrets infected with the 1986 virus and vaccinated with a single dose of the COBRA HA VLP vaccines elicited antibodies with HAI activity against 11 to 14 of the 15 H1N1 viruses isolated between 1934 and 2013. A subset of ferrets was infected with influenza viruses expressing the COBRA HA antigens. These COBRA preimmune ferrets had superior breadth of HAI activity after vaccination with COBRA HA VLP vaccines than COBRA preimmune ferrets vaccinated with VLP vaccines expressing wild-type HA proteins. Overall, priming naive ferrets with COBRA HA based viruses or using COBRA HA based vaccines to boost preexisting antibodies induced by wild-type H1N1 viruses, COBRA HA antigens elicited sera with the broadest HAI reactivity against multiple antigenic H1N1 viral variants. This is the first report demonstrating the effectiveness of a broadly reactive or universal influenza vaccine in a preimmune ferret model.IMPORTANCE Currently, many groups are testing influenza vaccine candidates to meet the challenge of developing a vaccine that elicits broadly reactive and long-lasting protective immune responses. The goal of these vaccines is to stimulate immune responses that react against most, if not all, circulating influenza strains, over a long period of time in all populations of people. Commonly, these experimental vaccines are tested in naive animal models that do not have anti-influenza immune responses; however, humans have preexisting immunity to influenza viral antigens, particularly antibodies to the HA and NA glycoproteins. Therefore, this study investigated how preexisting antibodies to historical influenza viruses influenced HAI-specific antibodies and protective efficacy using a broadly protective vaccine candidate.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/immunology
- Antigens, Viral/immunology
- Ferrets
- Hemagglutination Inhibition Tests
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H1N1 Subtype/classification
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Donald M Carter
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Christopher A Darby
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Scott K Johnson
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Michael A Carlock
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Greg A Kirchenbaum
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - James D Allen
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
| | - Thorsten U Vogel
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Simon Delagrave
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Joshua DiNapoli
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Harold Kleanthous
- Sanofi-Pasteur, Inc., Discovery North America, Cambridge, Massachusetts, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
10
|
Shim SM, Song EJ, Song D, Lee TY, Kim DJ, Nam JH, Gwin Jeong D, Lee CK, Kim SH, Kim JK. Nontoxic outer membrane vesicles efficiently increase the efficacy of an influenza vaccine in mice and ferrets. Vaccine 2017; 35:3741-3748. [PMID: 28576571 DOI: 10.1016/j.vaccine.2017.05.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 02/06/2023]
Abstract
In this study, we developed a further-modified outer membrane vesicle (fmOMV) from the ΔmsbB/ΔpagP mutant of Escherichia coli transformed with the plasmid, pLpxF, in order to use it as an adjuvant for pandemic H1N1 (pH1N1) influenza vaccine. We evaluated the efficacy of the pH1N1 influenza vaccine containing the fmOMV in animal models as compared to the commercial adjuvants, alum or AddaVaxTM. The fmOMV-adjuvanted pH1N1 influenza vaccine induced a significant increase in the humoral immunity; however, this effect was less than that of the AddaVaxTM. The fmOMV-adjuvanted vaccine displayed pronounced an enhanced protective efficacy with increased T cell immune response and reduced the viral load in the lungs of the infected mice after challenging them with a lethal dose of the homologous virus. Moreover, it resulted in a significantly higher cross-protection against heterologous virus challenge than that of the pH1N1 vaccine with alum or with no adjuvants. In ferrets, the fmOMV-adjuvanted vaccine elicited a superior antibody response based on the HI titer and efficiently protected the animals from the lethal viral challenges. Taken together, the nontoxic fmOMV could be a promising adjuvant for inducing robust T cell priming into the pH1N1 vaccine and might be broadly applicable to the development of preventive measures against influenza virus infection.
Collapse
Affiliation(s)
- Sang-Mu Shim
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Eun-Jung Song
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Daesub Song
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Tae-Young Lee
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Doo-Jin Kim
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong-Hyun Nam
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Dae Gwin Jeong
- Viral Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Chong-Kil Lee
- Department of Pharmacy, College of Pharmacy, Chungbuk National University, 52 Naesudong-ro, Heungdeok-gu, Cheongju, Chungcheongbuk-do 28644, Republic of Korea
| | - Sang-Hyun Kim
- College of Veterinary Medicine, Gyeongsang National University, 501 Jinjudae-ro, Jinju, Gyeongsangnam-do 52828, Republic of Korea.
| | - Jeong-Ki Kim
- Department of Pharmacy, College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea.
| |
Collapse
|
11
|
Poly-γ-glutamic acid/chitosan nanogel greatly enhances the efficacy and heterosubtypic cross-reactivity of H1N1 pandemic influenza vaccine. Sci Rep 2017; 7:44839. [PMID: 28322289 PMCID: PMC5359587 DOI: 10.1038/srep44839] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 02/14/2017] [Indexed: 11/09/2022] Open
Abstract
In 2009, the global outbreak of an influenza pandemic emphasized the need for an effective vaccine adjuvant. In this study, we examined the efficacy of poly-γ-glutamic acid/chitosan (PC) nanogel as an adjuvant for the influenza vaccine. PC nanogel significantly enhanced antigen-specific cross-presentation and cytotoxic T lymphocyte (CTL) activity. Compared with alum, the protective efficacy of the pandemic H1N1 influenza (pH1N1) vaccine was substantially increased by PC nanogel, with increased hemagglutination-inhibition titers, CTL activity, and earlier virus clearance after homologous and heterosubtypic [A/Philippines/2/82 (H3N2)] virus challenges. However, CD8+ T cell-depleted mice displayed no protection against the heterosubtypic virus challenge after immunization with PC nanogel-adjuvanted pH1N1 vaccine. We also observed that using PC nanogel as a vaccine adjuvant had a dose-sparing effect and significantly enhanced the long-lasting protection of the pH1N1 vaccine. Together, these results suggest that PC nanogel is a promising vaccine adjuvant that could broadly prevent influenza virus infection.
Collapse
|
12
|
Sterilizing immunity to influenza virus infection requires local antigen-specific T cell response in the lungs. Sci Rep 2016; 6:32973. [PMID: 27596047 PMCID: PMC5011745 DOI: 10.1038/srep32973] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 08/17/2016] [Indexed: 12/29/2022] Open
Abstract
Sterilizing immunity is a unique immune status, which prevents effective virus infection into the host. It is different from the immunity that allows infection but with subsequent successful eradication of the virus. Pre-infection induces sterilizing immunity to homologous influenza virus challenge in ferret. In our antigen-specific experimental system, mice pre-infected with PR8 influenza virus through nasal route are likewise resistant to reinfection of the same strain of virus. The virus is cleared before establishment of effective infection. Intramuscular influenza virus injection confers protection against re-infection with facilitated virus clearance but not sterilizing immunity. Pre-infection and intramuscular injection generates comparable innate immunity and antibody response, but only pre-infection induces virus receptor reduction and efficient antigen-specific T cell response in the lungs. Pre-infection with nH1N1 influenza virus induces virus receptor reduction but not PR8-specific T cell immune response in the lungs and cannot prevent infection of PR8 influenza virus. Pre-infection with PR8 virus induced PR8-specific T cell response in the lungs but cannot prevent infection of nH1N1 virus either. These results reveal that antigen-specific T cell immunity is required for sterilizing immunity.
Collapse
|
13
|
Lee YN, Kim MC, Lee YT, Kim YJ, Kang SM. Mechanisms of Cross-protection by Influenza Virus M2-based Vaccines. Immune Netw 2015; 15:213-21. [PMID: 26557805 PMCID: PMC4637342 DOI: 10.4110/in.2015.15.5.213] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 08/25/2015] [Accepted: 09/10/2015] [Indexed: 01/06/2023] Open
Abstract
Current influenza virus vaccines are based on strain-specific surface glycoprotein hemagglutinin (HA) antigens and effective only when the predicted vaccine strains and circulating viruses are well-matched. The current strategy of influenza vaccination does not prevent the pandemic outbreaks and protection efficacy is reduced or ineffective if mutant strains emerge. It is of high priority to develop effective vaccines and vaccination strategies conferring a broad range of cross protection. The extracellular domain of M2 (M2e) is highly conserved among human influenza A viruses and has been utilized to develop new vaccines inducing cross protection against different subtypes of influenza A virus. However, immune mechanisms of cross protection by M2e-based vaccines still remain to be fully elucidated. Here, we review immune correlates and mechanisms conferring cross protection by M2e-based vaccines. Molecular and cellular immune components that are known to be involved in M2 immune-mediated protection include antibodies, B cells, T cells, alveolar macrophages, Fc receptors, complements, and natural killer cells. Better understanding of protective mechanisms by immune responses induced by M2e vaccination will help facilitate development of broadly cross protective vaccines against influenza A virus.
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, Anyang 14089, 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
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
14
|
Price I, Mochan-Keef ED, Swigon D, Ermentrout GB, Lukens S, Toapanta FR, Ross TM, Clermont G. The inflammatory response to influenza A virus (H1N1): An experimental and mathematical study. J Theor Biol 2015; 374:83-93. [PMID: 25843213 DOI: 10.1016/j.jtbi.2015.03.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 03/12/2015] [Accepted: 03/13/2015] [Indexed: 10/23/2022]
Abstract
Mortality from influenza infections continues as a global public health issue, with the host inflammatory response contributing to fatalities related to the primary infection. Based on Ordinary Differential Equation (ODE) formalism, a computational model was developed for the in-host response to influenza A virus, merging inflammatory, innate, adaptive and humoral responses to virus and linking severity of infection, the inflammatory response, and mortality. The model was calibrated using dense cytokine and cell data from adult BALB/c mice infected with the H1N1 influenza strain A/PR/8/34 in sublethal and lethal doses. Uncertainty in model parameters and disease mechanisms was quantified using Bayesian inference and ensemble model methodology that generates probabilistic predictions of survival, defined as viral clearance and recovery of the respiratory epithelium. The ensemble recovers the expected relationship between magnitude of viral exposure and the duration of survival, and suggests mechanisms primarily responsible for survival, which could guide the development of immuno-modulatory interventions as adjuncts to current anti-viral treatments. The model is employed to extrapolate from available data survival curves for the population and their dependence on initial viral aliquot. In addition, the model allows us to illustrate the positive effect of controlled inflammation on influenza survival.
Collapse
Affiliation(s)
- Ian Price
- Department of Mathematics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ericka D Mochan-Keef
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David Swigon
- Department of Mathematics, University of Pittsburgh, Pittsburgh, PA, USA; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - G Bard Ermentrout
- Department of Mathematics, University of Pittsburgh, Pittsburgh, PA, USA; Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah Lukens
- Department of Mathematics, University of Pittsburgh, Pittsburgh, PA, USA; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Ted M Ross
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gilles Clermont
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Hahn M, Schnitzler P, Schweiger B, Kunz C, Ho AD, Goldschmidt H, Schmitt M. Efficacy of single versus boost vaccination against influenza virus in patients with multiple myeloma. Haematologica 2015; 100:e285-8. [PMID: 25820335 DOI: 10.3324/haematol.2014.116772] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Michael Hahn
- Internal Medicine V, University Hospital Heidelberg, Germany
| | | | - Brunhilde Schweiger
- Robert Koch-Institut, National Reference Center for Influenza, Berlin, Germany
| | - Christina Kunz
- German Cancer Research Center (DKFZ), Dept. of Biostatistics, Heidelberg, Germany
| | - Anthony D Ho
- Internal Medicine V, University Hospital Heidelberg, Germany
| | | | - Michael Schmitt
- Internal Medicine V, University Hospital Heidelberg, Germany
| |
Collapse
|
16
|
Yamanaka T, Cullinane A, Gildea S, Bannai H, Nemoto M, Tsujimura K, Kondo T, Matsumura T. The potential impact of a single amino-acid substitution on the efficacy of equine influenza vaccines. Equine Vet J 2014; 47:456-62. [DOI: 10.1111/evj.12290] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 04/09/2014] [Indexed: 11/29/2022]
Affiliation(s)
- T. Yamanaka
- Epizootic Research Center; Equine Research Institute; Japan Racing Association; Shimotsuke Japan
| | - A. Cullinane
- Virology Unit; Irish Equine Centre; Johnstown Naas Co.; Kildare Ireland
| | - S. Gildea
- Virology Unit; Irish Equine Centre; Johnstown Naas Co.; Kildare Ireland
| | - H. Bannai
- Epizootic Research Center; Equine Research Institute; Japan Racing Association; Shimotsuke Japan
| | - M. Nemoto
- Epizootic Research Center; Equine Research Institute; Japan Racing Association; Shimotsuke Japan
| | - K. Tsujimura
- Epizootic Research Center; Equine Research Institute; Japan Racing Association; Shimotsuke Japan
| | - T. Kondo
- Epizootic Research Center; Equine Research Institute; Japan Racing Association; Shimotsuke Japan
| | - T. Matsumura
- Epizootic Research Center; Equine Research Institute; Japan Racing Association; Shimotsuke Japan
| |
Collapse
|
17
|
Muramatsu M, Yoshida R, Yokoyama A, Miyamoto H, Kajihara M, Maruyama J, Nao N, Manzoor R, Takada A. Comparison of antiviral activity between IgA and IgG specific to influenza virus hemagglutinin: increased potential of IgA for heterosubtypic immunity. PLoS One 2014; 9:e85582. [PMID: 24465606 PMCID: PMC3895000 DOI: 10.1371/journal.pone.0085582] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/05/2013] [Indexed: 12/25/2022] Open
Abstract
Both IgA and IgG antibodies are known to play important roles in protection against influenza virus infection. While IgG is the major isotype induced systemically, IgA is predominant in mucosal tissues, including the upper respiratory tract. Although IgA antibodies are believed to have unique advantages in mucosal immunity, information on direct comparisons of the in vitro antiviral activities of IgA and IgG antibodies recognizing the same epitope is limited. In this study, we demonstrate differences in antiviral activities between these isotypes using monoclonal IgA and IgG antibodies obtained from hybridomas of the same origin. Polymeric IgA-producing hybridoma cells were successfully subcloned from those originally producing monoclonal antibody S139/1, a hemaggulutinin (HA)-specific IgG that was generated against an influenza A virus strain of the H3 subtype but had cross-neutralizing activities against the H1, H2, H13, and H16 subtypes. These monoclonal S139/1 IgA and IgG antibodies were assumed to recognize the same epitope and thus used to compare their antiviral activities. We found that both S139/1 IgA and IgG antibodies strongly bound to the homologous H3 virus in an enzyme-linked immunosorbent assay, and there were no significant differences in their hemagglutination-inhibiting and neutralizing activities against the H3 virus. In contrast, S139/1 IgA showed remarkably higher cross-binding to and antiviral activities against H1, H2, and H13 viruses than S139/1 IgG. It was also noted that S139/1 IgA, but not IgG, drastically suppressed the extracellular release of the viruses from infected cells. Electron microscopy revealed that S139/1 IgA deposited newly produced viral particles on the cell surface, most likely by tethering the particles. These results suggest that anti-HA IgA has greater potential to prevent influenza A virus infection than IgG antibodies, likely due to increased avidity conferred by its multivalency, and that this advantage may be particularly important for heterosubtypic immunity.
Collapse
Affiliation(s)
- Mieko Muramatsu
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Reiko Yoshida
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Ayaka Yokoyama
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hiroko Miyamoto
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Masahiro Kajihara
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Junki Maruyama
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Naganori Nao
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Rashid Manzoor
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Ayato Takada
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- School of Veterinary Medicine, the University of Zambia, Lusaka, Zambia
- * E-mail:
| |
Collapse
|
18
|
Epstein SL. Control of influenza virus infection by immunity to conserved viral features. Expert Rev Anti Infect Ther 2014; 1:627-38. [PMID: 15482160 DOI: 10.1586/14787210.1.4.627] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Influenza has circulated among humans for centuries and kills more people than many newly emerging diseases. The present methods for control of influenza are not adequate, especially for dealing with a pandemic. In the face of a rapidly spreading outbreak, a race to isolate the virus and prepare a vaccine would probably not succeed in time to avoid great losses. Thus, additional anti-infection strategies are needed. Broad cross-protection against widely divergent influenza A subtypes is readily achieved in animals by several means of immunization. How does cross-protection work in animals, and can we apply what we have learned about it to induce broad cross-protection in humans?
Collapse
Affiliation(s)
- Suzanne L Epstein
- Laboratory of Immunology and Developmental Biology, Division of Cellular and Gene Therapies, HFM-730, Office of Cellular, Tissue and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, USA.
| |
Collapse
|
19
|
Almansa R, Bermejo-Martín JF, de Lejarazu Leonardo RO. Immunopathogenesis of 2009 pandemic influenza. Enferm Infecc Microbiol Clin 2013; 30 Suppl 4:18-24. [PMID: 23116788 PMCID: PMC7130369 DOI: 10.1016/s0213-005x(12)70100-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Three years after the pandemic, major advances have been made in our understanding of the innate and adaptive immune responses to the influenza A(H1N1)pdm09 virus and those responses' contribution to the immunopathology associated with this infection. Severe disease is characterized by early secretion of proinflammatory and immunomodulatory cytokines. This cytokine secretion persisted in patients with severe viral pneumonia and was directly associated with the degree of viral replication in the respiratory tract. Cytokines play important roles in the antiviral defense, but persistent hypercytokinemia may cause inflammatory tissue damage and participate in the genesis of the respiratory failure observed in these patients. An absence of pre-existing protective antibodies was the rule for both mild and severe cases. A role for pathogenic immunocomplexes has been proposed for this disease. Defective T cell responses characterize severe cases of infection caused by the influenza A(H1N1)pdm09 virus. Immune alterations associated with accompanying conditions such as obesity, pregnancy or chronic obstructive pulmonary disease may interfere with the normal development of the specific response to the virus. The role of host immunogenetic factors associated with disease severity is also discussed in this review. In conclusion, currently available information suggests a complex immunological dysfunction/alteration that characterizes the severe cases of 2009 pandemic influenza. The potential benefits of prophylactic/therapeutic interventions aimed at preventing/correcting such dysfunction warrant investigation.
Collapse
Affiliation(s)
- Raquel Almansa
- Unidad de Investigación Médica en Infección e Inmunidad (IMI), Investigación Biomédica del Clínico (ibC), Hospital Clínico Universitario, Valladolid, Spain
| | | | | |
Collapse
|
20
|
Comparative serological assays for the study of h5 and h7 avian influenza viruses. INFLUENZA RESEARCH AND TREATMENT 2013; 2013:286158. [PMID: 24163763 PMCID: PMC3791816 DOI: 10.1155/2013/286158] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/16/2013] [Indexed: 01/20/2023]
Abstract
The nature of influenza virus to randomly mutate and evolve into new types is an important challenge in the control of influenza infection. It is necessary to monitor virus evolution for a better understanding of the pandemic risk posed by certain variants as evidenced by the highly pathogenic avian influenza (HPAI) viruses. This has been clearly recognized in Egypt following the notification of the first HPAI H5N1 outbreak. The continuous circulation of the virus and the mass vaccination programme undertaken in poultry have resulted in a progressive genetic evolution and a significant antigenic drift near the major antigenic sites. In order to establish if vaccination is sufficient to provide significant intra- and interclade cross-protection, lentiviral pseudotypes derived from H5N1 HPAI viruses (A/Vietnam/1194/04, A/chicken/Egypt-1709-01/2007) and an antigenic drift variant (A/chicken/Egypt-1709-06-2008) were constructed and used in pseudotype-based neutralization assays (pp-NT). pp-NT data obtained was confirmed and correlated with HI and MN assays. A panel of pseudotypes belonging to influenza Groups 1 and 2, with a combination of reporter systems, was also employed for testing avian sera in order to support further application of pp-NT as an alternative valid assay that can improve avian vaccination efficacy testing, vaccine virus selection, and the reliability of reference sera.
Collapse
|
21
|
Muramatsu M, Yoshida R, Miyamoto H, Tomabechi D, Kajihara M, Maruyama J, Kimura T, Manzoor R, Ito K, Takada A. Heterosubtypic antiviral activity of hemagglutinin-specific antibodies induced by intranasal immunization with inactivated influenza viruses in mice. PLoS One 2013; 8:e71534. [PMID: 23977065 PMCID: PMC3745432 DOI: 10.1371/journal.pone.0071534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 06/28/2013] [Indexed: 01/27/2023] Open
Abstract
Influenza A virus subtypes are classified on the basis of the antigenicity of their envelope glycoproteins, hemagglutinin (HA; H1–H17) and neuraminidase. Since HA-specific neutralizing antibodies are predominantly specific for a single HA subtype, the contribution of antibodies to the heterosubtypic immunity is not fully understood. In this study, mice were immunized intranasally or subcutaneously with viruses having the H1, H3, H5, H7, H9, or H13 HA subtype, and cross-reactivities of induced IgG and IgA antibodies to recombinant HAs of the H1–H16 subtypes were analyzed. We found that both subcutaneous and intranasal immunizations induced antibody responses to multiple HAs of different subtypes, whereas IgA was not detected remarkably in mice immunized subcutaneously. Using serum, nasal wash, and trachea-lung wash samples of H9 virus-immunized mice, neutralizing activities of cross-reactive antibodies were then evaluated by plaque-reduction assays. As expected, no heterosubtypic neutralizing activity was detected by a standard neutralization test in which viruses were mixed with antibodies prior to inoculation into cultured cells. Interestingly, however, a remarkable reduction of plaque formation and extracellular release of the H12 virus, which was bound by the H9-induced cross-reactive antibodies, was observed when infected cells were subsequently cultured with the samples containing HA-specific cross-reactive IgA. This heterosubtypic plaque reduction was interfered when the samples were pretreated with anti-mouse IgA polyclonal serum. These results suggest that the majority of HA-specific cross-reactive IgG and IgA antibodies produced by immunization do not block cellular entry of viruses, but cross-reactive IgA may have the potential to inhibit viral egress from infected cells and thus to play a role in heterosubtypic immunity against influenza A viruses.
Collapse
Affiliation(s)
- Mieko Muramatsu
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Reiko Yoshida
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hiroko Miyamoto
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Daisuke Tomabechi
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Masahiro Kajihara
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Junki Maruyama
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Takashi Kimura
- Division of Molecular Pathobiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Rashid Manzoor
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Kimihito Ito
- Division of Bioinformatics, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Ayato Takada
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- * E-mail:
| |
Collapse
|
22
|
Characterization of the 2012 highly pathogenic avian influenza H7N3 virus isolated from poultry in an outbreak in Mexico: pathobiology and vaccine protection. J Virol 2013; 87:9086-96. [PMID: 23760232 DOI: 10.1128/jvi.00666-13] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In June of 2012, an H7N3 highly pathogenic avian influenza (HPAI) virus was identified as the cause of a severe disease outbreak in commercial laying chicken farms in Mexico. The purpose of this study was to characterize the Mexican 2012 H7N3 HPAI virus (A/chicken/Jalisco/CPA1/2012) and determine the protection against the virus conferred by different H7 inactivated vaccines in chickens. Both adult and young chickens intranasally inoculated with the virus became infected and died at between 2 and 4 days postinoculation (p.i.). High virus titers and viral replication in many tissues were demonstrated at 2 days p.i. in infected birds. The virus from Jalisco, Mexico, had high sequence similarity of greater than 97% to the sequences of wild bird viruses from North America in all eight gene segments. The hemagglutinin gene of the virus contained a 24-nucleotide insert at the hemagglutinin cleavage site which had 100% sequence identity to chicken 28S rRNA, suggesting that the insert was the result of nonhomologous recombination with the host genome. For vaccine protection studies, both U.S. H7 low-pathogenic avian influenza (LPAI) viruses and a 2006 Mexican H7 LPAI virus were tested as antigens in experimental oil emulsion vaccines and injected into chickens 3 weeks prior to challenge. All H7 vaccines tested provided ≥90% protection against clinical disease after challenge and decreased the number of birds shedding virus and the titers of virus shed. This study demonstrates the pathological consequences of the infection of chickens with the 2012 Mexican lineage H7N3 HPAI virus and provides support for effective programs of vaccination against this virus in poultry.
Collapse
|
23
|
Campbell-Harding G, Sawkins H, Bedke N, Holgate ST, Davies DE, Andrews AL. The innate antiviral response upregulates IL-13 receptor α2 in bronchial fibroblasts. J Allergy Clin Immunol 2013; 131:849-55. [PMID: 23069489 DOI: 10.1016/j.jaci.2012.08.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 08/13/2012] [Accepted: 08/22/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND IL-13 is key mediator of allergic inflammation in asthmatic patients. We have previously shown that the decoy receptor IL-13 receptor (IL-13R) α2 attenuates responses of fibroblasts to IL-13. Because the expression of IL-13Rα2 can be regulated by IFN-γ, a type II interferon, we hypothesized that innate antiviral responses characterized by type I interferon expression can also induce IL-13Rα2 expression. OBJECTIVE We sought to induce an innate antiviral response in primary fibroblasts using exposure to double-stranded RNA (dsRNA) and to examine the expression and function of IL-13Rα2. METHODS Primary human fibroblasts were cultured from endobronchial biopsy specimens obtained from healthy or asthmatic volunteers and challenged with dsRNA. Upregulation of IL-13Rα2 mRNA was measured by using real-time quantitative PCR, and cell-surface IL-13Rα2 protein expression was measured by using fluorescence-activated cell sorting. Eotaxin release was determined by means of ELISA. RESULTS Direct treatment with IFN-β led to an upregulation of IL-13Rα2. Exposure to dsRNA rapidly induced IFN-β mRNA in fibroblasts, and this was followed by significant induction of IL-13Rα2 mRNA and cell-surface protein expression, which was dependent on de novo protein synthesis. A neutralizing antibody to the IFN-α/β receptor blocked cell-surface expression of IL-13Rα2 in the presence of dsRNA. Pretreatment of fibroblasts with dsRNA led to attenuation of IL-13-stimulated eotaxin production. However, the presence of an IL-13Rα2 neutralizing antibody restored IL-13-stimulated eotaxin production in dsRNA-treated cells. CONCLUSION IFN-β induces IL-13Rα2 expression, leading to a consequential suppression of responsiveness to IL-13. These data suggest cross-talk between TH1 and TH2 pathways and point to an immunomodulatory role for IL-13Rα2 in human bronchial fibroblasts during viral infection.
Collapse
Affiliation(s)
- Gemma Campbell-Harding
- Academic Unit of Clinical and Experimental Sciences and the Southampton NIHR Respiratory Biomedical Research Unit, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Southampton, United Kingdom
| | | | | | | | | | | |
Collapse
|
24
|
Enhancement of T cell-mediated immune responses to whole inactivated influenza virus by chloroquine treatment in vivo. Vaccine 2013; 31:1717-24. [PMID: 23380456 DOI: 10.1016/j.vaccine.2013.01.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/16/2012] [Accepted: 01/21/2013] [Indexed: 11/21/2022]
Abstract
Current influenza vaccines induce poor cross-reactive CD8+ T cell responses. Cellular immunity is generally specific for epitopes that are remarkably conserved among different subtypes, suggesting that strategies to improve the cross-presentation of viral antigens by dendritic cells (DC) could elicit a broadly protective immune response. Previous studies have shown that limited proteolysis within the endocytic pathway can favorably influence antigen processing and thus immune responses. Herein, we demonstrate that chloroquine improves the cross-presentation of non-replicating influenza virus in vitro and T cell responses in mice following a single administration of inactivated HI-X31 virus. CD8+ T cells were also recruited to lymph nodes draining the site of infection and able to reduce viral load following pulmonary challenge with the heterologous PR8 virus. These findings may have implications for vaccination strategies aimed at improving the cross-presentation capacity of DCs and thus the size of effector and memory CD8+ T cells against influenza vaccines.
Collapse
|
25
|
Jang YH, Seong BL. Principles underlying rational design of live attenuated influenza vaccines. Clin Exp Vaccine Res 2012; 1:35-49. [PMID: 23596576 PMCID: PMC3623510 DOI: 10.7774/cevr.2012.1.1.35] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 05/23/2012] [Accepted: 06/10/2012] [Indexed: 12/18/2022] Open
Abstract
Despite recent innovative advances in molecular virology and the developments of vaccines, influenza virus remains a serious burden for human health. Vaccination has been considered a primary countermeasure for prevention of influenza infection. Live attenuated influenza vaccines (LAIVs) are particularly attracting attention as an effective strategy due to several advantages over inactivated vaccines. Cold-adaptation, as a classical means for attenuating viral virulence, has been successfully used for generating safe and effective donor strains of LAIVs against seasonal epidemics and occasional pandemics. Recently, the advent of reverse genetics technique expedited a variety of rational strategies to broaden the pool of LAIVs. Considering the breadth of antigenic diversity of influenza virus, the pool of LAIVs is likely to equip us with better options for controlling influenza pandemics. With a brief reflection on classical attenuating strategies used at the initial stage of development of LAIVs, especially on the principles underlying the development of cold-adapted LAIVs, we further discuss and outline other attenuation strategies especially with respect to the rationales for attenuation, and their practicality for mass production. Finally, we propose important considerations for a rational vaccine design, which will provide us with practical guidelines for improving the safety and effectiveness of LAIVs.
Collapse
Affiliation(s)
- Yo Han Jang
- Laboratory of Molecular Medicine, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | | |
Collapse
|
26
|
Kheiri MT, Jamali A, Shenagari M, Hashemi H, Sabahi F, Atyabi F, Saghiri R. Influenza virosome/DNA vaccine complex as a new formulation to induce intra-subtypic protection against influenza virus challenge. Antiviral Res 2012; 95:229-36. [PMID: 22809863 DOI: 10.1016/j.antiviral.2012.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 07/04/2012] [Accepted: 07/07/2012] [Indexed: 11/28/2022]
Abstract
Influenza virosome is one of the commercially available vaccines that have been used for a number of years. Like other influenza vaccines, the efficacy of the virosomal vaccine is significantly compromised when circulating viruses do not have a good match with vaccine strains due to antigenic drift or less frequent emergence of a pandemic virus. A major advantage of virosome over other influenza vaccine platforms is its intrinsic adjuvant activity and potential carrier capability which have been exploited in this study to broaden vaccine protectivity by incorporating a conserved component of influenza virus in seasonal vaccine formulation. Influenza nucleoprotein (NP)-encoding plasmid was adsorbed onto surface of influenza virosomes as a virosome/DNA vaccine complex. Mice were immunized with a single dose of the influenza virosome attached with the NP plasmid or NP plasmid alone where both influenza virosomes and NP gene were derived from influenza A virus H1N1 New/Caledonia strain. Analysis of the cellular immune responses showed that 5μg (10-fold reduced dose) of the NP plasmid attached to the virosomes induced T cell responses equivalent to those elicited by 50μg of NP plasmid alone as assessed by IFN-γ and granzyme B ELISPOT. Furthermore, the influenza virosome/NP plasmid complex protected mice against intra-subtypic challenge with the mouse adapted H1N1 PR8 virus, while mice immunized with the virosome alone did not survive. Results of hemagglutination inhibition test showed that the observed intra-subtypic cross-protection could not be attributed to neutralizing antibodies. These findings suggest that influenza virosomes could be equipped with an NP-encoding plasmid in a dose-sparing fashion to elicit anti-influenza cytotoxic immune responses and broaden the vaccine coverage against antigenic drift.
Collapse
|
27
|
Killed Bacillus subtilis spores as a mucosal adjuvant for an H5N1 vaccine. Vaccine 2012; 30:3266-77. [PMID: 22446640 DOI: 10.1016/j.vaccine.2012.03.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 02/21/2012] [Accepted: 03/09/2012] [Indexed: 01/15/2023]
Abstract
Heat killed spores of the Gram-positive bacterium Bacillus subtilis have been evaluated as a vaccine delivery system with mucosal adjuvant properties for influenza. Killed spores were able to bind H5N1 virions (NIBRG-14; clade 1) and, when intra-nasally administered to mice, resulting immune responses, both humoral and cell mediated, were enhanced compared to immunization with the virion alone. Levels of both systemic IgG and mucosal sIgA specific to the virion were elevated. Levels of IgG2a (a Th(1) antibody type) were strongly enhanced when the virion was co-administered with killed spores. Cytokine induction in stimulated splenocytes was also apparent indicating balanced T(h)1 and T(h)2 responses. Evidence of cross-neutralization of clade 2.2 viruses was shown. In a challenge experiment mice dosed two times with spores adsorbed with just 20 ng HA (hemagglutinin) of inactivated NIBRG-14 were fully protected against challenge with 20 LD(50) of H5N2 virus. Interestingly, partial protection (60%) was observed in animals dosed only with killed spores. Mice dosed only with killed spores were shown to be fully protected against H5N2 (5 LD(50)) infection indicating that innate immunity and its stimulation by spores may play an important role in protection. Supporting this killed spores were (i) shown to stimulate TLR-mediated expression of NF-κB, and (ii) able to recruit NK cells into lungs and induce maturation of DCs. This work demonstrates the potential and underlying mechanism for the use of bacterial spores as an adjuvant for H5N1 vaccination.
Collapse
|
28
|
Lee YN, Lee DH, Park JK, Lim TH, Youn HN, Yuk SS, Lee YJ, Mo IP, Sung HW, Lee JB, Park SY, Choi IS, Song CS. Isolation and characterization of a novel H9N2 influenza virus in Korean native chicken farm. Avian Dis 2012; 55:724-7. [PMID: 22313001 DOI: 10.1637/9774-050911-case.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
An outbreak of avian influenza, caused by an H9N2 low-pathogenic avian influenza virus (AIV), occurred in a chicken farm and caused severe economic losses due to mortality and diarrhea. AIV was isolated and identified in a sample from an affected native Korean chicken. Genetic analysis of the isolate revealed a high sequence similarity to genes of novel reassortant H9N2 viruses isolated from slaughterhouses and live bird markets in Korea in 2008 and 2009. Animal challenge studies demonstrated that the replication kinetics and pathogenicity of the isolate were considerably altered due to adaptation in chickens. Vaccine protection studies indicated that commercial vaccine was not able to prevent virus shedding and clinical disease when chickens were challenged with the isolate. These results suggest that the novel H9N2 virus possesses the capacity to replicate efficiently in the respiratory system against vaccination and to cause severe disease in domestic chickens. The results also highlight the importance of appropriate updating of vaccine strains, based on continuous surveillance data, to prevent the possibility of a new H9N2 epidemic in Korea.
Collapse
Affiliation(s)
- Yu-Na Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, 143-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Choi SK, Lee C, Lee KS, Choe SY, Mo IP, Seong RH, Hong S, Jeon SH. DNA aptamers against the receptor binding region of hemagglutinin prevent avian influenza viral infection. Mol Cells 2011; 32:527-33. [PMID: 22058017 PMCID: PMC3887679 DOI: 10.1007/s10059-011-0156-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/05/2011] [Accepted: 10/05/2011] [Indexed: 01/09/2023] Open
Abstract
The entrance of influenza virus into host cells is facilitated by the attachment of the globular region of viral hemagglutinin to the sialic acid receptors on host cell surfaces. In this study, we have cloned the cDNA fragment encoding the entire globular region (residues 101-257) of hemagglutinin of the H9N2 type avian influenza virus (A/ck/Korea/ms96/96). The protein segment (denoted as the H9 peptide), which was expressed and purified in E. coli, was used for the immunization of BALB/c mice to obtain the anti-H9 antiserum. To identify specific DNA aptamers with high affinity to H9 peptide, we conducted the SELEX method; 19 aptamers were newly isolated. A random mixture of these aptamers showed an increased level of binding affinity to the H9 peptide. The sequence alignment analysis of these aptamers revealed that 6 aptamers have highly conserved consensus sequences. Among these, aptamer C7 showed the highest similarity to the consensus sequences. Therefore, based on the C7 aptamer, we synthesized a new modified aptamer designated as C7-35M. This new aptamer showed strong binding capability to the viral particles. Furthermore, it could prevent MDCK cells from viral infection by strong binding to the viral particles. These results suggest that our aptamers can recognize the hemagglutinin protein of avian influenza virus and inhibit the binding of the virus to target receptors required for the penetration of host cells.
Collapse
Affiliation(s)
- Seung Kwan Choi
- Department of Life Science, Hallym University, Chuncheon 200-702, Korea
- These authors contributed equally to this work
| | - Changjin Lee
- Research Center for Functional Cellulomics, Seoul National University, Seoul 151-742, Korea
- These authors contributed equally to this work
| | - Kwang Soo Lee
- Department of Life Science, Hallym University, Chuncheon 200-702, Korea
| | - Soo-Young Choe
- School of Life Science, Chungbuk National University, Cheongju 361-764, Korea
| | - In Pil Mo
- College of Veterinary Medicine, Chungbuk National University, Cheongju 361-764, Korea
| | - Rho Hyun Seong
- Research Center for Functional Cellulomics, Seoul National University, Seoul 151-742, Korea
| | - Seokmann Hong
- Department of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul 143-747, Korea
| | - Sung Ho Jeon
- Department of Life Science, Hallym University, Chuncheon 200-702, Korea
| |
Collapse
|
30
|
Sublingual immunization with M2-based vaccine induces broad protective immunity against influenza. PLoS One 2011; 6:e27953. [PMID: 22140491 PMCID: PMC3227615 DOI: 10.1371/journal.pone.0027953] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 10/28/2011] [Indexed: 12/12/2022] Open
Abstract
Background The ectodomain of matrix protein 2 (M2e) of influenza A virus is a rationale target antigen candidate for the development of a universal vaccine against influenza as M2e undergoes little sequence variation amongst human influenza A strains. Vaccine-induced M2e-specific antibodies (Abs) have been shown to display significant cross-protective activity in animal models. M2e-based vaccine constructs have been shown to be more protective when administered by the intranasal (i.n.) route than after parenteral injection. However, i.n. administration of vaccines poses rare but serious safety issues associated with retrograde passage of inhaled antigens and adjuvants through the olfactory epithelium. In this study, we examined whether the sublingual (s.l.) route could serve as a safe and effective alternative mucosal delivery route for administering a prototype M2e-based vaccine. The mechanism whereby s.l. immunization with M2e vaccine candidate induces broad protection against infection with different influenza virus subtypes was explored. Methods and Results A recombinant M2 protein with three tandem copies of the M2e (3M2eC) was expressed in Escherichia coli. Parenteral immunizations of mice with 3M2eC induced high levels of M2e-specific serum Abs but failed to provide complete protection against lethal challenge with influenza virus. In contrast, s.l. immunization with 3M2eC was superior for inducing protection in mice. In the latter animals, protection was associated with specific Ab responses in the lungs. Conclusions The results demonstrate that s.l. immunization with 3M2eC vaccine induced airway mucosal immune responses along with broad cross-protective immunity to influenza. These findings may contribute to the understanding of the M2-based vaccine approach to control epidemic and pandemic influenza infections.
Collapse
|
31
|
Sun Y, Pu J, Fan L, Sun H, Wang J, Zhang Y, Liu L, Liu J. Evaluation of the protective efficacy of a commercial vaccine against different antigenic groups of H9N2 influenza viruses in chickens. Vet Microbiol 2011; 156:193-9. [PMID: 22019289 DOI: 10.1016/j.vetmic.2011.10.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 09/23/2011] [Accepted: 10/03/2011] [Indexed: 12/15/2022]
Abstract
Despite the long-term vaccination programs implemented in China, H9N2 avian influenza viruses (AIVs) continue to persist in chicken populations, even in vaccinated flocks. We previously demonstrated that H9N2 AIV isolated from chickens in China also underwent antigenic drift and evolved into distinct antigenic groups (C, D and E). To understand whether antigenic drift of viruses away from the vaccine strain partially contributed to the circulation of H9N2 AIV in China, we evaluated the protective efficacy of a commercial vaccine against different antigenic groups of H9N2 AIV. Challenge experiments using vaccinated chickens indicated that the vaccine prevented shedding of antigenic group C viruses, but not those of the more recent groups D and E. Vaccinated chickens, even those with vaccine-induced HI titers of 1:1024, shed virus after being infected with A/chicken/Shandong/ZB/2007, a representative virus of antigenic group D. Genetic analysis showed that the representative viruses of antigenic groups D and E possessed greater numbers of amino acid substitutions in the hemagglutinin protein compared to the vaccine strain and the antigenic group C virus, and many of which were located in antigenic sites. Our results indicated that the persistence of H9N2 AIV in China might be due to incomplete vaccine protection, and that the avian influenza vaccine should be regularly evaluated and updated to maintain optimal protection. Furthermore, the avian influenza vaccination policy also needs to be re-assessed, and increased veterinary biosecurity on farms, rather than vaccine application alone, should be implemented to prevent and control avian influenza.
Collapse
Affiliation(s)
- Yipeng Sun
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Gueller S, Allwinn R, Mousset S, Martin H, Wieters I, Herrmann E, Serve H, Bickel M, Bug G. Enhanced Immune Response after a Second Dose of an AS03-Adjuvanted H1N1 Influenza A Vaccine in Patients after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2011; 17:1546-50. [DOI: 10.1016/j.bbmt.2011.02.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 02/07/2011] [Indexed: 10/18/2022]
|
33
|
Ichihashi T, Yoshida R, Sugimoto C, Takada A, Kajino K. Cross-protective peptide vaccine against influenza A viruses developed in HLA-A*2402 human immunity model. PLoS One 2011; 6:e24626. [PMID: 21949735 PMCID: PMC3176274 DOI: 10.1371/journal.pone.0024626] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 08/15/2011] [Indexed: 11/25/2022] Open
Abstract
Background The virus-specific cytotoxic T lymphocyte (CTL) induction is an important target for the development of a broadly protective human influenza vaccine, since most CTL epitopes are found on internal viral proteins and relatively conserved. In this study, the possibility of developing a strain/subtype-independent human influenza vaccine was explored by taking a bioinformatics approach to establish an immunogenic HLA-A24 restricted CTL epitope screening system in HLA-transgenic mice. Methodology/Principal Findings HLA-A24 restricted CTL epitope peptides derived from internal proteins of the H5N1 highly pathogenic avian influenza A virus were predicted by CTL epitope peptide prediction programs. Of 35 predicted peptides, six peptides exhibited remarkable cytotoxic activity in vivo. More than half of the mice which were subcutaneously vaccinated with the three most immunogenic and highly conserved epitopes among three different influenza A virus subtypes (H1N1, H3N2 and H5N1) survived lethal influenza virus challenge during both effector and memory CTL phases. Furthermore, mice that were intranasally vaccinated with these peptides remained free of clinical signs after lethal virus challenge during the effector phase. Conclusions/Significance This CTL epitope peptide selection system can be used as an effective tool for the development of a cross-protective human influenza vaccine. Furthermore this vaccine strategy can be applicable to the development of all intracellular pathogens vaccines to induce epitope-specific CTL that effectively eliminate infected cells.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cross Protection/immunology
- Epitopes/immunology
- HLA-A24 Antigen/genetics
- HLA-A24 Antigen/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H1N1 Subtype/enzymology
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza A Virus, H3N2 Subtype/enzymology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/pathogenicity
- Influenza A Virus, H5N1 Subtype/enzymology
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza A virus/enzymology
- Influenza A virus/immunology
- Influenza A virus/pathogenicity
- Influenza Vaccines/immunology
- Lung/virology
- Mice
- Mice, Transgenic
- Models, Animal
- Neuraminidase/immunology
- Reproducibility of Results
- T-Lymphocytes, Cytotoxic/immunology
- Time Factors
- Vaccination
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Toru Ichihashi
- Department of Collaboration and Education, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Reiko Yoshida
- Department of Global Epidemiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Chihiro Sugimoto
- Department of Collaboration and Education, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Ayato Takada
- Department of Global Epidemiology, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
| | - Kiichi Kajino
- Department of Collaboration and Education, Hokkaido University Research Center for Zoonosis Control, Sapporo, Japan
- * E-mail:
| |
Collapse
|
34
|
Improvement of the trivalent inactivated flu vaccine using PapMV nanoparticles. PLoS One 2011; 6:e21522. [PMID: 21747909 PMCID: PMC3126827 DOI: 10.1371/journal.pone.0021522] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/30/2011] [Indexed: 02/06/2023] Open
Abstract
Commercial seasonal flu vaccines induce production of antibodies directed mostly towards hemaglutinin (HA). Because HA changes rapidly in the circulating virus, the protection remains partial. Several conserved viral proteins, e.g., nucleocapsid (NP) and matrix proteins (M1), are present in the vaccine, but are not immunogenic. To improve the protection provided by these vaccines, we used nanoparticles made of the coat protein of a plant virus (papaya mosaic virus; PapMV) as an adjuvant. Immunization of mice and ferrets with the adjuvanted formulation increased the magnitude and breadth of the humoral response to NP and to highly conserved regions of HA. They also triggered a cellular mediated immune response to NP and M1, and long-lasting protection in animals challenged with a heterosubtypic influenza strain (WSN/33). Thus, seasonal flu vaccine adjuvanted with PapMV nanoparticles can induce universal protection to influenza, which is a major advancement when facing a pandemic.
Collapse
|
35
|
Genetic characterization of highly pathogenic H5N1 avian influenza viruses isolated from poultry farms in Egypt. Virus Genes 2011; 43:272-80. [DOI: 10.1007/s11262-011-0633-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 05/28/2011] [Indexed: 10/18/2022]
|
36
|
Kapczynski DR, Liljebjelke K, Kulkarni G, Hunt H, Jiang HJ, Petkov D. Cross reactive cellular immune responses in chickens previously exposed to low pathogenic avian influenza. BMC Proc 2011; 5 Suppl 4:S13. [PMID: 21645292 PMCID: PMC3108207 DOI: 10.1186/1753-6561-5-s4-s13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Avian influenza (AI) infection in poultry can result in high morbidity and mortality, and negatively affect international trade. Because most AI vaccines used for poultry are inactivated, our knowledge of immunity against AI is based largely on humoral immune responses. In fact, little is known about cellular immunity following a primary AI infection in poultry, especially regarding cytotoxic T lymphocytes (CTL’s). Methods In these studies, major histocompatibility complex (MHC)-defined (B2/B2) chickens were infected with low pathogenic AI (LPAI) H9N2 and clinical signs of disease were monitored over a two weeks period. Splenic lymphocytes from infected and naïve birds were examined for cross reactivity against homologous and heterologous (H7N2) LPAI by ex vivo stimulation. Cellular immunity was determined by cytotoxic lysis of B2/B2 infected lung target cells and proliferation of T cells following exposure to LPAI. Results Infection with H9N2 resulted in statistically significant weight loss compared to sham-infected birds. Splenic lymphocytes derived from H9N2-infected birds displayed lysis of both homologous (H9N2) and heterologous (H7N2) infected target cells, whereas lymphocytes obtained from sham-infected birds did not. T cell proliferation was determined to be highest when exposed to the homologous virus. Conclusions Taken together these data extend the findings that cellular immunity, including CTL’s, is cross reactive against heterologous isolates of AI and contribute to protection following infection.
Collapse
Affiliation(s)
- Darrell R Kapczynski
- Southeast Poultry Research Laboratory, Agricultural Research Service, USA, Department of Agriculture, 934 College Station Road, Athens, GA 30605, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Murphy EA, Davis JM, McClellan JL, Carmichael MD, Rooijen NV, Gangemi JD. Susceptibility to Infection and Inflammatory Response Following Influenza Virus (H1N1, A/PR/8/34) Challenge: Role of Macrophages. J Interferon Cytokine Res 2011; 31:501-8. [DOI: 10.1089/jir.2010.0143] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- E. Angela Murphy
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina
| | - J. Mark Davis
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Jamie L. McClellan
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Martin D. Carmichael
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina
| | - Nico Van Rooijen
- Department of Cell Biology and Immunology, Vrije Universiteit of Amsterdam, Amsterdam, Netherlands
| | - J. David Gangemi
- Department of Microbiology and Molecular Medicine, Clemson University, Clemson, South Carolina
| |
Collapse
|
38
|
Benson JM, Wolf ML, Kajon A, Tibbetts BM, Bourdelais AJ, Baden DG, March TH. Brevetoxin inhalation alters the pulmonary response to influenza A in the male F344 rat. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2011; 74:313-324. [PMID: 21240731 PMCID: PMC3444170 DOI: 10.1080/15287394.2010.519316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Epidemiological studies demonstrated that the number of emergency-room visits for respiratory indications increases during periods of Florida Red Tides. The purpose of this study was to examine whether or not repeated brevetoxin inhalation, as may occur during a Florida Red Tide, affects pulmonary responses to influenza A. Male F344 rats were divided into four groups: (1) sham aerosol/no influenza; (2) sham aerosol/influenza; (3) brevetoxin/no influenza; and (4) brevetoxin/influenza. Animals were exposed by nose-only inhalation to vehicle or 50 μg brevetoxin-3/m3, 2 h/d for 12 d. On d 6 of aerosol exposure, groups 2 and 4 were administered 10,000 plaque-forming units of influenza A, strain HKX-31 (H3N2), by intratracheal instillation. Subgroups were euthanized at 2, 4, and 7 d post influenza treatment. Lungs were evaluated for viral load, cytokine content, and histopathologic changes. Influenza virus was cleared from the lungs over the 7-d period; however, there was significantly more virus remaining in the group 4 lungs compared to group 2. Influenza virus significantly increased interleukins-1α and -6 and monocyte chemotactic protein-1 in lung; brevetoxin exposure significantly enhanced the influenza-induced response. At 7 d, the severity of perivascular and peribronchiolar inflammatory cell infiltrates was greatest in group 4. Bronchiolitis persisted, with low incidence and severity, only in group 4 at d 7. These results suggest that repeated inhalation exposure to brevetoxin may delay virus particle clearance and recovery from influenza A infection in the rat lung.
Collapse
Affiliation(s)
- Janet M. Benson
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM 87108
| | - Molly L. Wolf
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM 87108
| | - Adriana Kajon
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM 87108
| | - Brad M. Tibbetts
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM 87108
| | - Andrea J. Bourdelais
- Center for Marine Science Research, University of North Carolina at Wilmington, 5600 Marvin K. Moss Lane, Wilmington, NC 28409
| | - Daniel G. Baden
- Center for Marine Science Research, University of North Carolina at Wilmington, 5600 Marvin K. Moss Lane, Wilmington, NC 28409
| | - Thomas H. March
- Lovelace Respiratory Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM 87108
| |
Collapse
|
39
|
Bickel M, von Hentig N, Wieters I, Khaykin P, Nisius G, Haberl A, Stephan C, Herrmann E, Doerr HW, Brodt HR, Allwinn R. Immune Response after Two Doses of the Novel Split Virion, Adjuvanted Pandemic H1N1 Influenza A Vaccine in HIV-1-Infected Patients. Clin Infect Dis 2011; 52:122-7. [DOI: 10.1093/cid/ciq003] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
40
|
The 2009 pandemic H1N1 and triple-reassortant swine H1N1 influenza viruses replicate efficiently but elicit an attenuated inflammatory response in polarized human bronchial epithelial cells. J Virol 2010; 85:686-96. [PMID: 21047961 DOI: 10.1128/jvi.01568-10] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pandemic H1N1 virus of 2009 (2009 H1N1) produced a spectrum of disease ranging from mild illness to severe illness and death. Respiratory symptoms were frequently associated with virus infection, with relatively high rate of gastrointestinal symptoms reported. To better understand 2009 H1N1 virus pathogenesis in humans, we studied virus and host responses following infection of two cell types: polarized bronchial and pharyngeal epithelial cells, which exhibit many features of the human airway epithelium, and colon epithelial cells to serve as a human intestinal cell model. Selected 2009 H1N1 viruses were compared to both seasonal H1N1 and triple-reassortant swine H1N1 influenza viruses that have circulated among North American pigs since before the 2009 pandemic. All H1N1 viruses replicated productively in airway cells; however, in contrast to seasonal H1N1 virus infection, infection with the 2009 H1N1 and triple-reassortant swine H1N1 viruses resulted in an attenuated inflammatory response, a weaker interferon response, and reduced cell death. Additionally, the H1N1 viruses of swine origin replicated less efficiently at the temperature of the human proximal airways (33°C). We also observed that the 2009 H1N1 viruses replicated to significantly higher titers than seasonal H1N1 virus in polarized colon epithelial cells. These studies reveal that in comparison to seasonal influenza virus, H1N1 viruses of swine origin poorly activate multiple aspects of the human innate response, which may contribute to the virulence of these viruses. In addition, their less efficient replication at human upper airway temperatures has implications for the understanding of pandemic H1N1 virus adaptation to humans.
Collapse
|
41
|
Immune responses to common respiratory pathogens: problems and perspectives in equine immunology. Equine Vet J 2010. [PMCID: PMC7163740 DOI: 10.1111/j.2042-3306.1991.tb04751.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
Low rate of seroconversion after vaccination with a split virion, adjuvanted pandemic H1N1 influenza vaccine in HIV-1-infected patients. AIDS 2010; 24:F31-5. [PMID: 20559034 DOI: 10.1097/qad.0b013e3283398da1] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine rates of seroconversion after single vaccination with a novel split virion, inactivated, adjuvanted pandemic H1N1 influenza vaccine (A/California/7/2009) in HIV-1-infected patients (ClinicalTrials.gov Identifier: NCT01017172). DESIGN Single center diagnostic study. SETTING Institutional HIV outpatient department of an urban university clinic. PARTICIPANTS Adult HIV-1-infected individuals. INTERVENTION Serum samples were taken before and 21 days after vaccination. MAIN OUTCOME MEASURES Antibody titers determined by hemagglutination inhibition assay. Seroconversion to vaccination was defined by either an antibody titer of 1: 10 or less before and of at least 1: 40 after or at least 1: 10 before and at least four-fold increase in antibody titer 21 days after single vaccination. RESULTS One hundred and sixty patients (125 men/35 women) were analyzed. Before vaccination, 23 patients (14.4%) had a hemagglutination inhibition assay titer of at least 1: 40. A median of 22 +/- 3 days after vaccination, 110 (69%) patients seroconverted. Seroconverters were younger (45.1 +/- 10.0 vs. 48.8 +/- 11.3 years; P = 0.04), had a higher CD4 cell count (532 +/- 227 vs. 475 +/- 281 cells/microl; P = 0.03) and were more likely to have received a previous H5N1 vaccination in 2009 (25 vs. 8%; P = 0.02) when compared to nonresponders. No other significant differences were found comparing the two groups (prevaccination hemagglutination inhibition assay titer of > or =1: 40, AIDS, HAART, HIV RNA PCR <50 copies/ml or CD4 nadir, CD4 and CD8 percentage, sex, BMI, chronic hepatitis B or C). CONCLUSION Seroconversion after one dose of a split virion, inactivated, adjuvanted pandemic H1N1 influenza vaccine of HIV-infected patients was 69%. Studies to investigate whether a second dose of the vaccine will increase seroconversion rate are needed.
Collapse
|
43
|
Eggert D, Thomas C, Spackman E, Pritchard N, Rojo F, Bublot M, Swayne DE. Characterization and efficacy determination of commercially available Central American H5N2 avian influenza vaccines for poultry. Vaccine 2010; 28:4609-15. [DOI: 10.1016/j.vaccine.2010.04.081] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 04/23/2010] [Accepted: 04/26/2010] [Indexed: 10/19/2022]
|
44
|
Liljebjelke KA, Petkov DI, Kapczynski DR. Mucosal vaccination with a codon-optimized hemagglutinin gene expressed by attenuated Salmonella elicits a protective immune response in chickens against highly pathogenic avian influenza. Vaccine 2010; 28:4430-7. [PMID: 20406663 DOI: 10.1016/j.vaccine.2010.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 03/25/2010] [Accepted: 04/05/2010] [Indexed: 10/19/2022]
Abstract
The purpose of this study was to evaluate clinical protection from challenge conferred by two attenuated Salmonella enteria serovar typhimurium vaccine strains expressing the hemagglutinin (HA1) gene from a highly pathogenic avian influenza (HPAI) H5N1 (A/whooper swan/Mongolia/3/2005), under control of the anaerobically inducible nir15 promoter. Two-week-old White Leghorn chickens were immunized by oral gavage with one milliliter doses of >109 Salmonella colony-forming units once weekly for 4 weeks prior to challenge. Expression of recombinant protein was confirmed via Western blot. Serum and mucosal gavage samples were collected prior to, and following immunization and antibodies against avian influenza HA were confirmed by Western blot and hemagglutination-inhibition (HI) assay. Chickens were challenged with homologous (A/whooper swan/Mongolia/3/2005), or heterologous (A/Chicken/Queretaro/14588-19/95) HPAI virus strains. Chickens immunized with attenuated Salmonella strains containing plasmid expression vector (pTETnir15HA) demonstrated a statistically significant increase in survival compared to control groups. Results provide evidence of effectiveness of attenuated Salmonella strains for delivery of recombinant avian influenza HA antigens and induction of mucosal and systemic immune responses protective against lethal challenge with HPAI.
Collapse
|
45
|
Cytotoxic T cells are the predominant players providing cross-protective immunity induced by {gamma}-irradiated influenza A viruses. J Virol 2010; 84:4212-21. [PMID: 20164231 DOI: 10.1128/jvi.02508-09] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously demonstrated that a single dose of nonadjuvanted intranasal gamma-irradiated influenza A virus can provide robust protection in mice against both homologous and heterosubtypic challenges, including challenge with an H5N1 avian virus strain. We investigated the mechanism behind the observed cross-protection to define which arms of the adaptive immune response are involved in mediating this protection. Studies with gene knockout mice showed the cross-protective immunity to be mediated mainly by T cells and to be dependent on the cytolytic effector molecule perforin. Adoptive transfer of memory T cells from immunized mice, but not of memory B cells, protected naïve recipients against lethal heterosubtypic influenza virus challenge. Furthermore, gamma-irradiated influenza viruses induced cross-reactive Tc-cell responses but not cross-neutralizing or cross-protective antibodies. In addition, histological analysis showed reduced lung inflammation in vaccinated mice compared to that in unvaccinated controls following heterosubtypic challenge. This reduced inflammation was associated with enhanced early recruitment of T cells, both CD4(+) and CD8(+), and with early influenza virus-specific cytotoxic T-cell responses. Therefore, cross-protective immunity induced by vaccination with gamma-irradiated influenza A virus is mediated mainly by Tc-cell responses.
Collapse
|
46
|
Furuya Y, Regner M, Lobigs M, Koskinen A, Müllbacher A, Alsharifi M. Effect of inactivation method on the cross-protective immunity induced by whole 'killed' influenza A viruses and commercial vaccine preparations. J Gen Virol 2010; 91:1450-60. [PMID: 20147516 DOI: 10.1099/vir.0.018168-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have recently shown that intranasal (i.n.) administration of gamma-irradiated A/PR/8 [A/Puerto Rico/8/34 (H1N1)] protects mice against lethal avian influenza A/Vietnam/1203/2004 (H5N1) and other heterosubtypic influenza A infections. Here, we used gamma-irradiated, formalin- and UV-inactivated A/PC [A/Port Chalmers/1/73 (H3N2)] virus preparations and compared their ability to induce both homologous and heterosubtypic protective immunity. Our data show that, in contrast to i.n. vaccination with formalin- or UV-inactivated virus, or the present commercially available trivalent influenza vaccine, a single dose of gamma-ray-inactivated A/PC (gamma-A/PC) conferred significant protection in mice against both homologous and heterosubtypic virus challenges. A multiple immunization regime was required for formalin-inactivated virus preparations to induce protective immunity against a homotypic virus challenge, but did not induce influenza A strain cross-protective immunity. The highly immunogenic gamma-A/PC, but not formalin- or UV-inactivated A/PC, nor the currently available subvirion vaccine, elicited cytotoxic T-cell responses that are most likely responsible for the cross-protective and long-lasting immunity against highly lethal influenza A infections in mice. Finally, freeze-drying of gamma-A/PC did not affect the ability to induce cross-protective immunity.
Collapse
Affiliation(s)
- Yoichi Furuya
- Viral Immunology and Molecular Virology, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
A key question in pandemic influenza is the relative roles of innate immunity and target cell depletion in limiting primary infection and modulating pathology. Here, we model these interactions using detailed data from equine influenza virus infection, combining viral and immune (type I interferon) kinetics with estimates of cell depletion. The resulting dynamics indicate a powerful role for innate immunity in controlling the rapid peak in virus shedding. As a corollary, cells are much less depleted than suggested by a model of human influenza based only on virus-shedding data. We then explore how differences in the influence of viral proteins on interferon kinetics can account for the observed spectrum of virus shedding, immune response, and influenza pathology. In particular, induction of high levels of interferon ("cytokine storms"), coupled with evasion of its effects, could lead to severe pathology, as hypothesized for some fatal cases of influenza.
Collapse
|
48
|
Immunization by influenza virus-like particles protects aged mice against lethal influenza virus challenge. Antiviral Res 2009; 84:215-24. [PMID: 19772876 DOI: 10.1016/j.antiviral.2009.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 08/31/2009] [Accepted: 09/03/2009] [Indexed: 11/24/2022]
Abstract
Influenza virus-like particles (VLPs) were produced in Sf9 insect cells by co-expressing the matrix protein M1 and the surface glycoproteins hemagglutinin (HA) and neuraminidase (NA) using the recombinant baculovirus expression system. The VLPs were morphologically similar to influenza virions. Both HA and NA proteins were incorporated into VLPs and these proteins retained their functional activities. Further, influenza VLPs but not inactivated influenza viruses (IIV) stimulated secretion of inflammatory cytokines from mouse bone marrow-derived dendritic cells (BMDC). Immunogenicity of influenza VLPs and their protective efficacies against lethal influenza virus challenge were evaluated in young and aged mice. Immunization with influenza VLPs induced strong antibody responses against HA that inhibited hemagglutination by influenza virus, similar to IIV vaccines. Compared to young mice, antibody responses in aged mice immunized with a low dose of either influenza VLPs or IIV vaccines exhibited markedly reduced avidity for HA. However, immunization of aged mice with a high dose of influenza VLPs induced antibody responses with high avidity similar to those in young mice. Furthermore, all vaccinated animals survived a lethal challenge by a mouse-adapted influenza virus (A/PR/8/34), indicating that influenza VLPs are highly efficacious for protection against influenza virus infection in both young and aged mice.
Collapse
|
49
|
Alsharifi M, Furuya Y, Bowden TR, Lobigs M, Koskinen A, Regner M, Trinidad L, Boyle DB, Müllbacher A. Intranasal flu vaccine protective against seasonal and H5N1 avian influenza infections. PLoS One 2009; 4:e5336. [PMID: 19401775 PMCID: PMC2671162 DOI: 10.1371/journal.pone.0005336] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 04/01/2009] [Indexed: 11/18/2022] Open
Abstract
Background Influenza A (flu) virus causes significant morbidity and mortality worldwide, and current vaccines require annual updating to protect against the rapidly arising antigenic variations due to antigenic shift and drift. In fact, current subunit or split flu vaccines rely exclusively on antibody responses for protection and do not induce cytotoxic T (Tc) cell responses, which are broadly cross-reactive between virus strains. We have previously reported that γ-ray inactivated flu virus can induce cross-reactive Tc cell responses. Methodology/Principal Finding Here, we report that intranasal administration of purified γ-ray inactivated human influenza A virus preparations (γ-Flu) effectively induces heterotypic and cross-protective immunity. A single intranasal administration of γ-A/PR8[H1N1] protects mice against lethal H5N1 and other heterotypic infections. Conclusions/Significance Intranasal γ-Flu represents a unique approach for a cross-protective vaccine against both seasonal as well as possible future pandemic influenza A virus infections.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Cross Reactions
- Female
- Gamma Rays
- Humans
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/pathogenicity
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- RNA, Viral/analysis
- RNA, Viral/genetics
- T-Lymphocytes, Cytotoxic/immunology
- Vaccination/methods
- Vaccines, Inactivated/administration & dosage
Collapse
Affiliation(s)
- Mohammed Alsharifi
- Division of Immunology and Genetics, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- Microbiology and Infectious Diseases, Institute of Medical and Veterinary Science, Adelaide, South Australia, Australia
- * E-mail: (MA); (AM)
| | - Yoichi Furuya
- Division of Immunology and Genetics, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Timothy R. Bowden
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Mario Lobigs
- Division of Immunology and Genetics, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Aulikki Koskinen
- Division of Immunology and Genetics, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Matthias Regner
- Division of Immunology and Genetics, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Lee Trinidad
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - David B. Boyle
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Arno Müllbacher
- Division of Immunology and Genetics, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- * E-mail: (MA); (AM)
| |
Collapse
|
50
|
Intranasal vaccination with 1918 influenza virus-like particles protects mice and ferrets from lethal 1918 and H5N1 influenza virus challenge. J Virol 2009; 83:5726-34. [PMID: 19321609 DOI: 10.1128/jvi.00207-09] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Influenza vaccines capable of inducing cross-reactive or heterotypic immunity could be an important first line of prevention against a novel subtype virus. Influenza virus-like particles (VLPs) displaying functional viral proteins are effective vaccines against replication-competent homologous virus, but their ability to induce heterotypic immunity has not been adequately tested. To measure VLP vaccine efficacy against a known influenza pandemic virus, recombinant VLPs were generated from structural proteins of the 1918 H1N1 virus. Mucosal and traditional parenteral administrations of H1N1 VLPs were compared for the ability to protect against the reconstructed 1918 virus and a highly pathogenic avian H5N1 virus isolated from a fatal human case. Mice that received two intranasal immunizations of H1N1 VLPs were largely protected against a lethal challenge with both the 1918 virus and the H5N1 virus. In contrast, mice that received two intramuscular immunizations of 1918 VLPs were only protected against a homologous virus challenge. Mucosal vaccination of mice with 1918 VLPs induced higher levels of cross-reactive immunoglobulin G (IgG) and IgA antibodies than did parenteral vaccination. Similarly, ferrets mucosally vaccinated with 1918 VLPs completely survived a lethal challenge with the H5N1 virus, while only a 50% survival rate was observed in parenterally vaccinated animals. These results suggest a strategy of VLP vaccination against a pandemic virus and one that stimulates heterotypic immunity against an influenza virus strain with threatening pandemic potential.
Collapse
|