1
|
Del Bene A, D'Aniello A, Mottola S, Mazzarella V, Cutolo R, Campagna E, Benedetti R, Altucci L, Cosconati S, Di Maro S, Messere A. From genetic code to global health: the impact of nucleic acid vaccines on disease prevention and treatment. RSC Med Chem 2025:d5md00032g. [PMID: 40337306 PMCID: PMC12053015 DOI: 10.1039/d5md00032g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/19/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccinology has revolutionized modern medicine, delivering groundbreaking solutions to prevent and control infectious diseases while pioneering innovative strategies to tackle non-infectious challenges, including cancer. Traditional vaccines faced inherent limitations, driving the evolution of next-generation vaccines such as subunit vaccines, peptide-based vaccines, and nucleic acid-based platforms. Among these, nucleic acid-based vaccines, including DNA and mRNA technologies, represent a major innovation. Pioneering studies in the 1990s demonstrated their ability to elicit immune responses by encoding specific antigens. Recent advancements in delivery systems and molecular engineering have overcome initial challenges, enabling their rapid development and clinical success. This review explores nucleic acid-based vaccines, including chemically modified variants, by examining their mechanisms, structural features, and therapeutic potential, while underscoring their pivotal role in modern immunization strategies and expanding applications across contemporary medicine.
Collapse
Affiliation(s)
- Alessandra Del Bene
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | | | - Salvatore Mottola
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Vincenzo Mazzarella
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Roberto Cutolo
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Erica Campagna
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Vico Luigi De Crecchio 1 80138 Naples Italy
- Program of Medical Epigenetics, Vanvitelli Hospital 80138 Naples Italy
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Vico Luigi De Crecchio 1 80138 Naples Italy
- Program of Medical Epigenetics, Vanvitelli Hospital 80138 Naples Italy
- Biogem Institute of Molecular and Genetic Biology 83031 Ariano Irpino Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Vico Luigi De Crecchio 1 80138 Naples Italy
| | - Sandro Cosconati
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Salvatore Di Maro
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Anna Messere
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| |
Collapse
|
2
|
Mahooti M, Abdolalipour E, Farahmand B, Shirian S, Ghaemi A. Immunomodulatory effects of probiotic Lactobacillus casei on GM-CSF-adjuvanted influenza DNA vaccine. Future Virol 2022. [DOI: 10.2217/fvl-2021-0327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: This study investigates the protective efficacy of influenza DNA vaccine combined with a granulocyte macrophage-colony stimulating factor (GM-CSF) adjuvant, and probiotic Lactobacillus casei, an oral immunomodulator, in a BALB/c mice. Materials & methods: The mice were immunized with HA1 DNA vaccine along with GM-CSF and probiotic twice within a one-week interval. Results: The results showed that both adjuvants exert a synergistic effect in enhancing the humoral and cellular immune responses of the DNA vaccine. This combination also deceased IL-6 and IL-17A levels in the lung homogenates. The protection patterns were closely associated with influenza virus-specific splenocyte proliferative and serum IgG antibody (Ab) responses. Conclusion: The Findings demonstrate L. casei modulate balanced Th1/Th2 immune responses toward HA1 DNA vaccine adjuvanted by GM-CSF.
Collapse
Affiliation(s)
- Mehran Mahooti
- Department of Influenza & other respiratory viruses, Pasteur Institute of Iran, Tehran, 1316943551, Iran
- Department of Biotechnology, Iranian Research Organization for Science & Technology, Tehran, Iran
| | - Elahe Abdolalipour
- Department of Influenza & other respiratory viruses, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Behrokh Farahmand
- Department of Influenza & other respiratory viruses, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Amir Ghaemi
- Department of Influenza & other respiratory viruses, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| |
Collapse
|
3
|
Chen YP, Lin CC, Xie YX, Chen CY, Qiu JT. Enhancing immunogenicity of HPV16 E 7 DNA vaccine by conjugating codon-optimized GM-CSF to HPV16 E 7 DNA. Taiwan J Obstet Gynecol 2021; 60:700-705. [PMID: 34247810 DOI: 10.1016/j.tjog.2021.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE To generate immunity against human papillomavirus (HPV), the use of a recombinant DNA vaccine to carry an appropriate target gene is a promising and cost-effective approach. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a potent immunomodulatory cytokine that enhances the efficacy of vaccines by promoting the development and prolongation of humoral and cellular immunity. In this study, we linked codon-optimized GM-CSF (cGM-CSF) to the HPV16 E7 sequence as fused protein and evaluated the immunogenic potential of this DNA vaccine. MATERIALS AND METHODS We have demonstrated that cGM-CSF enhanced immunity against tumor challenges by generating and promoting the proliferation of HPV16 E7-specific CD8+ T cells, which secrete IFN-γ in the murine model. In this study, we aimed to evaluate the immunogenic potential of DNA vaccine that constructed by linking codon-optimized GM-CSF to HPV16 E7 sequence in the animal model. We study the half-life of RNA decay and cellular location of HPV16 E7 by Q-PCR and Western blot. We also assess immune response in the animal model by flow cytometry and ELISA. RESULTS The cGM-CSF-E7 sequence increased and extended the expression of E7 mRNA, in comparison with the E7 sequence alone. Mice vaccinated with the cGM-CSF-E7 DNA vaccine exhibited a slower rate of tumor growth than those vaccinated with the unconjugated E7 DNA vaccine. We also found that the CD4 and CD8+ T cells from these mice showed strong secretion of IFN-γ. CONCLUSION Through in vivo antibody depletion experiments, we demonstrated that both CD4+ and CD8+ T cells play an important role in the suppression of tumor growth.
Collapse
Affiliation(s)
- Yi-Pin Chen
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan, ROC; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chu-Chi Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Yu-Xin Xie
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chia-Yuan Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - J Timothy Qiu
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC; College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
4
|
Tannig P, Peter AS, Lapuente D, Klessing S, Schmidt A, Damm D, Tenbusch M, Überla K, Temchura V. Genetic Co-Administration of Soluble PD-1 Ectodomains Modifies Immune Responses against Influenza A Virus Induced by DNA Vaccination. Vaccines (Basel) 2020; 8:vaccines8040570. [PMID: 33019546 PMCID: PMC7712647 DOI: 10.3390/vaccines8040570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/08/2023] Open
Abstract
Due to the low efficacy and the need for seasonal adaptation of currently licensed influenza A vaccines, the importance of alternative vaccination strategies is increasingly recognized. Considering that DNA vaccines can be rapidly manufactured and readily adapted with novel antigen sequences, genetic vaccination is a promising immunization platform. However, the applicability of different genetic adjuvants to this approach still represents a complex challenge. Immune checkpoints are a class of molecules involved in adaptive immune responses and germinal center reactions. In this study, we immunized mice by intramuscular electroporation with a DNA-vaccine encoding hemagglutinin (HA) and nucleoprotein (NP) of the influenza A virus. The DNA-vaccine was applied either alone or in combination with genetic adjuvants encoding the soluble ectodomains of programmed cell death protein-1 (sPD-1) or its ligand (sPD-L1). Co-administration of genetic checkpoint adjuvants did not significantly alter immune responses against NP. In contrast, sPD-1 co-electroporation elevated HA-specific CD4+ T cell responses, decreased regulatory CD4+ T cell pools, and modulated the IgG2a-biased HA antibody pattern towards an isotype-balanced IgG response with a trend to higher influenza neutralization in vitro. Taken together, our data demonstrate that a genetic DNA-adjuvant encoding soluble ectodomains of sPD-1 was able to modulate immune responses induced by a co-administered influenza DNA vaccine.
Collapse
|
5
|
Lapuente D, Stab V, Storcksdieck Genannt Bonsmann M, Maaske A, Köster M, Xiao H, Ehrhardt C, Tenbusch M. Innate signalling molecules as genetic adjuvants do not alter the efficacy of a DNA-based influenza A vaccine. PLoS One 2020; 15:e0231138. [PMID: 32243477 PMCID: PMC7122823 DOI: 10.1371/journal.pone.0231138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/14/2020] [Indexed: 01/07/2023] Open
Abstract
In respect to the heterogeneity among influenza A virus strains and the shortcomings of current vaccination programs, there is a huge interest in the development of alternative vaccines that provide a broader and more long-lasting protection. Gene-based approaches are considered as promising candidates for such flu vaccines. In our study, innate signalling molecules from the RIG-I and the NALP3 pathways were evaluated as genetic adjuvants in intramuscular DNA immunizations. Plasmids encoding a constitutive active form of RIG-I (cRIG-I), IPS-1, IL-1β, or IL-18 were co-administered with plasmids encoding the hemagglutinin and nucleoprotein derived from H1N1/Puerto Rico/8/1934 via electroporation in BALB/c mice. Immunogenicity was analysed in detail and efficacy was demonstrated in homologous and heterologous influenza challenge experiments. Although the biological activities of the adjuvants have been confirmed by in vitro reporter assays, their single or combined inclusion in the vaccine did not result in superior vaccine efficacy. With the exception of significantly increased levels of antigen-specific IgG1 after the co-administration of IL-1β, there were only minor alterations concerning the immunogenicity. Since DNA electroporation alone induced substantial inflammation at the injection site, as demonstrated in this study using Mx2-Luc reporter mice, it might override the adjuvants´ contribution to the inflammatory microenvironment and thereby minimizes the influence on the immunogenicity. Taken together, the DNA immunization was protective against subsequent challenge infections but could not be further improved by the genetic adjuvants analysed in this study.
Collapse
Affiliation(s)
- Dennis Lapuente
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Viktoria Stab
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | | | - Andre Maaske
- Environmental Medicine, UNIKA-T Augsburg, Technische Universität München and Helmholtz Zentrum, Neuherberg, Germany
| | - Mario Köster
- Model Systems for Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Han Xiao
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christina Ehrhardt
- Section of Experimental Virology, Institute of Medical Microbiology, University Hospital Jena, Jena, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
6
|
Tsunekuni R, Tanikawa T, Nakaya T, Saito T. Improvement of a recombinant avian avulavirus serotype 10 vectored vaccine by the addition of untranslated regions. Vaccine 2019; 38:822-829. [PMID: 31718900 DOI: 10.1016/j.vaccine.2019.10.098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND We have previously developed a recombinant avian avulavirus serotype 10 (rAAvV-10/HA) expressing the hemagglutinin (HA) gene of a highly pathogenic avian influenza virus (HPAIV) as an emergency vaccine for poultry. rAAvV-10/HA can overcome the activity of the anti-AAvV-1 (Newcastle disease virus) antibody acquired by commercial chickens upon routine vaccination. Most chickens do not have the anti-AAvV-10 antibody, which could interfere with the vaccine efficacy. However, the vaccine efficacy of rAAvV-10/HA is not satisfactory in chickens even though it affords protection against an HPAIV challenge. In the present study, we improved the rAAvV-10/HA vaccine by enhancing the expression of the exogenous HA protein. METHODS The 5' and 3' untranslated regions (UTR) of each AAvV-10 gene were flanked with the exogenous HA gene cassette to modify rAAvV-10/HA, yielding different rAAv10-UTRs. As a control, rAAv10-nonUTR that did not contain any UTRs was generated. The effects of UTRs on mRNA transcription, HA protein expression, and vaccine efficacy were then examined using embryonated chicken eggs and white leghorn chickens. RESULTS The proportion of the HA gene mRNA among the vector-derived mRNAs (1.55-1.84-fold increase vs. the control) and HA protein levels (148-1151-fold increase vs. the control) in cells infected with rAAv10-UTRs were higher than in those infected with rAAv10-nonUTR. In vivo, vaccination of chickens with rAAv10-UTRs resulted in 100% protection against an HPAIV challenge. No chickens vaccinated with rAAv10-NP-UTR, rAAv10-F-UTR, or rAAv10-HN-UTR shed the virus in the throat and cloaca swabs. By contrast, rAAv10-nonUTR vaccination offered 70% protection, with 50% of chickens shedding the virus in the cloaca or throat swabs after the challenge. We conclude that the AAvV-10 UTRs can enhance the expression of the exogenous HA gene, resulting in improved efficacy of the rAAvV-10/HA vector vaccine. This improvement aids in the protection of flocks worldwide from the highly pathogenic avian influenza.
Collapse
Affiliation(s)
- Ryota Tsunekuni
- Division of Transboundary Animal Disease, National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki 305-0854, Japan.
| | - Taichiro Tanikawa
- Division of Transboundary Animal Disease, National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki 305-0854, Japan.
| | - Takaaki Nakaya
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan.
| | - Takehiko Saito
- Division of Transboundary Animal Disease, National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki 305-0854, Japan; United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu City, Gifu 501-1193, Japan.
| |
Collapse
|
7
|
Analysis of codon usage pattern in the viral proteins of chicken anaemia virus and its possible biological relevance. INFECTION GENETICS AND EVOLUTION 2019; 69:93-106. [DOI: 10.1016/j.meegid.2019.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/19/2018] [Accepted: 01/02/2019] [Indexed: 01/05/2023]
|
8
|
Lee LYY, Izzard L, Hurt AC. A Review of DNA Vaccines Against Influenza. Front Immunol 2018; 9:1568. [PMID: 30038621 PMCID: PMC6046547 DOI: 10.3389/fimmu.2018.01568] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/25/2018] [Indexed: 01/07/2023] Open
Abstract
The challenges of effective vaccination against influenza are gaining more mainstream attention, as recent influenza seasons have reported low efficacy in annual vaccination programs worldwide. Combined with the potential emergence of novel influenza viruses resulting in a pandemic, the need for effective alternatives to egg-produced conventional vaccines has been made increasingly clear. DNA vaccines against influenza have been in development since the 1990s, but the initial excitement over success in murine model trials has been tempered by comparatively poor performance in larger animal models. In the intervening years, much progress has been made to refine the DNA vaccine platform-the rational design of antigens and expression vectors, the development of novel vaccine adjuvants, and the employment of innovative gene delivery methods. This review discusses how these advances have been applied in recent efforts to develop an effective influenza DNA vaccine.
Collapse
|
9
|
IL-1β as mucosal vaccine adjuvant: the specific induction of tissue-resident memory T cells improves the heterosubtypic immunity against influenza A viruses. Mucosal Immunol 2018; 11:1265-1278. [PMID: 29545648 DOI: 10.1038/s41385-018-0017-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/19/2018] [Accepted: 02/08/2018] [Indexed: 02/04/2023]
Abstract
A universal influenza vaccine must provide protection against antigenically divergent influenza viruses either through broadly neutralizing antibodies or cross-reactive T cells. Here, intranasal immunizations with recombinant adenoviral vectors (rAd) encoding hemagglutinin (HA) and nucleoprotein (NP) in combination with rAd-Interleukin-(IL)-1β or rAd-IL-18 were evaluated for their efficacy in BALB/c mice. Mucosal delivery of rAd-IL-1β enhanced HA-specific antibody responses including strain-specific neutralizing antibodies. Nevertheless, the beneficial effects on the local T cell responses were much more impressive reflected by increased numbers of CD103+CD69+ tissue-resident memory T cells (TRM). This increased immunogenicity translated into superior protection against infections with homologous and heterologous strains including H1N1, pH1N1, H3N2, and H7N7. Inhibition of the egress of circulating T cells out of the lymph nodes during the heterologous infection had no impact on the degree of protection underscoring the unique potential of TRM for the local containment of mucosal infections. The local co-expression of IL-1β and antigen lead to the activation of critical checkpoints in the formation of TRM including activation of epithelial cells, expression of chemokines and adhesion molecules, recruitment of lung-derived CD103+ DCs, and finally local TRM imprinting. Given the importance of TRM-mediated protection at mucosal barriers, this study has major implications for vaccine development.
Collapse
|
10
|
Evaluation of adenovirus 19a as a novel vector for mucosal vaccination against influenza A viruses. Vaccine 2018; 36:2712-2720. [PMID: 29628150 DOI: 10.1016/j.vaccine.2018.02.075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 01/19/2023]
Abstract
Since preexisting immunity and enhanced infection rates in a clinical trial of an HIV vaccine have raised some concerns on adenovirus (Ad) serotype 5-based vaccines, we evaluated the subgroup D adenovirus serotype Ad19a for its suitability as novel viral vector vaccine against mucosal infections. In BALB/c mice, we compared the immunogenicity and efficacy of E1/E3-deleted Ad19a vectors encoding the influenza A virus (IAV)-derived antigens hemagglutinin (HA) and nucleoprotein (NP) to the most commonly used Ad5 vectors. The adenoviral vectors were applied intranasally and induced detectable antigen-specific T cell responses in the lung and in the spleen as well as robust antibody responses. A prior DNA immunization significantly improved the immunogenicity of both vectors and resulted in full protection against a lethal infection with a heterologous H3N2 virus. Nevertheless, the Ad5-based vectors were slightly superior in reducing viral replication in the lung which corresponded to higher NP-specific T cell responses measured in the lungs.
Collapse
|
11
|
Cloning and Expression of a Secretory form of Truncated ORF2 (aa 112-607) from Hepatitis E Virus in the pVAX1 Vector. Jundishapur J Microbiol 2017. [DOI: 10.5812/jjm.13543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
12
|
Sarsenbayeva G, Volgin Y, Kassenov M, Issagulov T, Bogdanov N, Nurpeisova A, Sagymbay A, Abitay R, Stukova M, Sansyzbay A, Khairullin B. A novel preservative-free seasonal influenza vaccine safety and immune response study in the frame of preclinical research. J Med Virol 2017; 89:1168-1173. [PMID: 28160490 DOI: 10.1002/jmv.24771] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/08/2016] [Accepted: 12/14/2016] [Indexed: 11/08/2022]
Abstract
The paper describes the results of preclinical testing of the preparation "Vaccine allantoic split-virus inactivated against seasonal influenza." Acute toxicity and local irritating effect, anaphylactic reactions to different antigens (vaccine and ovalbumin), delayed-type hypersensitivity to ram erythrocytes, humoral immune response in hemaggtination reaction, immunogenic activity was studied in laboratory animals of various species (mice, rats, guinea pigs). Comparative analysis of the results from testing immunogenic activity of the preparation under study and the commercial influenza vaccines was performed. The preclinical testing has demonstrated safety and immune response of the seasonal split influenza vaccine, so it may be recommended for clinical study on limited contingent of volunteers.
Collapse
Affiliation(s)
| | - Yevgeniy Volgin
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Markhabat Kassenov
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Timur Issagulov
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Nikolay Bogdanov
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Ainur Nurpeisova
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Altynay Sagymbay
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Ruslan Abitay
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Marina Stukova
- Influenza Research Institute, St.Petersburg, Russian Federation
| | - Abylay Sansyzbay
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| | - Berik Khairullin
- Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan
| |
Collapse
|
13
|
Cheng BYH, Nogales A, de la Torre JC, Martínez-Sobrido L. Development of live-attenuated arenavirus vaccines based on codon deoptimization of the viral glycoprotein. Virology 2016; 501:35-46. [PMID: 27855284 DOI: 10.1016/j.virol.2016.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 11/18/2022]
Abstract
Several arenaviruses, chiefly Lassa (LASV) in West Africa, cause hemorrhagic fever (HF) disease in humans and pose important public health problems in their endemic regions. To date, there are no FDA-approved arenavirus vaccines and current anti-arenaviral therapy is limited to the use of ribavirin that has very limited efficacy. In this work we document that a recombinant prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) with a codon deoptimized (CD) surface glycoprotein (GP), rLCMV/CD, exhibited wild type (WT)-like growth properties in cultured cells despite barely detectable GP expression levels in rLCMV/CD-infected cells. Importantly, rLCMV/CD was highly attenuated in vivo but able to induce complete protection against a subsequent lethal challenge with rLCMV/WT. Our findings support the feasibility of implementing an arenavirus GP CD-based approach for the development of safe and effective live-attenuated vaccines (LAVs) to combat diseases caused by human pathogenic arenaviruses.
Collapse
Affiliation(s)
- Benson Y H Cheng
- Department of Microbiology and Immunology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Juan Carlos de la Torre
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| |
Collapse
|
14
|
Makvandi M, Teimoori A, Neisi N, Samarbafzadeh A. Designing, Construction and Expression of a Recombinant Fusion Protein Comprising the Hepatitis E Virus ORF2 and Rotavirus NSP4 in the Baculovirus Expression System. Jundishapur J Microbiol 2016; 9:e40303. [PMID: 28138375 PMCID: PMC5240165 DOI: 10.5812/jjm.40303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 12/27/2022] Open
Abstract
Background The hepatitis E virus (HEV) accounts for hepatitis E infection with relatively high mortality rate in pregnant women that can lead to fulminant hepatitis. The baculovirus expression system (BES) has the capability to produce high-level recombinant proteins and could be useful for vaccine designing. Objectives The aim of this study was designing a recombinant hepatitis E virus ORF2 and Rotavirus NSP4 (ORF2-NSP4) and to evaluating construction these recombinant proteins in the BES. Methods The truncated ORF2 gene (112-607) and truncated ORF2-NSP4 were subcloned in pFastBac1 plasmid, separately, followed by digestion and confirmed by digestion and sequencing. Then the products were transformed into Escherichia coli DH5α and retransformed in DH10Bac competent cells. Finally the white colonies containing Bacmid DNA subjected to PCR for confirming transformation. Bacmid DNA containing HEV truncated ORF2 and HEV truncated ORF2-NSP4 genes were transfected into SF9 cells using BES. The expressed proteins in the cell lysate were evaluated by SDS-PAGE and determined by the western blot assay. Results The lengths of subcloned genes, truncated ORF2 and truncated ORF2-NSP4 were 1500 and 2000bp, respectively. After retransforming in DH10Bac, the size of PCR products were 300 bp in Bacmid DNA without recombination while it was 4300 and 3800 bp in Bacmid truncated ORF2-NSP4 and Bacmid truncated ORF2 PCR products. The analysis of protein expression by SDS-PAGE and immunoblotting revealed the presence of 56 KDa for truncated ORF2 and 74.5 KDa for truncated ORF2-NSP4 proteins. Conclusions The results of the present study showed that the baculovirus expression system (SF9 cells) was able to express truncated ORF2 and truncated ORF2-NSP4 proteins as a potential candidate vaccine.
Collapse
Affiliation(s)
- Manoochehr Makvandi
- Infectious and Tropical Disease Research Center, Health Research Institute, Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Ali Teimoori
- Infectious and Tropical Disease Research Center, Health Research Institute, Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
- Research center for Infectious Diseases of Digestive System; Imam Khomeini hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Niloofar Neisi
- Infectious and Tropical Disease Research Center, Health Research Institute, Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
- Corresponding author: Niloofar Neisi, Infectious and Tropical Disease Research Center, Health Research Institute, Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran. Tel: +98-9166136984, Fax: +98-6133332036; +98-6133362411, E-mail:
| | - Alireza Samarbafzadeh
- Infectious and Tropical Disease Research Center, Health Research Institute, Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| |
Collapse
|
15
|
Stachyra A, Redkiewicz P, Kosson P, Protasiuk A, Góra-Sochacka A, Kudla G, Sirko A. Codon optimization of antigen coding sequences improves the immune potential of DNA vaccines against avian influenza virus H5N1 in mice and chickens. Virol J 2016; 13:143. [PMID: 27562235 PMCID: PMC5000471 DOI: 10.1186/s12985-016-0599-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/12/2016] [Indexed: 01/12/2023] Open
Abstract
Background Highly pathogenic avian influenza viruses are a serious threat to domestic poultry and can be a source of new human pandemic and annual influenza strains. Vaccination is the main strategy of protection against influenza, thus new generation vaccines, including DNA vaccines, are needed. One promising approach for enhancing the immunogenicity of a DNA vaccine is to maximize its expression in the immunized host. Methods The immunogenicity of three variants of a DNA vaccine encoding hemagglutinin (HA) from the avian influenza virus A/swan/Poland/305-135V08/2006 (H5N1) was compared in two animal models, mice (BALB/c) and chickens (broilers and layers). One variant encoded the wild type HA while the other two encoded HA without proteolytic site between HA1 and HA2 subunits and differed in usage of synonymous codons. One of them was enriched for codons preferentially used in chicken genes, while in the other modified variant the third position of codons was occupied in almost 100 % by G or C nucleotides. Results The variant of the DNA vaccine containing almost 100 % of the GC content in the third position of codons stimulated strongest immune response in two animal models, mice and chickens. These results indicate that such modification can improve not only gene expression but also immunogenicity of DNA vaccine. Conclusion Enhancement of the GC content in the third position of the codon might be a good strategy for development of a variant of a DNA vaccine against influenza that could be highly effective in distant hosts, such as birds and mammals, including humans. Electronic supplementary material The online version of this article (doi:10.1186/s12985-016-0599-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Stachyra
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul., Pawinskiego 5A, 02-106, Warsaw, Poland
| | - Patrycja Redkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul., Pawinskiego 5A, 02-106, Warsaw, Poland
| | - Piotr Kosson
- Mossakowski Medical Research Centre Polish Academy of Sciences, ul., Pawinskiego 5, 02-106, Warsaw, Poland
| | - Anna Protasiuk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul., Pawinskiego 5A, 02-106, Warsaw, Poland
| | - Anna Góra-Sochacka
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul., Pawinskiego 5A, 02-106, Warsaw, Poland
| | - Grzegorz Kudla
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, Scotland, UK
| | - Agnieszka Sirko
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul., Pawinskiego 5A, 02-106, Warsaw, Poland.
| |
Collapse
|
16
|
Martinez-Sobrido L, de la Torre JC. Novel strategies for development of hemorrhagic fever arenavirus live-attenuated vaccines. Expert Rev Vaccines 2016; 15:1113-21. [PMID: 27118328 DOI: 10.1080/14760584.2016.1182024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Several arenaviruses, chiefly Lassa virus (LASV), cause hemorrhagic fever (HF) disease in humans and pose significant public health problems in their endemic regions. Moreover, HF arenaviruses represent credible biodefense threats. There are not FDA-approved arenavirus vaccines and current anti-arenaviral therapy is limited to an off-label use of ribavirin that is only partially effective. AREAS COVERED Live-attenuated vaccines (LAV) represent the most feasible approach to control HF arenaviruses within their endemic regions. Different platforms, including recombinant viral vectors expressing LASV antigens, and the use of attenuated reassortant arenaviruses, have been used to develop LAV candidates against LASV with promising results in animal models of LASV infection, but none of them has entered a clinical trial. These vaccine efforts have been the subject of recent reviews and will not be examined in this review, which is focused on new avenues for the development of safe and effective LAV to combat HF arenaviruses. Expert commentary: The development of arenavirus reverse genetics has provided investigators with a novel powerful approach to manipulate the genomes of HF arenaviruses, which has opened new avenues for the rapid development of safe and effective LAV to combat these human pathogens.
Collapse
Affiliation(s)
- Luis Martinez-Sobrido
- a Department of Microbiology and Immunology , University of Rochester Medical Center Ringgold standard institution , Rochester , NY , USA
| | - Juan Carlos de la Torre
- b Department of Immunology and Microbial Science , The Scripps Research Institute , La Jolla , CA , USA
| |
Collapse
|
17
|
Starodubova ES, Kuzmenko YV, Latanova AA, Preobrazhenskaya OV, Karpov VL. Creation of DNA vaccine vector based on codon-optimized gene of rabies virus glycoprotein (G protein) with consensus amino acid sequence. Mol Biol 2016. [DOI: 10.1134/s0026893316020242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Exploiting tRNAs to Boost Virulence. Life (Basel) 2016; 6:life6010004. [PMID: 26797637 PMCID: PMC4810235 DOI: 10.3390/life6010004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/08/2016] [Accepted: 01/14/2016] [Indexed: 01/22/2023] Open
Abstract
Transfer RNAs (tRNAs) are powerful small RNA entities that are used to translate nucleotide language of genes into the amino acid language of proteins. Their near-uniform length and tertiary structure as well as their high nucleotide similarity and post-transcriptional modifications have made it difficult to characterize individual species quantitatively. However, due to the central role of the tRNA pool in protein biosynthesis as well as newly emerging roles played by tRNAs, their quantitative assessment yields important information, particularly relevant for virus research. Viruses which depend on the host protein expression machinery have evolved various strategies to optimize tRNA usage—either by adapting to the host codon usage or encoding their own tRNAs. Additionally, several viruses bear tRNA-like elements (TLE) in the 5′- and 3′-UTR of their mRNAs. There are different hypotheses concerning the manner in which such structures boost viral protein expression. Furthermore, retroviruses use special tRNAs for packaging and initiating reverse transcription of their genetic material. Since there is a strong specificity of different viruses towards certain tRNAs, different strategies for recruitment are employed. Interestingly, modifications on tRNAs strongly impact their functionality in viruses. Here, we review those intersection points between virus and tRNA research and describe methods for assessing the tRNA pool in terms of concentration, aminoacylation and modification.
Collapse
|
19
|
Niezold T, Storcksdieck Genannt Bonsmann M, Maaske A, Temchura V, Heinecke V, Hannaman D, Buer J, Ehrhardt C, Hansen W, Überla K, Tenbusch M. DNA vaccines encoding DEC205-targeted antigens: immunity or tolerance? Immunology 2015; 145:519-33. [PMID: 25819746 DOI: 10.1111/imm.12467] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 01/02/2023] Open
Abstract
Targeting of antigens to the endocytic uptake receptor DEC205 resulted in enhanced antigen presentation by dendritic cells (DCs). In combination with adjuvants for DC maturation, proteins coupled to an antibody against DEC205 induced strong pathogen-specific immune responses, whereas without additional adjuvant tolerance could be induced. As less is known about DNA vaccines encoding DEC205-targeted antigens, we explored the immunogenicity and efficacy of a dendritic cell-targeted DNA vaccine against influenza A virus (IAV) delivered by electroporation. Although coupling of haemagglutinin to a single-chain antibody against DEC205 enhanced antigen presentation on MHC class II and activation of T-cell receptor-transgenic CD4 T cells, the T-cell responses induced by the targeted DNA vaccine in wild-type BALB/c mice were significantly reduced compared with DNA encoding non-targeted antigens. Consistently, these mice were less protected against an IAV infection. Adoptive transfer experiments were performed to assess the fate of the antigen-specific T cells in animals vaccinated with DNA encoding DEC205-targeted antigens. By this, we could exclude the general deletion of antigen-specific T cells as cause for the reduced efficacy, but observed a local expansion of antigen-specific regulatory T cells, which could suppress the activation of effector cells. In conclusion, DNA vaccines encoding DEC205-targeted antigens induce peripheral tolerance rather than immunity in our study. Finally, we evaluated our DNA vaccines as prophylactic or therapeutic treatment in an allergen-induced asthma mouse model.
Collapse
Affiliation(s)
- Thomas Niezold
- Department of Molecular and Medical Virology, Ruhr-University, Bochum, Germany
| | | | - André Maaske
- Department of Molecular and Medical Virology, Ruhr-University, Bochum, Germany
| | - Vladimir Temchura
- Department of Molecular and Medical Virology, Ruhr-University, Bochum, Germany
| | - Vanessa Heinecke
- Department of Molecular and Medical Virology, Ruhr-University, Bochum, Germany
| | | | - Jan Buer
- Institute of Medical Microbiology University Hospital, Essen
| | - Christina Ehrhardt
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation, University of Münster, Münster
| | - Wiebke Hansen
- Institute of Medical Microbiology University Hospital, Essen
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-University, Bochum, Germany.,Universitätsklinikum Erlangen, Institute of Clinical and Medical Virology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University, Bochum, Germany
| |
Collapse
|
20
|
Klingbeil K, Lange E, Teifke JP, Mettenleiter TC, Fuchs W. Immunization of pigs with an attenuated pseudorabies virus recombinant expressing the haemagglutinin of pandemic swine origin H1N1 influenza A virus. J Gen Virol 2014; 95:948-959. [PMID: 24431235 DOI: 10.1099/vir.0.059253-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pigs can be severely harmed by influenza, and represent important reservoir hosts, in which new human pathogens such as the recent pandemic swine-origin H1N1 influenza A virus can arise by mutation and reassortment of genome segments. To obtain novel, safe influenza vaccines for pigs, and to investigate the antigen-specific immune response, we modified an established live-virus vaccine against Aujeszky's disease of swine, pseudorabies virus (PrV) strain Bartha (PrV-Ba), to serve as vector for the expression of haemagglutinin (HA) of swine-origin H1N1 virus. To facilitate transgene insertion, the genome of PrV-Ba was cloned as a bacterial artificial chromosome. HA expression occurred under control of the human or murine cytomegalovirus immediate early promoters (P-HCMV, P-MCMV), but could be substantially enhanced by synthetic introns and adaptation of the codon usage to that of PrV. However, despite abundant expression, the heterologous glycoprotein was not detectably incorporated into mature PrV particles. Replication of HA-expressing PrV in cell culture was only slightly affected compared to that of the parental virus strain. A single immunization of pigs with the PrV vector expressing the codon-optimized HA gene under control of P-MCMV induced high levels of HA-specific antibodies. The vaccinated animals were protected from clinical signs after challenge with a related swine-origin H1N1 influenza A virus, and challenge virus shedding was significantly reduced.
Collapse
Affiliation(s)
- Katharina Klingbeil
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Elke Lange
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Jens P Teifke
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Walter Fuchs
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| |
Collapse
|
21
|
Flingai S, Czerwonko M, Goodman J, Kudchodkar SB, Muthumani K, Weiner DB. Synthetic DNA vaccines: improved vaccine potency by electroporation and co-delivered genetic adjuvants. Front Immunol 2013; 4:354. [PMID: 24204366 PMCID: PMC3816528 DOI: 10.3389/fimmu.2013.00354] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/20/2013] [Indexed: 01/07/2023] Open
Abstract
In recent years, DNA vaccines have undergone a number of technological advancements that have incited renewed interest and heightened promise in the field. Two such improvements are the use of genetically engineered cytokine adjuvants and plasmid delivery via in vivo electroporation (EP), the latter of which has been shown to increase antigen delivery by nearly 1000-fold compared to naked DNA plasmid delivery alone. Both strategies, either separately or in combination, have been shown to augment cellular and humoral immune responses in not only mice, but also in large animal models. These promising results, coupled with recent clinical trials that have shown enhanced immune responses in humans, highlight the bright prospects for DNA vaccines to address many human diseases.
Collapse
Affiliation(s)
- Seleeke Flingai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania , Philadelphia, PA , USA
| | | | | | | | | | | |
Collapse
|
22
|
Stab V, Nitsche S, Niezold T, Storcksdieck genannt Bonsmann M, Wiechers A, Tippler B, Hannaman D, Ehrhardt C, Überla K, Grunwald T, Tenbusch M. Protective efficacy and immunogenicity of a combinatory DNA vaccine against Influenza A Virus and the Respiratory Syncytial Virus. PLoS One 2013; 8:e72217. [PMID: 23967287 PMCID: PMC3743785 DOI: 10.1371/journal.pone.0072217] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 07/05/2013] [Indexed: 12/18/2022] Open
Abstract
The Respiratory Syncytial Virus (RSV) and Influenza A Virus (IAV) are both two major causative agents of severe respiratory tract infections in humans leading to hospitalization and thousands of deaths each year. In this study, we evaluated the immunogenicity and efficacy of a combinatory DNA vaccine in comparison to the single component vaccines against both diseases in a mouse model. Intramuscular electroporation with plasmids expressing the hemagglutinin (HA) of IAV and the F protein of RSV induced strong humoral immune responses regardless if they were delivered in combination or alone. In consequence, high neutralizing antibody titers were detected, which conferred protection against a lethal challenge with IAV. Furthermore, the viral load in the lungs after a RSV infection could be dramatically reduced in vaccinated mice. Concurrently, substantial amounts of antigen-specific, polyfunctional CD8⁺ T-cells were measured after vaccination. Interestingly, the cellular response to the hemagglutinin was significantly reduced in the presence of the RSV-F encoding plasmid, but not vice versa. Although these results indicate a suppressive effect of the RSV-F protein, the protective efficacy of the combinatory vaccine was comparable to the efficacy of both single-component vaccines. In conclusion, the novel combinatory vaccine against RSV and IAV may have great potential to reduce the rate of severe respiratory tract infections in humans without increasing the number of necessary vaccinations.
Collapse
Affiliation(s)
- Viktoria Stab
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Sandra Nitsche
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Niezold
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | | | - Andrea Wiechers
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Bettina Tippler
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Drew Hannaman
- Ichor Medical Systems, San Diego, California, United States of America
| | - Christina Ehrhardt
- Institute of Molecular Virology, Centre of Molecular Biology of Inflammation, Westfaelische Wilhelms University, Muenster, Germany
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Grunwald
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
- * E-mail:
| |
Collapse
|
23
|
Tello M, Vergara F, Spencer E. Genomic adaptation of the ISA virus to Salmo salar codon usage. Virol J 2013; 10:223. [PMID: 23829271 PMCID: PMC3706250 DOI: 10.1186/1743-422x-10-223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 07/01/2013] [Indexed: 01/09/2023] Open
Abstract
Background The ISA virus (ISAV) is an Orthomyxovirus whose genome encodes for at least 10 proteins. Low protein identity and lack of genetic tools have hampered the study of the molecular mechanism behind its virulence. It has been shown that viral codon usage controls several processes such as translational efficiency, folding, tuning of protein expression, antigenicity and virulence. Despite this, the possible role that adaptation to host codon usage plays in virulence and viral evolution has not been studied in ISAV. Methods Intergenomic adaptation between viral and host genomes was calculated using the codon adaptation index score with EMBOSS software and the Kazusa database. Classification of host genes according to GeneOnthology was performed using Blast2go. A non parametric test was applied to determine the presence of significant correlations among CAI, mortality and time. Results Using the codon adaptation index (CAI) score, we found that the encoding genes for nucleoprotein, matrix protein M1 and antagonist of Interferon I signaling (NS1) are the ISAV genes that are more adapted to host codon usage, in agreement with their requirement for production of viral particles and inactivation of antiviral responses. Comparison to host genes showed that ISAV shares CAI values with less than 0.45% of Salmo salar genes. GeneOntology classification of host genes showed that ISAV genes share CAI values with genes from less than 3% of the host biological process, far from the 14% shown by Influenza A viruses and closer to the 5% shown by Influenza B and C. As well, we identified a positive correlation (p<0.05) between CAI values of a virus and the duration of the outbreak disease in given salmon farms, as well as a weak relationship between codon adaptation values of PB1 and the mortality rates of a set of ISA viruses. Conclusions Our analysis shows that ISAV is the least adapted viral Salmo salar pathogen and Orthomyxovirus family member less adapted to host codon usage, avoiding the general behavior of host genes. This is probably due to its recent emergence among farmed Salmon populations.
Collapse
Affiliation(s)
- Mario Tello
- Centro de Biotecnología Acuícola, Laboratorio de Virología, Facultad de Química y Biología, Universidad de Santiago de Chile, Avenida Libertador Bernardo O'Higgins 3363, Santiago, Chile.
| | | | | |
Collapse
|
24
|
Pavon-Eternod M, David A, Dittmar K, Berglund P, Pan T, Bennink JR, Yewdell JW. Vaccinia and influenza A viruses select rather than adjust tRNAs to optimize translation. Nucleic Acids Res 2012; 41:1914-21. [PMID: 23254333 PMCID: PMC3561966 DOI: 10.1093/nar/gks986] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Transfer RNAs (tRNAs) are central to protein synthesis and impact translational speed and
fidelity by their abundance. Here we examine the extent to which viruses manipulate tRNA
populations to favor translation of their own genes. We study two very different viruses:
influenza A virus (IAV), a medium-sized (13 kB genome) RNA virus; and vaccinia virus (VV),
a large (200 kB genome) DNA virus. We show that the total cellular tRNA population remains
unchanged following viral infection, whereas the polysome-associated tRNA population
changes dramatically in a virus-specific manner. The changes in polysome-associated tRNA
levels reflect the codon usage of viral genes, suggesting the existence of local tRNA
pools optimized for viral translation.
Collapse
Affiliation(s)
- Mariana Pavon-Eternod
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Li L, Saade F, Petrovsky N. The future of human DNA vaccines. J Biotechnol 2012; 162:171-82. [PMID: 22981627 DOI: 10.1016/j.jbiotec.2012.08.012] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/01/2012] [Accepted: 08/06/2012] [Indexed: 01/03/2023]
Abstract
DNA vaccines have evolved greatly over the last 20 years since their invention, but have yet to become a competitive alternative to conventional protein or carbohydrate based human vaccines. Whilst safety concerns were an initial barrier, the Achilles heel of DNA vaccines remains their poor immunogenicity when compared to protein vaccines. A wide variety of strategies have been developed to optimize DNA vaccine immunogenicity, including codon optimization, genetic adjuvants, electroporation and sophisticated prime-boost regimens, with each of these methods having its advantages and limitations. Whilst each of these methods has contributed to incremental improvements in DNA vaccine efficacy, more is still needed if human DNA vaccines are to succeed commercially. This review foresees a final breakthrough in human DNA vaccines will come from application of the latest cutting-edge technologies, including "epigenetics" and "omics" approaches, alongside traditional techniques to improve immunogenicity such as adjuvants and electroporation, thereby overcoming the current limitations of DNA vaccines in humans.
Collapse
Affiliation(s)
- Lei Li
- Vaxine Pty Ltd, Bedford Park, Adelaide 5042, Australia
| | | | | |
Collapse
|
26
|
Kamdem Toukam D, Tenbusch M, Stang A, Temchura V, Storcksdieck genannt Bonsmann M, Grewe B, Koch S, Meyerhans A, Nchinda G, Kaptue L, Überla K. Targeting antibody responses to the membrane proximal external region of the envelope glycoprotein of human immunodeficiency virus. PLoS One 2012; 7:e38068. [PMID: 22666448 PMCID: PMC3364164 DOI: 10.1371/journal.pone.0038068] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 04/30/2012] [Indexed: 01/20/2023] Open
Abstract
Although human immunodeficiency type 1 (HIV-1) infection induces strong antibody responses to the viral envelope glycoprotein (Env) only a few of these antibodies possess the capacity to neutralize a broad range of strains. The induction of such antibodies represents an important goal in the development of a preventive vaccine against the infection. Among the broadly neutralizing monoclonal antibodies discovered so far, three (2F5, Z13 and 4E10) target the short and hidden membrane proximal external region (MPER) of the gp41 transmembrane protein. Antibody responses to MPER are rarely observed in HIV-infected individuals or after immunization with Env immunogens. To initiate antibody responses to MPER in its membrane-embedded native conformation, we generated expression plasmids encoding the membrane-anchored ectodomain of gp41 with N-terminal deletions of various sizes. Following transfection of these plasmids, the MPER domains are displayed on the cell surface and incorporated into HIV virus like particles (VLP). Transfected cells displaying MPER mutants bound as efficiently to both 2F5 and 4E10 as cells transfected with a plasmid encoding full-length Env. Mice immunized with VLPs containing the MPER mutants produced MPER-specific antibodies, the levels of which could be increased by the trimerization of the displayed proteins as well as by a DNA prime-VLP boost immunization strategy. Although 2F5 competed for binding to MPER with antibodies in sera of some of the immunized mice, neutralizing activity could not be detected. Whether this is due to inefficient binding of the induced antibodies to MPER in the context of wild type Env or whether the overall MPER-specific antibody response induced by the MPER display mutants is too low to reveal neutralizing activity, remains to be determined.
Collapse
Affiliation(s)
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Stang
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Vladimir Temchura
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | | | - Bastian Grewe
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Stefanie Koch
- Fraunhofer Institute for Biomedical Engineering, Sulzbach, Germany
| | - Andreas Meyerhans
- ICREA Infection Biology Laboratory, Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
| | - Godwin Nchinda
- Laboratory of Immunology, The Chantal Biya International Reference Centre for Research on HIV/AIDS Prevention and Management (CIRCB), Yaounde, Cameroon
| | - Lazare Kaptue
- Institut Supérieur des Sciences de la Santé, Université des Montagnes, Banganté, Cameroon
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
- * E-mail:
| |
Collapse
|