1
|
Chai S, Wang L, Du H, Jiang H. Achievement and Challenges in Orthohantavirus Vaccines. Vaccines (Basel) 2025; 13:198. [PMID: 40006744 PMCID: PMC11861054 DOI: 10.3390/vaccines13020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Orthohantaviruses (also known as hantaviruses) are pathogens that cause two distinct, yet related forms of severe human disease: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). These diseases pose a significant threat to global public health due to their high case fatality rates, which can range from 1% to 50%. In recent years, an increasing number of countries and regions have reported human cases, underscoring the urgent need for improved understanding, prevention, and treatment strategies. Given the severity of these diseases and the lack of specific post-exposure antiviral treatments, preventive measures are critical. For several decades, substantial efforts have been dedicated to developing orthohantavirus vaccines, leading to significant advancements. The first large-scale deployment involved inactivated vaccines, which played a crucial role in reducing HFRS incidence in South Korea and China. Subunit vaccines, viral vector vaccines, and virus-like particle (VLP) vaccines have also been extensively researched. Nucleic acid vaccines, including both mRNA and DNA vaccines, hold the greatest potential for future development due to their rapid design and production cycles, ability to elicit robust immune responses, ease of storage and transportation, and adaptable production platforms. Ongoing advancements in computer technology and artificial intelligence promise to further enhance the development of more effective orthohantavirus vaccines.
Collapse
Affiliation(s)
- Shiqi Chai
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| | - Limei Wang
- Department of Microbiology and Pathogenic Biology, School of Basic Medicine, The Fourth Military Medical University, Xi’an 710032, China;
| | - Hong Du
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| | - Hong Jiang
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| |
Collapse
|
2
|
Garcia-Atutxa I, Mondragon-Teran P, Huerta-Saquero A, Villanueva-Flores F. Advancements in monkeypox vaccines development: a critical review of emerging technologies. Front Immunol 2024; 15:1456060. [PMID: 39464881 PMCID: PMC11502315 DOI: 10.3389/fimmu.2024.1456060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024] Open
Abstract
Monkeypox (mpox) is a zoonotic illness caused by the monkeypox virus (MPXV), with higher health concerns among people who are pregnant, children, and persons who are immunocompromised, including people with untreated and advanced HIV disease. Significant progress has been made in developing vaccines against mpox, yet critical challenges and limitations persist in ensuring their effectiveness, safety, and accessibility. The pertinence of this review is highlighted by the World Health Organization's declaration of a global health emergency on August 14, 2024, due to the recent mpox outbreak, underscoring the critical necessity for effective vaccine solutions in the face of a rapidly evolving virus. Here, we comprehensively analyze various vaccine platforms utilized in mpox prevention, including attenuated and non-replicating virus vaccines, viral vector-based vaccines, recombinant protein vaccines, and DNA and mRNA vaccines. We evaluate the advantages and limitations of each platform, highlighting the urgent need for ongoing research and innovation to enhance vaccine efficacy and safety. Recent advancements, such as incorporating immunostimulatory sequences, improved delivery systems, and developing polyvalent vaccines, are explored for their potential to offer broader protection against diverse orthopoxvirus strains. This work underscores the need to optimize currently available vaccines and investigate novel vaccination strategies to address future public health emergencies effectively. By focusing on these advanced methodologies, we aim to contribute to the development of robust and adaptable vaccine solutions for mpox and other related viral threats.
Collapse
Affiliation(s)
- Igor Garcia-Atutxa
- Computer Science Department, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Paul Mondragon-Teran
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| | - Alejandro Huerta-Saquero
- Departamento de Bionanotecnología, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México (UNAM), Ensenada, Mexico
| | - Francisca Villanueva-Flores
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Xochitepec, Morelos, Mexico
| |
Collapse
|
3
|
Kuang J, Liu M, Yu Q, Cheng Y, Huang J, Han S, Shi J, Huang L, Li P. Antiviral Effect and Mechanism of Edaravone against Grouper Iridovirus Infection. Viruses 2023; 15:2237. [PMID: 38005914 PMCID: PMC10674758 DOI: 10.3390/v15112237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Singapore grouper iridovirus (SGIV) is a virus with high fatality rate in the grouper culture industry. The outbreak of SGIV is often accompanied by a large number of grouper deaths, which has a great impact on the economy. Therefore, it is of great significance to find effective drugs against SGIV. It has been reported that edaravone is a broad-spectrum antiviral drug, most widely used clinically in recent years, but no report has been found exploring the effect of edaravone on SGIV infections. In this study, we evaluated the antiviral effect of edaravone against SGIV, and the anti-SGIV mechanism of edaravone was also explored. It was found that the safe concentration of edaravone on grouper spleen (GS) cells was 50 µg/mL, and it possessed antiviral activity against SGIV infection in a dose-dependent manner. Furthermore, edaravone could significantly disrupt SGIV particles and interference with SGIV binding to host cells, as well as SGIV replication in host cells. However, edaravone was not effective during the SGIV invasion into host cells. This study was the first time that it was determined that edaravone could exert antiviral effects in response to SGIV infection by directly interfering with the processes of SGIV infecting cells, aiming to provide a theoretical basis for the control of grouper virus disease.
Collapse
Affiliation(s)
- Jihui Kuang
- School of Resources, Environment and Materials, Guangxi University, Nanning 537100, China;
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
| | - Mingzhu Liu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Qing Yu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Yuan Cheng
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Jing Huang
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Shuyu Han
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- Guangxi Fisheries Technology Extension Station, Nanning 530022, China
| | - Jingu Shi
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- Guangxi Fisheries Technology Extension Station, Nanning 530022, China
| | - Lin Huang
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Pengfei Li
- School of Resources, Environment and Materials, Guangxi University, Nanning 537100, China;
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| |
Collapse
|
4
|
Fang CY, Liu CC. Novel strategies for the development of hand, foot, and mouth disease vaccines and antiviral therapies. Expert Opin Drug Discov 2022; 17:27-39. [PMID: 34382876 DOI: 10.1080/17460441.2021.1965987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/05/2021] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Hand, foot, and mouth disease (HFMD) poses a great threat to young children in the Asia-Pacific region. HFMD is usually caused by enterovirus A, and infection with enterovirus A71 (EV-A71) is particularly associated with severe complications. However, coxsackievirus CV-A16, CV-A6, and CV-A10 pandemics have been observed in recent HFMD outbreaks. Inactivated monovalent EV-A71 vaccines are available to prevent EV-A71 infection; however, they cannot prevent infections by non-EV-A71 enteroviruses. Anti-enteroviral drugs are still in the developmental stage. Application of novel strategies will facilitate the development of new therapies against these emerging HFMD-associated enteroviruses. AREAS COVERED The authors highlight the current approaches for anti-enterovirus therapeutic development and discuss the application of these novel strategies for the discovery of vaccines and antiviral drugs for enteroviruses. EXPERT OPINION The maturation of DNA/RNA vaccine technology could be applied for rapid and robust development of multivalent enterovirus vaccines. Structure biology and neutralization antibody studies decipher the immunodominant sites of enteroviruses for vaccine design. Nucleotide aptamer library screening is a novel, fast, and cost-effective strategy for the development of antiviral agents. Animal models carrying viral receptors and attachment factors are required for enterovirus study and vaccine/antiviral development. Currently developed antivirals require effectiveness evaluation in clinical trials.
Collapse
Affiliation(s)
- Chih-Yeu Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Chia-Chyi Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| |
Collapse
|
5
|
Rockman S, Laurie KL, Parkes S, Wheatley A, Barr IG. New Technologies for Influenza Vaccines. Microorganisms 2020; 8:microorganisms8111745. [PMID: 33172191 PMCID: PMC7694987 DOI: 10.3390/microorganisms8111745] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/04/2020] [Accepted: 11/04/2020] [Indexed: 12/22/2022] Open
Abstract
Vaccine development has been hampered by the long lead times and the high cost required to reach the market. The 2020 pandemic, caused by a new coronavirus (SARS-CoV-2) that was first reported in late 2019, has seen unprecedented rapid activity to generate a vaccine, which belies the traditional vaccine development cycle. Critically, much of this progress has been leveraged off existing technologies, many of which had their beginnings in influenza vaccine development. This commentary outlines the most promising of the next generation of non-egg-based influenza vaccines including new manufacturing platforms, structure-based antigen design/computational biology, protein-based vaccines including recombinant technologies, nanoparticles, gene- and vector-based technologies, as well as an update on activities around a universal influenza vaccine.
Collapse
Affiliation(s)
- Steven Rockman
- Technical Development, Seqirus Ltd, Parkville, Victoria 3052, Australia; (S.R.); (S.P.)
- Department of Immunology and Microbiology, The University of Melbourne, Parkville, Victoria 3052, Australia; (A.W.); (I.G.B.)
| | - Karen L. Laurie
- Technical Development, Seqirus Ltd, Parkville, Victoria 3052, Australia; (S.R.); (S.P.)
- Correspondence:
| | - Simone Parkes
- Technical Development, Seqirus Ltd, Parkville, Victoria 3052, Australia; (S.R.); (S.P.)
| | - Adam Wheatley
- Department of Immunology and Microbiology, The University of Melbourne, Parkville, Victoria 3052, Australia; (A.W.); (I.G.B.)
| | - Ian G. Barr
- Department of Immunology and Microbiology, The University of Melbourne, Parkville, Victoria 3052, Australia; (A.W.); (I.G.B.)
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria 3052, Australia
| |
Collapse
|
6
|
Yu Q, Liu M, Xiao H, Wu S, Qin X, Lu Z, Shi D, Li S, Mi H, Wang Y, Su H, Wang T, Li P. The inhibitory activities and antiviral mechanism of Viola philippica aqueous extracts against grouper iridovirus infection in vitro and in vivo. JOURNAL OF FISH DISEASES 2019; 42:859-868. [PMID: 30893481 DOI: 10.1111/jfd.12987] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 06/09/2023]
Abstract
Grouper iridovirus (GIV) is one of the most serious pathogens in mariculture and causes high mortality rates in cultured groupers; then, effective medicines for controlling GIV infections are urgently needed. Viola philippica is a well-known medicinal plant, and the application of V. philippica aqueous extracts against GIV infection was assessed by different methods in this study. The results showed that the working concentration of V. philippica aqueous extracts was 10 mg/ml. V. philippica aqueous extracts below 10 mg/ml have no significant cytotoxic effects on cell viability, while extracts over 15 mg/ml decreased cell viability and showed cytotoxic activity. V. philippica aqueous extracts had excellent inhibitory effects against GIV infection in vitro and in vivo. The possible antiviral mechanism of V. philippica was further analysed, which indicated that V. philippica did no damages to GIV particles, but it could disturb GIV binding, entry and replication in host cells. V. philippica had the best inhibitory effects against GIV during viral infection stage of binding and replication in host cells. Overall, the results suggest that appropriate concentration of V. philippica aqueous extracts has great antiviral effects, making it an interesting candidate for developing effective medicines for preventing and controlling GIV infection in farmed groupers.
Collapse
Affiliation(s)
- Qing Yu
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Sciences, Nanning, China
| | - Mingzhu Liu
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, Nanning, China
| | - Hehe Xiao
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Siting Wu
- Guangxi Key Lab for Marine Biotechnology, Guangxi Institute of Oceanography, Guangxi Academy of Sciences, Beihai, China
| | - Xianling Qin
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Sciences, Nanning, China
| | - Zijun Lu
- School of Marine Sciences and Biotechnology, Guangxi University for Nationalities, Nanning, China
| | - Deqiang Shi
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Sciences, Nanning, China
| | - Siqiao Li
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, Nanning, China
| | - Huizhi Mi
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, Nanning, China
| | - Yibing Wang
- School of Marine Sciences and Biotechnology, Guangxi University for Nationalities, Nanning, China
| | - Hongfei Su
- Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, College of Marine Sciences, Guangxi University, Nanning, China
| | - Taixia Wang
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Pengfei Li
- Guangxi Key Laboratory of Marine Environmental Science, Guangxi Academy of Sciences, Nanning, China
| |
Collapse
|
7
|
Immunogenicity of a bovine herpesvirus 1 glycoprotein D DNA vaccine complexed with bovine neutrophil beta-defensin 3. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 22:79-90. [PMID: 25378352 DOI: 10.1128/cvi.00476-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Protective efficacy against bovine herpesvirus 1 (BoHV-1) has been demonstrated to be induced by a plasmid encoding bovine neutrophil beta-defensin 3 (BNBD3) as a fusion construct with truncated glycoprotein D (tgD). However, in spite of the increased cell-mediated immune responses induced by this DNA vaccine, the clinical responses of BoHV-1-challenged cattle were not reduced over those observed in animals vaccinated with the plasmid encoding tgD alone; this might have been because the vaccine failed to improve humoral responses. We hypothesized that an alternative vaccine design strategy that utilized the DNA vaccine pMASIA-tgD as a complex with BNBD3 might improve humoral responses while maintaining robust Th1-type cell-mediated responses. C57BL/6 mice were vaccinated with pMASIA-tgD complexed with 0, 0.01875, 0.1875, or 1.875 nmol of a stable synthesized analog of BNBD3 (aBNBD3). The best results were seen in mice immunized with the vaccine composed of pMASIA-tgD complexed to 0.1875 nmol aBNBD3. In this group, humoral responses were improved, as evidenced by increased virus neutralization, tgD-specific early IgG1, and later IgG2a titers, while the strong cell-mediated immune responses, measured based on specific gamma interferon (IFN-γ)-secreting cells, were maintained relative to pMASIA-tgD. Modulation of the immune response might have been due in part to the effect of BNBD3 on dendritic cells (DCs). In vitro studies showed that murine bone marrow-derived DCs (BMDCs) pretreated with aBNBD3 were activated, as evidenced by CD11c downregulation, and were functionally mature, as shown by increased allostimulatory ability. Native, synthetic, and analog forms of BNBD3 were equally capable of inducing functional maturation of BMDCs.
Collapse
|
8
|
Li P, Yan Y, Wei S, Wei J, Gao R, Huang X, Huang Y, Jiang G, Qin Q. Isolation and characterization of a new class of DNA aptamers specific binding to Singapore grouper iridovirus (SGIV) with antiviral activities. Virus Res 2014; 188:146-54. [PMID: 24768847 DOI: 10.1016/j.virusres.2014.04.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/11/2014] [Accepted: 04/11/2014] [Indexed: 11/18/2022]
Abstract
The Singapore grouper iridovirus (SGIV), a member of the genus Ranavirus, is a major viral pathogen that has caused heavy economic losses to the grouper aquaculture industry in China and Southeast Asia. No efficient method of controlling SGIV outbreaks is currently available. Systematic evolution of ligands by exponential enrichment (SELEX) is now widely used for the in vitro selection of artificial ssDNA or RNA ligands, known as aptamers, which bind to targets through their stable three-dimensional structures. In our current study, we generated ssDNA aptamers against the SGIV, and evaluated their ability to block SGIV infection in cultured fish cells and cultured fish in vivo. The anti-SGIV DNA aptamers, LMB-761, LMB-764, LMB-748, LMB-439, LMB-755, and LMB-767, were selected from a pool of oligonucleotides randomly generated using a SELEX iterative method. The analysis of the secondary structure of the aptamers revealed that they all formed similar stem-loop structures. Electrophoretic mobility shift assays showed that the aptamers bound SGIV specifically, as evidenced by a lack cross-reactivity with the soft shell turtle iridovirus. The aptamers produced no cytotoxic effects in cultured grouper spleen cells (GS). Assessment of cytopathic effects (CPE) and viral titer assays showed that LMB-761, LMB-764, LMB-748, LMB-755, and LMB-767 significantly inhibited SGIV infection in GS cells. The in vivo experiments showed that LMB-761 and LMB-764 reduced SGIV-related mortality, and no negative effects were observed in orange-spotted grouper, Epinephelus coioides, indicating that these DNA aptamers may be suitable antiviral candidates for controlling SGIV infections in fish reared in marine aquaculture facilities.
Collapse
Affiliation(s)
- Pengfei Li
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Yang Yan
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China
| | - Shina Wei
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China
| | - Jingguang Wei
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China
| | - Ren Gao
- State Key Laboratory Breeding Base for Sustainable Exploitation of Tropical Biotic Resources, College of Marine Science, Hainan University, Haikou 570228, China
| | - Xiaohong Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China
| | - Youhua Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China
| | - Guohua Jiang
- Analytical and Testing Center, Beijing Normal University, Xinjiekouwai Street, Beijing 100875, China.
| | - Qiwei Qin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China.
| |
Collapse
|
9
|
Ou-yang Z, Wang P, Huang Y, Huang X, Wan Q, Zhou S, Wei J, Zhou Y, Qin Q. Selection and identification of Singapore grouper iridovirus vaccine candidate antigens using bioinformatics and DNA vaccination. Vet Immunol Immunopathol 2012; 149:38-45. [DOI: 10.1016/j.vetimm.2012.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 05/15/2012] [Accepted: 05/26/2012] [Indexed: 11/29/2022]
|
10
|
Chen HM, Wang PH, Aravindaram K, Chen YH, Yu HH, Yang WC, Yang NS. Shikonin enhances efficacy of a gene-based cancer vaccine via induction of RANTES. J Biomed Sci 2012; 19:42. [PMID: 22494696 PMCID: PMC3353861 DOI: 10.1186/1423-0127-19-42] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 04/12/2012] [Indexed: 01/01/2023] Open
Abstract
Background Shikonin, a phytochemical purified from Lithospermum erythrorhizon, has been shown to confer diverse pharmacological activities, including accelerating granuloma formation, wound healing, anti-inflammation and others, and is explored for immune-modifier activities for vaccination in this study. Transdermal gene-based vaccine is an attractive approach for delivery of DNA transgenes encoding specific tumor antigens to host skin tissues. Skin dendritic cells (DCs), a potent antigen-presenting cell type, is known to play a critical role in transmitting and orchestrating tumor antigen-specific immunities against cancers. The present study hence employs these various components for experimentation. Method The mRNA and protein expression of RANTES were detected by RT-PCR and ELISA, respectively. The regional expression of RANTES and tissue damage in test skin were evaluated via immunohistochemistry assay. Fluorescein isothiocyanate sensitization assay was performed to trace the trafficking of DCs from the skin vaccination site to draining lymph nodes. Adjuvantic effect of shikonin on gene gun-delivered human gp100 (hgp100) DNA cancer vaccine was studied in a human gp100-transfected B16 (B16/hgp100) tumor model. Results Among various phytochemicals tested, shikonin induced the highest level of expression of RANTES in normal skin tissues. In comparison, mouse RANTES cDNA gene transfection induced a higher level of mRANTES expression for a longer period, but caused more extensive skin damage. Topical application of shikonin onto the immunization site before gene gun-mediated vaccination augmented the population of skin DCs migrating into the draining lymph nodes. A hgp100 cDNA gene vaccination regimen with shikonin pretreatment as an adjuvant in a B16/hgp100 tumor model increased cytotoxic T lymphocyte activities in splenocytes and lymph node cells on target tumor cells. Conclusion Together, our findings suggest that shikonin can effectively enhance anti-tumor potency of a gene-based cancer vaccine via the induction of RANTES expression at the skin immunization site.
Collapse
Affiliation(s)
- Hui-Ming Chen
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
11
|
Kim YC, Song JM, Lipatov AS, Choi SO, Lee JW, Donis RO, Compans RW, Kang SM, Prausnitz MR. Increased immunogenicity of avian influenza DNA vaccine delivered to the skin using a microneedle patch. Eur J Pharm Biopharm 2012; 81:239-47. [PMID: 22504442 DOI: 10.1016/j.ejpb.2012.03.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 02/16/2012] [Accepted: 03/14/2012] [Indexed: 10/28/2022]
Abstract
Effective public health responses to an influenza pandemic require an effective vaccine that can be manufactured and administered to large populations in the shortest possible time. In this study, we evaluated a method for vaccination against avian influenza virus that uses a DNA vaccine for rapid manufacturing and delivered by a microneedle skin patch for simplified administration and increased immunogenicity. We prepared patches containing 700-μm long microneedles coated with an avian H5 influenza hemagglutinin DNA vaccine from A/Viet Nam/1203/04 influenza virus. The coating DNA dose increased with DNA concentration in the coating solution and the number of dip-coating cycles. Coated DNA was released into the skin tissue by dissolution within minutes. Vaccination of mice using microneedles induced higher levels of antibody responses and hemagglutination inhibition titers, and improved protection against lethal infection with avian influenza as compared to conventional intramuscular delivery of the same dose of the DNA vaccine. Additional analysis showed that the microneedle coating solution containing carboxymethylcellulose and a surfactant may have negatively affected the immunogenicity of the DNA vaccine. Overall, this study shows that DNA vaccine delivery by microneedles can be a promising approach for improved vaccination to mitigate an influenza pandemic.
Collapse
Affiliation(s)
- Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
ELISPOT analysis of a new CTL based DNA vaccine for HIV-1 using GM-CSF in DNA prime/peptide boost strategy: GM-CSF induced long-lived memory responses. Immunol Lett 2011; 140:14-20. [DOI: 10.1016/j.imlet.2011.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 05/15/2011] [Accepted: 05/15/2011] [Indexed: 11/19/2022]
|
13
|
Liu F, Liang M, Cao S, Liu Q, Zhang Q, Li C, Zhang S, Wang S, Li D. Fusion with extracellular domain of cytotoxic T-lymphocyte-associated-antigen 4 leads to enhancement of immunogenicity of Hantaan virus DNA vaccines in C57BL/6 mice. Virol J 2011; 8:448. [PMID: 21943202 PMCID: PMC3204296 DOI: 10.1186/1743-422x-8-448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/23/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Hantaan virus (HTNV) is the causative agent of the most severe form of a rodent-borne disease known as hemorrhagic fever with renal syndrome (HFRS). A safe and effective HTNV vaccine is needed. Vaccination with DNA constructs expressing fused antigen with bioactive factors, has shown promising improvement of immunogenicity for viral agents in animal models, but the effect of fusion strategy on HTNV DNA vaccine has not been investigated. RESULTS DNA plasmids encoding the HTNV nucleocapsid protein (N) and glycoprotein (Gn and Gc) in fusion to the extracellular domain of cytotoxic T-lymphocyte-associated-antigen 4 (eCTLA-4) targeting to antigen presenting cells (APCs) were constructed. Intramuscular immunization of mice with plasmids expressing eCTLA-4-HTNV-N/GP fusion proteins leads to a significant enhancement of the specific antibody response as well as cytotoxic T-lymphocyte (CTL) response in C57BL/6 mice. Moreover, this effect could be further augmented when co-administered with CpG motifs. CONCLUSIONS Modification of viral antigen in fusion to bioactive factor will be promising to confer efficient antigen presentation and improve the potency of DNA vaccine in mice.
Collapse
Affiliation(s)
- Feng Liu
- Laboratory for Viral Hemorrhagic Fever, National Institute for Viral Disease Control and Prevention, China CDC 100 Ying Xin Jie, Xuan Wu Qu, Beijing 100052, China
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Shimmoto H, Kawai K, Ikawa T, Oshima SI. Protection of red sea bream Pagrus major against red sea bream iridovirus infection by vaccination with a recombinant viral protein. Microbiol Immunol 2010; 54:135-42. [DOI: 10.1111/j.1348-0421.2010.00204.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
Choudhury S, Ganguly A, Chakrabarti K, Sharma RK, Gupta SK. DNA vaccine encoding chimeric protein encompassing epitopes of human ZP3 and ZP4: immunogenicity and characterization of antibodies. J Reprod Immunol 2008; 79:137-47. [PMID: 19004505 DOI: 10.1016/j.jri.2008.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 08/19/2008] [Accepted: 09/15/2008] [Indexed: 10/21/2022]
Abstract
Immunization with zona pellucida (ZP) glycoproteins leads to curtailment of fertility often associated with ovarian dysfunction. To avoid ovarian dysfunction, synthetic peptides corresponding to ZP glycoproteins have been proposed as candidate immunogens. In the present study, plasmid DNA encoding a human ZP glycoprotein-3 (ZP3) epitope corresponding to amino acid (aa) residues 334-343 and a human ZP glycoprotein-4 (ZP4) epitope corresponding to aa residues 251-273 separated by a triglycine spacer was constructed using the mammalian expression vector, VR1020. The plasmid DNA construct expressed both human ZP3 and ZP4 epitopes, as revealed by transient transfection of COS-1 (African green monkey, kidney) mammalian cells. Active immunization of female BALB/cJ mice with the DNA vaccine led to generation of antibodies reactive with baculovirus-expressed recombinant human ZP3, ZP4 and ZP3((334-343aa))-GGG-ZP4((251-273aa)) synthetic peptide in an ELISA as well as T cell responses. Antibodies generated by the DNA vaccine also recognized native ZP. The immune sera significantly inhibited (p<0.005) the binding of FITC-labeled ZP3 to capacitated human sperm, whereas no inhibition in the binding of FITC-labeled ZP4 was observed. However, a significant decrease in acrosomal exocytosis mediated by both recombinant human ZP3 (p<0.005) and ZP4 (p<0.005) was observed in presence of the immune sera. These studies demonstrate that a DNA vaccine can be designed to elicit antibodies against small epitopes of ZP glycoproteins.
Collapse
Affiliation(s)
- Sangeeta Choudhury
- Gamete Antigen Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | | | | | | | |
Collapse
|
16
|
Kim TJ, Jang EJ, Lee JI. Vaccination of rock bream, Oplegnathus fasciatus (Temminck & Schlegel), using a recombinant major capsid protein of fish iridovirus. JOURNAL OF FISH DISEASES 2008; 31:547-551. [PMID: 18577102 DOI: 10.1111/j.1365-2761.2007.00853.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Affiliation(s)
- T J Kim
- Biotherapy Human Resources Center, Chonnam National University, Gwangju, Korea
| | | | | |
Collapse
|
17
|
Ma K, Xu W, Shao X, Hu L, Xu H, Yuan Z, Zheng X, Xiong S. Coimmunization with RANTES plasmid polarized Th1 immune response against hepatitis B virus envelope via recruitment of dendritic cells. Antiviral Res 2007; 76:140-9. [PMID: 17655942 DOI: 10.1016/j.antiviral.2007.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2007] [Revised: 06/16/2007] [Accepted: 06/19/2007] [Indexed: 11/25/2022]
Abstract
Induction of T help cell type 1 (Th1) response seems to be a prerequisite of HBV clearance. DNA vaccines have shown its potential to elicit Th1-biased immune response. However, its immunogenicity needs to be improved. Regulated upon activation normal T cell expressed and secreted (RANTES) is an inflammatory chemokine that promotes the accumulation and activation of CD4+, CD8+ T cells, and dendritic cells (DCs), which would favor antiviral immunity. In this study, the efficacy of a DNA vaccine encoding hepatitis B virus (HBV) preS2 plus S protein was enhanced through co-injection of a plasmid encoding RANTES in a BALB/c model. Co-injection of RANTES gene resulted in a moderate increase in the HBV specific humoral and cellular immune responses and a significant increase following an HBsAg booster vaccination compared to DNA encoding HBsAg alone. This enhancement was due to an enrichment of DCs in the draining lymph node and an up-regulation of DCs maturation by RANTES. More importantly, RANTES polarized the specific immunity towards a dominant Th1 profile and even converted an established Th2 response to a Th1 phenotype. Our study suggested the feasibility of using a plasmid-encoded RANTES as a modulatory Th1 adjuvant in genetic vaccination.
Collapse
Affiliation(s)
- Ke Ma
- Institute for ImmunoBiology and Department of Immunology, Shanghai Medical College of Fudan University, 138, Yi Xue Yuan Road, Shanghai 200032, PR China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Caipang CMA, Takano T, Hirono I, Aoki T. Genetic vaccines protect red seabream, Pagrus major, upon challenge with red seabream iridovirus (RSIV). FISH & SHELLFISH IMMUNOLOGY 2006; 21:130-8. [PMID: 16359871 DOI: 10.1016/j.fsi.2005.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 10/31/2005] [Accepted: 10/31/2005] [Indexed: 05/05/2023]
Abstract
We have investigated the protective effect of immunization of juvenile red seabream, Pagrus major, with DNA plasmids encoding the viral major capsid protein (MCP) and an open reading frame (ORF) containing a transmembrane domain against red seabream iridovirus (RSIV). The expression of the MHC class I transcript in the DNA-vaccinated fish was significantly upregulated at the 15th day post-vaccination and the relative level of expression was maintained until the 30th day post-vaccination. This pattern of expression was similar in fish vaccinated with a commercially prepared formalin-inactivated RSIV vaccine. In vaccine efficiency tests, the relative percentage survival (RPS) of fish receiving the DNA vaccines and their combination ranged from 42.8 to 71.4% in two experimental runs, and these were significantly different from the control groups. Our results clearly demonstrate that DNA vaccines are able to induce robust protection in fish against RSIV infection, and a cellular immune response as shown by the upregulation of the MHC class I transcript after vaccination, which may be associated with such protection.
Collapse
Affiliation(s)
- Christopher Marlowe A Caipang
- Laboratory of Genome Science, Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato 108-8477, Tokyo, Japan
| | | | | | | |
Collapse
|
19
|
Loots K, Vleugels B, Ons E, Vanrompay D, Goddeeris BM. Evaluation of the persistence and gene expression of an anti-Chlamydophila psittaci DNA vaccine in turkey muscle. BMC Vet Res 2006; 2:18. [PMID: 16764710 PMCID: PMC1524740 DOI: 10.1186/1746-6148-2-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Accepted: 06/09/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND DNA vaccination has been shown to elicit specific cellular and humoral immune responses to many different agents in a broad variety of species. However, looking at a commercial use, the duration of the immune response against the vaccine is critical. Therefore the persistence of the DNA vaccine, as well as its expression, should be investigated. We conducted these investigations on a DNA vaccine against Chlamydophila psittaci, a Gram-negative intracellular bacterium which causes respiratory disease in turkeys and humans. Previous studies showed that the DNA vaccine confers partial protection against C. psittaci infection in turkeys. Turkeys were injected intramuscularly with the DNA vaccine : a eukaryotic expression vector (pcDNA1::MOMP) expressing the major outer membrane protein (MOMP) of an avian C. psittaci serovar D strain. Over a period of 11 weeks, cellular uptake of the DNA vaccine was examined by PCR, transcription of the insert by reverse transcript-PCR (RT-PCR) and mRNA translation by immunofluorescence staining of muscle biopsies. RESULTS The results indicate that the DNA vaccine persists in turkey muscle for at least 10 weeks. Moreover, during this period of time MOMP was continuously expressed, as evidenced by the immunofluorescence staining and RT-PCR. CONCLUSION Since C. psittaci infections occur at the age of 3 to 6 and 8 to 12 weeks, a vaccine persistence of 10 weeks seems adequate. Therefore, further research should concentrate on improving the elicited immune response, more specifically the cell-mediated immune response, rather than prolonging the lifespan of the plasmid.
Collapse
Affiliation(s)
- Karolien Loots
- Division of Gene Technology, Department of Biosystems, Faculty of Bioscience Engineering, Catholic University Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
| | - Bart Vleugels
- Division of Gene Technology, Department of Biosystems, Faculty of Bioscience Engineering, Catholic University Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
| | - Ellen Ons
- Division of Gene Technology, Department of Biosystems, Faculty of Bioscience Engineering, Catholic University Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
| | - Daisy Vanrompay
- Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Bruno Maria Goddeeris
- Division of Gene Technology, Department of Biosystems, Faculty of Bioscience Engineering, Catholic University Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
- Laboratory of Veterinary Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
20
|
Abstract
Strategies for gene delivery comprise a diverse range of live and synthetic approaches; DNA delivery for the purposes of immunisation in turn comprises a large part of this research. This review mainly discusses synthetic systems for application in the delivery of plasmid DNA vaccines, outlining polylactide-co-glycolide, liposome, chitosan and complex combination delivery systems. Areas of promise for DNA vaccine candidates include immune modulation of allergic responses and veterinarian application. The potential for realistic consideration of DNA vaccines as an alternative to existing approaches is dependent on the development of efficient DNA vaccine vectors and improved systems for DNA vaccine delivery. DNA vaccine technology may yet prove to be an important asset in an environment where there is a critical need for therapeutic and prophylactic strategies to combat a wide range of disease states.
Collapse
Affiliation(s)
- H Oya Alpar
- University of London, School of Pharmacy, UK.
| | | | | |
Collapse
|
21
|
Azizi A, Aucoin S, Tadesse H, Frost R, Ghorbani M, Soare C, Naas T, Diaz-Mitoma F. A combined nucleocapsid vaccine induces vigorous SARS-CD8+ T-cell immune responses. GENETIC VACCINES AND THERAPY 2005; 3:7. [PMID: 16115319 PMCID: PMC1249587 DOI: 10.1186/1479-0556-3-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2005] [Accepted: 08/22/2005] [Indexed: 12/04/2022]
Abstract
Several studies have shown that cell-mediated immune responses play a crucial role in controlling viral replication. As such, a candidate SARS vaccine should elicit broad CD8+ T-cell immune responses. Several groups of mice were immunized alone or in combination with SARS-nucleocapsid immunogen. A high level of specific SARS-CD8+ T-cell response was demonstrated in mice that received DNA encoding the SARS-nucleocapsid, protein and XIAP as an adjuvant. We also observed that co-administration of a plasmid expressing nucleocapsid, recombinant protein and montanide/CpG induces high antibody titers in immunized mice. Moreover, this vaccine approach merits further investigation as a potential candidate vaccine against SARS.
Collapse
Affiliation(s)
- Ali Azizi
- Infectious Disease and Vaccine Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M2, Canada
| | - Susan Aucoin
- Infectious Disease and Vaccine Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Helina Tadesse
- Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M2, Canada
| | - Rita Frost
- Infectious Disease and Vaccine Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Masoud Ghorbani
- Infectious Disease and Vaccine Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Catalina Soare
- Infectious Disease and Vaccine Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M2, Canada
| | - Turaya Naas
- Infectious Disease and Vaccine Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M2, Canada
| | - Francisco Diaz-Mitoma
- Infectious Disease and Vaccine Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M2, Canada
| |
Collapse
|
22
|
Xu W, Chu Y, Zhang R, Xu H, Wang Y, Xiong S. Endoplasmic reticulum targeting sequence enhances HBV-specific cytotoxic T lymphocytes induced by a CTL epitope-based DNA vaccine. Virology 2005; 334:255-63. [PMID: 15780875 DOI: 10.1016/j.virol.2005.01.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2004] [Revised: 12/20/2004] [Accepted: 01/28/2005] [Indexed: 01/12/2023]
Abstract
CD8(+) T cells play a critical role in protective immunity against Hepatitis B Virus (HBV). Epitope-based DNA vaccines expressing HBV-dominant CTL epitopes can be used as candidate vaccines capable of inducing cytotoxic T Lymphocytes (CTL) responses. A plasmid DNA encoding a CTL epitope of HBV core antigen, HBc(18-27), was constructed. Intramuscular immunization of C57BL/6 mice with this DNA vaccine resulted in successful induction of HBV-specific CTL responses. In order to promote transportation of the peptide into endoplasmic reticulum (ER) to bind to MHC class I molecules for optimal class I antigen presentation, an ER targeting sequence (ERTS) was fused with the C(18-27) encoding gene. ERTS fusion significantly enhanced specific CD8(+) T cell responses in terms of CTL cytolysis as well as IFN-gamma secretion. This enhancement was correlated with promoted epitope presentation on target cell surface. We report here an enhanced immunogenicity of an epitope-based DNA vaccine using an ER targeting signal sequence, which has significant implications for future design of therapeutic HBV vaccine.
Collapse
Affiliation(s)
- Wei Xu
- Department of Immunology, Shanghai Medical College of Fudan University, Shanghai, PR China
| | | | | | | | | | | |
Collapse
|
23
|
Sun CJ, Pan SP, Xie QX, Xiao LJ. Preparation of chitosan-plasmid DNA nanoparticles encoding zona pellucida glycoprotein-3alpha and its expression in mouse. Mol Reprod Dev 2005; 68:182-8. [PMID: 15095339 DOI: 10.1002/mrd.20058] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the present study, the porcine zona pellucida (ZP)-3alpha eukaryotic expression vector pVAX1-pZP3alpha was constructed by genetic recombinant technology, then the recombinant plasmid was encapsulated in nanoparticles with chitosan, and the imaging of chitosan/pVAX1-pZP3alpha nanoparticles by Atomic Force Microscope (AFM) was processed. Feeding mouse with those microencapsulation by gastric larvae, and after 5 days, detecting its expression in mouse intestine by RT-PCR and indirect immunofluorescence (IIF). Results show that the porcine ZP-3alpha eukaryotic expression vector pVAX1-pZP3alpha had been constructed correctly, and the chitosan-DNA expressing ZP microencapsulation was prepared successfully. After 5 days of feeding mouse, the transcription and expression of those DNA vaccines were found in mouse alvine chorion. The preparation of chitosan/pVAX1-pZP3alpha plasmid DNA nanoparticles and its expression in mice will help to investigate the feasibility of ZP DNA vaccine to induce oviduct local mucosal immunity against ZP to block the fertilization without causing ovarian dysfunction, which will provide new ideas and ways for research and exploiting more effective, more convenient oral contraceptive vaccines.
Collapse
Affiliation(s)
- Cai-Jun Sun
- Research Center for Reproductive Immunology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, China
| | | | | | | |
Collapse
|
24
|
Abstract
Classical immunization protocols have produced an antibody-based humoral response that is very effective against susceptible infectious diseases. Immunization introduces an external substance to induce the host immune system to respond specifically. Typically an antigen is used, but DNA, or a primed, pre-existing leukocyte or antigen-presenting cell, can also be used. Immunization is currently being used or investigated for the prevention and treatment of infectious diseases, cancer, addictions, allergies, pregnancy, and autoimmune diseases. It is also being used to produce biologically active materials such as polyclonal and monoclonal antibodies, antivenins, and anti-toxins for treating a wide range of conditions. Animals have been integral to the development of immunization techniques, as producers of toxoids and antitoxins, as models (e.g., to validate materials and protocols used for immunization, to understand the impact of immunization itself on the immune system, and to help investigators devise methods for determining the efficacy of vaccines) and as beneficiaries themselves of vaccines and antitoxins. The choice of immunization protocols is complex, and results may be affected by many factors such as dose and concentration of antigen, choice of adjuvants, time between inoculation and response measurement, and method of detection. The immune system responses to an antigen are also complex and continue to develop with advancing age. Anatomical, physiological, and immune system differences between species influence responses to immunization, as do the purity and presentation of the antigens and adjuvants. When directly comparing results, animals should be sourced from the same supplier. This review highlights the many uses of immunization techniques and introduces important considerations for the choice of protocols and animal models.
Collapse
Affiliation(s)
- Michael K Schunk
- Laboratories de Contrôle, Sanofi-Pasteur, Marcy L'Etoile, France
| | | |
Collapse
|
25
|
Purcell MK, Kurath G, Garver KA, Herwig RP, Winton JR. Quantitative expression profiling of immune response genes in rainbow trout following infectious haematopoietic necrosis virus (IHNV) infection or DNA vaccination. FISH & SHELLFISH IMMUNOLOGY 2004; 17:447-62. [PMID: 15313511 DOI: 10.1016/j.fsi.2004.04.017] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Accepted: 04/28/2004] [Indexed: 05/20/2023]
Abstract
Infectious haematopoietic necrosis virus (IHNV) is a well-studied virus of salmonid fishes. A highly efficacious DNA vaccine has been developed against this virus and studies have demonstrated that this vaccine induces both an early and transient non-specific anti-viral phase as well as long-term specific protection. The mechanisms of the early anti-viral phase are not known, but previous studies noted changes in Mx gene expression, suggesting a role for type I interferon. This study used quantitative real-time reverse transcriptase PCR methodology to compare expression changes over time of a number of cytokine or cytokine-related genes in the spleen of rainbow trout following injection with poly I:C, live IHNV, the IHNV DNA vaccine or a control plasmid encoding the non-antigenic luciferase gene. The target genes included Mx-1, viral haemorrhagic septicaemia virus induced gene 8 (Vig-8), TNF-alpha1, TNF-alpha2, IL-1beta1, IL-8, TGF-beta1 and Hsp70. Poly I:C stimulation induced several genes but the strongest and significant response was observed in the Mx-1 and Vig-8 genes. The live IHN virus induced a significant response in all genes examined except TGF-beta1. The control plasmid construct and the IHNV DNA vaccine marginally induced a number of genes, but the main difference between these two groups was a statistically significant induction of the Mx-1 and Vig-8 genes by the IHNV vaccine only. The gene expression profiles elicited by the live virus and the IHNV DNA vaccine differed in a number of aspects but this study confirms the clear role for a type I interferon-like response in early anti-viral defence.
Collapse
Affiliation(s)
- Maureen K Purcell
- School of Aquatic and Fishery Sciences, University of Washington, Box 355020, Seattle, WA 98195, USA.
| | | | | | | | | |
Collapse
|
26
|
Takano T, Iwahori A, Hirono I, Aoki T. Development of a DNA vaccine against hirame rhabdovirus and analysis of the expression of immune-related genes after vaccination. FISH & SHELLFISH IMMUNOLOGY 2004; 17:367-374. [PMID: 15312663 DOI: 10.1016/j.fsi.2004.04.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Accepted: 04/14/2004] [Indexed: 05/24/2023]
Abstract
Intramuscular injection of Japanese flounder, Paralichthys olivaceus (average weight approximately 2 g) with 1 and 10 microg of a plasmid DNA vaccine encoding the hirame rhabdovirus (HIRRV) glycoprotein gene (pCMV-HRVg) was found to provide strong protection against HIRRV. We also conducted a real-time PCR analysis to quantify immune-related genes, e.g. MHC class Ialpha, IIalpha, IIbeta, TCR-alpha, beta1, beta2 and delta, to characterize the immune response at 1 and 7 days after DNA vaccination. In general, the copy numbers were at least 2-fold higher than those of the non-vaccinated fish. Interestingly, the gene expression of TCR beta1 and beta2 increased 1 day post-DNA vaccination, after which their copy numbers returned to levels similar to those before vaccination. These results suggest that the immune system of Japanese flounder was activated immediately after DNA immunization.
Collapse
Affiliation(s)
- Tomokazu Takano
- Laboratory of Genome Science, Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Konan 4-5-7, Minato, Tokyo 108-8477, Japan
| | | | | | | |
Collapse
|
27
|
Otero M, Calarota SA, Felber B, Laddy D, Pavlakis G, Boyer JD, Weiner DB. Resiquimod is a modest adjuvant for HIV-1 gag-based genetic immunization in a mouse model. Vaccine 2004; 22:1782-90. [PMID: 15068862 DOI: 10.1016/j.vaccine.2004.01.037] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
DNA vaccines have been effective at generating useful immune responses in many animal species. However, it is clearly desirable to increase their potency. The identification of adjuvants that increase their cell-mediated immune (CMI) response is therefore an important goal. Resiquimod is an imiquimod analog proven to activate dendritic cells through TLR-7. The adjuvant capacity of resiquimod has not, to our knowledge, been studied in the context of genetic immunization. Here, we studied resiquimod as an adjuvant for plasmid vaccine therapy by intra-muscular immunization of BALB/c mice with HIV-1 gag DNA vaccine without and with several concentrations of resiquimod (ranging from 5-100nM). We observed that resiquimod moderately enhanced IFN-gamma production as measured by a peptide-based ELISPOT assay compared to that obtained in mice immunized with DNA gag only. Antigen-specific T-cell proliferation studies showed a several-fold increase in the stimulation index in mice immunized with DNA gag +50 nM of resiquimod as compared to mice receiving DNA gag alone. Antibody titer also increased, while the antibody isotyping data showed a strong Th1 biased type response. Analysis of cytokine production in serum samples demonstrated a stronger Th1 cytokine bias in the presence of resiquimod. Furthermore, relevant increase in IL-4 production, as measured by ELISPOT assay, was not observed. Our results show that resiquimod can have modest adjuvant activity, in a DNA formulation, driving the immune system towards a cell-mediated immune response. Additional studies involving this adjuvant for DNA vaccines are underway.
Collapse
Affiliation(s)
- Miguel Otero
- Department of Pathology, School of Medicine, University of Pennsylvania, 422 Curie Blvd., 505 Stellar-Chance Bldg., Philadelphia, PA 19104-6100, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Li BW, Rush A, Zhang SR, Curtis KC, Weil GJ. Antibody responses to Brugia malayi antigens induced by DNA vaccination. FILARIA JOURNAL 2004; 3:1. [PMID: 14738569 PMCID: PMC343290 DOI: 10.1186/1475-2883-3-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2003] [Accepted: 01/22/2004] [Indexed: 01/14/2023]
Abstract
Background DNA vaccination is a convenient means of immunizing animals with recombinant parasite antigens. DNA delivery methods are believed to affect the qualitative nature of immune responses to DNA vaccines in ways that may affect their protective activity. However, relatively few studies have directly compared immune responses to plasmids encoding the same antigens after injection by different routes. Therefore, the purpose of this study was to explore the influence of the route of administration on antibody responses to plasmids encoding antigens from the filarial nematode parasite Brugia malayi. Methods Four B. malayi genes and partial genes encoding paramyosin (BM5), heat shock protein (BMHSP-70), intermediate filament (BMIF) and a serodiagnostic antigen (BM14) were inserted in eukaryotic expression vectors (pJW4303 and pCR™3.1). BALB/c mice were immunized with individual recombinant plasmids or with a cocktail of all four plasmids by intramuscular injection (IM) or by gene gun-intradermal inoculation (GG). Antibody responses to recombinant antigens were measured by ELISA. Mean IgG1 to IgG2a antibody ratios were used as an indicator of Th1 or Th2 bias in immune responses induced with particular antigens by IM or GG immunization. The statistical significance of group differences in antibody responses was assessed by the non-parametric Kruskal-Wallis test. Results Mice produced antibody responses to all four filarial antigens after DNA vaccination by either the IM or GG route. Antibody responses to BM5 paramyosin were strongly biased toward IgG1 with lower levels of IgG2a after GG vaccination, while IM vaccination produced dominant IgG2a antibody responses. Antibody responses were biased toward IgG1 after both IM and GG immunization with BMIF, but antibodies were biased toward IgG2a after IM and GG vaccination with BMHSP-70 and BM14. Animals injected with a mixture of four recombinant plasmid DNAs produced antibodies to all four antigens. Conclusions Our results show that monovalent and polyvalent DNA vaccination successfully induced antibody responses to a variety of filarial antigens. However, antibody responses to different antigens varied in magnitude and with respect to isotype bias. The isotype bias of antibody responses following DNA vaccination can be affected by route of administration and by intrinsic characteristics of individual antigens.
Collapse
Affiliation(s)
- Ben-Wen Li
- Infectious Diseases Division, Campus Box 8051, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Amy Rush
- Infectious Diseases Division, Campus Box 8051, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Shaorong R Zhang
- Infectious Diseases Division, Campus Box 8051, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Kurt C Curtis
- Infectious Diseases Division, Campus Box 8051, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Gary J Weil
- Infectious Diseases Division, Campus Box 8051, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
29
|
|
30
|
Blanchard P, Mahé D, Cariolet R, Keranflec'h A, Baudouard MA, Cordioli P, Albina E, Jestin A. Protection of swine against post-weaning multisystemic wasting syndrome (PMWS) by porcine circovirus type 2 (PCV2) proteins. Vaccine 2003; 21:4565-75. [PMID: 14575769 DOI: 10.1016/s0264-410x(03)00503-6] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Porcine circovirus type 2 (PCV2) is known to be associated with post-weaning multisystemic wasting syndrome (PMWS), a recently described disease of young pigs. Since no PCV2 vaccine was available so far, we have developed a specific PCV2 vaccine candidate. The Orf1-encoded replication protein and Orf2-encoded capsid protein of PCV2 were expressed and detected in either mammalian or insect expression systems. In a first trial, Orf2 protein was found to be a major immunogen, inducing protection in a prime-boost protocol; the piglets received a first injection with plasmids directing Orf2 protein and granulocyte-macrophage colony-stimulating factor (GM-CSF) expression, followed by a second injection, a fortnight later, associated with baculovirus-expressed Orf2 protein. As evaluated by growth parameters, clinical signs (fever), seroconversion, the pigs were protected against a PCV2 challenge after vaccination. In a second trial, protection induced by a subunit vaccine was even better than the one induced by DNA vaccine, since PCV2 replication was completely inhibited.
Collapse
Affiliation(s)
- P Blanchard
- Agence Française de Sécurité Sanitaire des Aliments (AFSSA) Ploufragan, Unité Génétique Virale et Biosécurité, BP 53, Zoopôle Les Croix, Fr 22440 Ploufragan, France.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Verri T, Ingrosso L, Chiloiro R, Danieli A, Zonno V, Alifano P, Romano N, Scapigliati G, Vilella S, Storelli C. Assessment of DNA vaccine potential for gilthead sea bream (Sparus aurata) by intramuscular injection of a reporter gene. FISH & SHELLFISH IMMUNOLOGY 2003; 15:283-295. [PMID: 12969650 DOI: 10.1016/s1050-4648(02)00166-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Naked circular plasmid DNA containing the cytomegalovirus (CMV)-promoter-driven lacZ reporter gene (pCMV-LacZ) was injected in the epaxial muscle of gilthead sea bream (Sparus aurata). A mosaic pattern of expression of beta-galactosidase (beta-gal) in the myofibres at the site of injection was visualised by in situ histochemical staining using 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside. As measured by o-nitrophenyl-beta-D-galactopyranoside assay, beta-gal enzymatic activity was found to steadily increase for at least 50 days post injection (p.i.) in pCMV-LacZ-injected muscle. In parallel, foreign DNA was detected by polymerase chain reaction in injected muscles (but not in other tissues) up to 60 days p.i., persisting most probably in an extrachromosomal, non-replicative, circular form. Neither beta-gal activity nor pCMV-LacZ-related amplification products were found 90 days p.i. Antibodies against beta-gal were demonstrated in pCMV-LacZ-injected fish sampled 45 days p.i. The results suggest that intramuscular delivery of foreign genes represents a realistic approach for DNA vaccine technology for the prevention of infectious diseases in gilthead sea bream.
Collapse
Affiliation(s)
- Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies, University of Lecce, via Provinciale Lecce-Monteroni, I-73100, Lecce, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Temperton NJ, Quenelle DC, Lawson KM, Zuckerman JN, Kern ER, Griffiths PD, Emery VC. Enhancement of humoral immune responses to a human cytomegalovirus DNA vaccine: adjuvant effects of aluminum phosphate and CpG oligodeoxynucleotides. J Med Virol 2003; 70:86-90. [PMID: 12629648 DOI: 10.1002/jmv.10357] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A human cytomegalovirus (HCMV) glycoprotein B (gpUL55) DNA vaccine has been evaluated in BALB/c mice. Intramuscular immunization of these mice with pRc/CMV2-gB resulted in the generation of high levels of gpUL55-specific antibody (geometric mean titer [GMT] 1:8900) and neutralizing antibody (GMT 1:74) after 2 booster doses given 5 and 10 weeks after primary inoculation. Emulsifying the construct with the aluminum phosphate gel adjuvant Adju-Phos before immunization enhanced gpUL55-specific antibody responses (GMT 1:17800, P = 0.04). Co-immunization with CpG oligodeoxynucleotides was shown to enhance levels of neutralizing antibodies generated by immunization of mice with a pRc/CMV2-gB/Adju-Phos emulsion (P = 0.04). The results provide a rationale for evaluating combinations of other HCMV proteins for incorporation into a multi-target DNA vaccine, and for the optimization of adjuvant usage, to elicit enhanced levels of neutralizing antibodies. 2003.
Collapse
Affiliation(s)
- Nigel J Temperton
- Department of Virology, Royal Free and University College Medical School, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
33
|
Grosfeld H, Cohen S, Bino T, Flashner Y, Ber R, Mamroud E, Kronman C, Shafferman A, Velan B. Effective protective immunity to Yersinia pestis infection conferred by DNA vaccine coding for derivatives of the F1 capsular antigen. Infect Immun 2003; 71:374-83. [PMID: 12496187 PMCID: PMC143422 DOI: 10.1128/iai.71.1.374-383.2003] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Three plasmids expressing derivatives of the Yersinia pestis capsular F1 antigen were evaluated for their potential as DNA vaccines. These included plasmids expressing the full-length F1, F1 devoid of its putative signal peptide (deF1), and F1 fused to the signal-bearing E3 polypeptide of Semliki Forest virus (E3/F1). Expression of these derivatives in transfected HEK293 cells revealed that deF1 is expressed in the cytosol, E3/F1 is targeted to the secretory cisternae, and the nonmodified F1 is rapidly eliminated from the cell. Intramuscular vaccination of mice with these plasmids revealed that the vector expressing deF1 was the most effective in eliciting anti-F1 antibodies. This response was not limited to specific mouse strains or to the mode of DNA administration, though gene gun-mediated vaccination was by far more effective than intramuscular needle injection. Vaccination of mice with deF1 DNA conferred protection against subcutaneous infection with the virulent Y. pestis Kimberley53 strain, even at challenge amounts as high as 4,000 50% lethal doses. Antibodies appear to play a major role in mediating this protection, as demonstrated by passive transfer of anti-deF1 DNA antiserum. Taken together, these observations indicate that a tailored genetic vaccine based on a bacterial protein can be used to confer protection against plague in mice without resorting to regimens involving the use of purified proteins.
Collapse
Affiliation(s)
- Haim Grosfeld
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Rodrigues MM, Soares MBP, Vasconcelos JRC. Endogenous interleukin-4 downregulates the type 1 CD4 T cell-mediated immune response induced by intramuscular DNA immunization. J Interferon Cytokine Res 2002; 22:1137-41. [PMID: 12513913 DOI: 10.1089/10799900260442566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intramuscular (i.m.) administration of eukaryotic plasmid vectors containing foreign genes is a general immunization strategy capable of inducing protective type 1 immune responses against viral, bacterial, fungal, and parasitic infections. We have described that immunization with a plasmid containing a gene encoding a parasite antigen elicits specific type 1 protective immune responses against experimental infection with the human protozoan parasite Trypanosoma cruzi. However, we had evidence suggesting that DNA immunization concomitantly activated specific type 2 immune responses. To determine precisely the influence of the type 2 cytokine interleukin-4 (IL-4) during DNA immunization, we compared the immune responses of genetically modified IL-4-deficient or wild-type (wt) BALB/c mice. IL-4-deficient mice had a significantly lower ratio of specific serum IgG1/IgG2a, and on in vitro restimulation with antigen, their spleen cells secreted significantly higher amounts of interferon-gamma (IFN-gamma). In contrast, absence of IL-4 did not affect total serum antibody response, T cell proliferative responses, or activation of IFN-gamma-producing CD8(+) T cells. Our results suggested that in contrast to conventional adjuvants, such as alum and complete Freund's adjuvant, specific IgG1 in DNA-immunized BALB/c mice was highly dependent on IL-4. To our knowledge, our study provides the first evidence that endogenous IL-4 selectively downregulates the type 1 CD4(+) T cell-mediated immune response induced by i.m. genetic immunization, a fact that may have implications for the design of certain DNA vaccines.
Collapse
Affiliation(s)
- Mauricio M Rodrigues
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo-Escola Paulista de Medicina, São Paulo, SP, Brazil, 04023-062.
| | | | | |
Collapse
|
35
|
Tischer BK, Schumacher D, Beer M, Beyer J, Teifke JP, Osterrieder K, Wink K, Zelnik V, Fehler F, Osterrieder N. A DNA vaccine containing an infectious Marek's disease virus genome can confer protection against tumorigenic Marek's disease in chickens. J Gen Virol 2002; 83:2367-2376. [PMID: 12237417 DOI: 10.1099/0022-1317-83-10-2367] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A DNA vaccine containing the infectious BAC20 clone of serotype 1 Marek's disease virus (MDV) was tested for its potential to protect against Marek's disease (MD). Chickens were immunized at 1 day old with BAC20 DNA suspended either in PBS, as calcium phosphate precipitates, incorporated into chitosan nanoparticles, in Escherichia coli DH10B cells, or bound to gold particles for gene-gun delivery. Challenge infection with MDV strain EU1 was performed at 12 days old, and four out of seven birds immunized with BAC20 DNA in saline by the intramuscular route remained free of MD until day 77 after challenge infection. A delay in the development of the disease could be observed in some animals vaccinated with other BAC20 DNA formulations, but clinical MD and tumour formation were evident in all but one bird. Five out of seven animals immunized with the vaccine virus CVI988 were protected against MD, but none out of seven birds survived EU1 challenge infection after injection of negative-control plasmid DNA. In a second animal experiment, five out of 12 chickens immunized with BAC20 DNA and six out of eight birds immunized with virus reconstituted from BAC20 DNA remained free of MD after challenge infection. In contrast, none out of 12 chickens survived challenge infection after immunization with BAC20 DNA lacking the essential gE gene or with gE-negative BAC20 virus. The results suggested that an MDV BAC DNA vaccine has potential to protect chickens against MD, but that in vivo reconstitution of vaccine virus is a prerequisite for protection.
Collapse
Affiliation(s)
- B Karsten Tischer
- Institute of Molecular Biology1, Virus Diagnostics2 and Infectology3, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, Boddenblick 5a, D-17498 Insel Riems, Germany
| | - Daniel Schumacher
- Institute of Molecular Biology1, Virus Diagnostics2 and Infectology3, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, Boddenblick 5a, D-17498 Insel Riems, Germany
| | - Martin Beer
- Institute of Molecular Biology1, Virus Diagnostics2 and Infectology3, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, Boddenblick 5a, D-17498 Insel Riems, Germany
| | - Jörg Beyer
- Institute of Molecular Biology1, Virus Diagnostics2 and Infectology3, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, Boddenblick 5a, D-17498 Insel Riems, Germany
| | - Jens Peter Teifke
- Institute of Molecular Biology1, Virus Diagnostics2 and Infectology3, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, Boddenblick 5a, D-17498 Insel Riems, Germany
| | | | - Kerstin Wink
- Institute of Molecular Biology1, Virus Diagnostics2 and Infectology3, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, Boddenblick 5a, D-17498 Insel Riems, Germany
| | - Vladimir Zelnik
- Institute of Virology, Slovak Academy of Sciences, Bratislava 842 45, Slovakia5
| | | | - Nikolaus Osterrieder
- Institute of Molecular Biology1, Virus Diagnostics2 and Infectology3, Friedrich-Loeffler-Institutes, Federal Research Centre for Virus Diseases of Animals, Boddenblick 5a, D-17498 Insel Riems, Germany
| |
Collapse
|
36
|
Lorenzen N, Lorenzen E, Einer-Jensen K, LaPatra SE. DNA vaccines as a tool for analysing the protective immune response against rhabdoviruses in rainbow trout. FISH & SHELLFISH IMMUNOLOGY 2002; 12:439-453. [PMID: 12194454 DOI: 10.1006/fsim.2002.0422] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
DNA vaccines based on the glycoprotein genes of the salmonid rhabdoviruses VHSV and IHNV have been demonstrated to be very efficient in inducing a protective immune response against the respective diseases in rainbow trout. Nanogram doses of plasmid DNA delivered by intramuscular injection are sufficient to induce high levels of immunity in fingerling-size fish, whereas larger fish require more vaccine for protection. The protection is long lasting and, more surprisingly, is partly established already 4 days post vaccination. The early protection involves cross-protective anti-viral defence mechanisms, while the long duration immunity is highly specific. The nature of these immune response mechanisms is discussed and it is suggested that the efficacy of the vaccines is related to their ability to activate the innate immune system as it is activated by live virus.
Collapse
Affiliation(s)
- N Lorenzen
- Danish Veterinary Institute, Hangøvej 2, DK-8200, Aarhus N, Denmark
| | | | | | | |
Collapse
|
37
|
Abstract
The development of a vaccine for the prevention of primary cytomegalovirus (CMV) infection is a major public health priority. Live attenuated virus, recombinant viral vector, recombinant protein and peptide vaccines have been studied as potential vaccine candidates. In recent years, DNA vaccination strategies have been developed for many pathogens, including CMV. This review aims to bring together many aspects of this relatively new vaccine technology as applied to current research into the development of vaccines against CMV.
Collapse
Affiliation(s)
- N J Temperton
- Academic Centre for Travel Medicine and Vaccines, Royal Free and University College Medical School, Rowland Hill Street, London NW3 2PF, UK.
| |
Collapse
|
38
|
Liu C, Kokuho T, Kubota T, Watanabe S, Inumaru S, Yokomizo Y, Onodera T. DNA mediated immunization with encoding the nucleoprotein gene of porcine transmissible gastroenteritis virus. Virus Res 2001; 80:75-82. [PMID: 11597750 DOI: 10.1016/s0168-1702(01)00333-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The immune response to a naked plasmid DNA encoding the nucleoprotein (N protein) of porcine transmissible gastroenteritis virus (TGEV) was investigated in this study. A complementary DNA of the entire N gene was amplified by RT-PCR, and inserted into a mammalian expression vector (pcDNA3.1) to construct a recombinant plasmid (pcDNA/N). To evaluate the immunogenicity of the construct, BALB/c mice were intramuscularly immunized with different doses (50, 100 and 200 microg/mouse) of pcDNA/N twice at a 5-week interval. An optimal antibody response was achieved with 100 microg of pcDNA/N. The response lasted at least 11 weeks after primary immunization. By western blotting analysis, the antibodies specifically recognized a 47 kDa protein corresponding to the viral N protein, but they did not reveal neutralizing activity against infectious TGEV in vitro. Immunoglobulin G2a was predominant among these antibodies, which was indicative of Th1 type cell activation in pcDNA/N immunized mice. Moreover, spleen cells from these mice showed stronger immune responses than those from live vaccine or parental vector immunized mice. These results suggest that the construct can elicit both humoral and cell-mediated immune (CMI) responses against TGEV N protein in mice.
Collapse
Affiliation(s)
- C Liu
- National Institute of Animal Health, 3-1-1 Kannondai, Tsukuba, 305-0856, Ibaraki, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Koletzki D, Schirmbeck R, Lundkvist A, Meisel H, Krüger DH, Ulrich R. DNA vaccination of mice with a plasmid encoding Puumala hantavirus nucleocapsid protein mimics the B-cell response induced by virus infection. J Biotechnol 2001; 84:73-8. [PMID: 11035190 DOI: 10.1016/s0168-1656(00)00329-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Inoculation of naked DNA has been applied for the development of prophylactic and therapeutic vaccines against different viral infections. To study the humoral immune response induced by DNA vaccination we cloned the entire nucleocapsid protein-encoding sequence of the Puumala hantavirus strain Vranica/Hällnäs into the CMV promoter-driven expression unit of the plasmid pcDNA3, generating pcDNA3-VR1. A single dose injection of 50 microg of plasmid DNA into each M. tibialis anterior of BALB/c mice induced a high-titered antibody response against the nucleocapsid protein as documented 6 and 11 weeks after immunisation. PEPSCAN analysis of a serum pool of the pcDNA3-VR1-vaccinated animals revealed antibodies reacting with epitopes covering the whole nucleocapsid protein. The epitope-specificity of the immune response induced by DNA vaccination seems to reflect the antibody response in experimentally virus-infected bank voles (the natural host of the Puumala virus) and humans. The data suggest that DNA vaccination could be used for the identification of highly immunogenic epitopes in viral proteins.
Collapse
Affiliation(s)
- D Koletzki
- Microbiology and Tumorbiology Centre, Karolinska Institute, S-171 77, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Hantaviruses are rodent-borne bunyaviruses which cause haemorrhagic fever with renal syndrome and Hantavirus pulmonary syndrome in humans. This review covers the host interactions of the viruses, including the rodent reservoirs, the clinical outcome of human infections as well as the pathogenesis and laboratory diagnosis of infections. The current stage in prophylaxis and therapy of hantaviral diseases is described and different approaches in vaccine development are discussed.
Collapse
Affiliation(s)
- D H Krüger
- Institute of Virology, Humboldt University, School of Medicine Charité, D-10098, Berlin, Germany.
| | | | | |
Collapse
|
41
|
Spack EG, Sorgi FL. Developing non-viral DNA delivery systems for cancer and infectious disease. Drug Discov Today 2001; 6:186-197. [PMID: 11173266 DOI: 10.1016/s1359-6446(00)01633-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Efforts to deliver therapeutic genes are frequently rebuffed by the body's adaptive immune response against viral delivery vectors. Attempts to circumvent this problem using non-viral delivery systems have encountered problems with transient expression and inflammatory responses induced by reaction of the innate immune system reacting against bacterial DNA. However, within the past decade, these barriers to non-viral DNA delivery have been recognized as potential allies in the development of novel vaccines for cancer and infectious disease. This review summarizes preclinical and current clinical studies testing the formulation, delivery route and adjuvant options in the development of novel DNA-based vaccines.
Collapse
Affiliation(s)
- E G. Spack
- InterMune Pharmaceuticals, CA-94010, Burlingame, USA
| | | |
Collapse
|
42
|
Fischer T, Büttner M, Rziha HJ. T helper 1-type cytokine transcription in peripheral blood mononuclear cells of pseudorabies virus (Suid herpesvirus 1)-primed swine indicates efficient immunization. Immunology 2000; 101:378-87. [PMID: 11106942 PMCID: PMC2327083 DOI: 10.1046/j.1365-2567.2000.00124.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The induction of porcine cytokines, which are believed to be important for the regulation of T helper (Th)1- and Th2-specific immune responses of pigs, was analysed after in vitro restimulation with a herpesvirus, Suid herpes 1 (pseudorabies virus [PRV]), in peripheral blood mononuclear cells (PBMC). To this end, quantitative, competitive reverse transcription-polymerase chain reaction (RT-qcPCR) was established using constructed heterologous DNA MIMICS, which contain cytokine- or glyceraldehyde-3-phosphate dehydrogenase (GAPDH)-specific primer-binding sites. This is a simple method that allows reliable determination of the differing regulation of cytokine mRNAs specific for porcine interleukin (IL)-2, -4 and -10, interferon gamma (IFN-gamma) and the housekeeping gene, GAPDH, as an endogenous control. PBMC derived from naive (innate response) and PRV-primed (memory response) outbred swine were analysed comparatively. The results demonstrated that restimulation with PRV significantly enhanced the transcription of Th1-type cytokines (IL-2 and IFN-gamma) but not of Th2-type cytokines (IL-4 and IL-10). This virus-specific cytokine response was only found with PBMC from swine protected against lethal PRV challenge infection, but not with naive PBMC or with PBMC from pigs immunized with plasmid DNA encoding PRV glycoprotein gC. Notably, PBMC derived from immune and naive pigs constitutively produced relatively high amounts of IL-10-specific mRNA, exceeding that of GAPDH mRNA, independently of the addition of viral antigen or the mitogen concanavalin A (Con A). The results of this work should help to provide a better understanding of the effector cell/cytokine network response to infection with, or vaccination against, PRV. Additionally, the simple, reliable and sensitive RT-qcPCR, when used to determine the porcine cytokine pattern, might be of prognostic value for the induction of protective immunity.
Collapse
Affiliation(s)
- T Fischer
- Federal Research Centre for Virus Diseases of Animals, Institute for Immunology, Tübingen, Germany
| | | | | |
Collapse
|
43
|
Abstract
Viruses that belong to six different families are a significant cause for neoplasia in man and animals. Among them are the Papillomaviruses that cause uterine cervical cancer in women. Efforts to develop prophylactic vaccines against viruses that cause cancer are now a major research engagement. Vaccinology, the science of vaccines, engages the sciences of immunology and of microbiology, both relying heavily on molecular biology. Successful development of vaccines relies on extensive knowledge of immunology and vaccinology. Present efforts to develop vaccines against cervical cancer caused by Papillomaviruses are focused on use of the structural antigens L1 and L2 of the virus and on the oncoproteins E6 and E7. Work on Papillomavirus vaccines has been brilliantly conceived and executed and some of vaccines are now in clinical trial. Success may follow and Papillomavirus vaccine may join with the hepatitis B virus anti-cancer vaccine in the battle against cancers of man.
Collapse
Affiliation(s)
- M R Hilleman
- Merck Institute for Therapeutic Research, Merck Research Laboratories, 770 Sumneytown Pike, West Point, PA 19486, USA
| |
Collapse
|
44
|
Zhao H, Cheng SH, Yew NS. Requirements for effective inhibition of immunostimulatory CpG motifs by neutralizing motifs. ANTISENSE & NUCLEIC ACID DRUG DEVELOPMENT 2000; 10:381-9. [PMID: 11079577 DOI: 10.1089/oli.1.2000.10.381] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The DNA of bacteria and many viruses contain unmethylated CpG dinucleotides in particular sequence contexts that activate vertebrate immune cells. A subset of these CpG motifs was previously found to oppose the effects of immunostimulatory (CpG-S) motifs and has been termed neutralizing (CpG-N) motifs. Here we show that oligodeoxynucleotides (ODNs) composed of clusters of CpG-N motifs could partially inhibit the induction of interleukin-12 (IK-12) from mouse spleen cells by ODN containing CpG-S motifs. However, non-CpG-containing ODN were also inhibitory, suggesting that neutralization of CpG-S ODNs by CpG-N ODNs in trans was nonspecific. Neutralization of CpG-S motifs by CpG-N motifs in cis was specific, but the degree of inhibition was strongly dependent on the particular CpG-S motif being neutralized, with motifs having an A residue 5' to the CG being much more resistant to inhibition than motifs having a T residue 5' to the CG. The degree of inhibition was dependent on the spacing between the CpG-S and CpG-N motifs, with the ability to neutralize inversely correlating with distance. In addition, whereas ODNs containing extended clusters of CpG-N motifs were nonstimulatory, isolated CpG-N motifs remained stimulatory in most sequence contexts. Finally, CpG-N ODNs were shown to be nonstimulatory when instilled into the lungs of BALB/c mice, but the ability of CpG-N motifs to neutralize CpG-S motifs in cis was not observed. These results show that there are precise and fairly complex interactions between immunostimulatory and inhibitory sequence motifs that govern whether a given DNA is able to activate the vertebrate immune system.
Collapse
Affiliation(s)
- H Zhao
- Genzyme Corporation, Framingham, MA 01701-9322, USA
| | | | | |
Collapse
|
45
|
McCluskie MJ, Weeratna RD, Davis HL. The role of CpG in DNA vaccines. SPRINGER SEMINARS IN IMMUNOPATHOLOGY 2000; 22:125-32. [PMID: 10944807 DOI: 10.1007/s002810000014] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
One of the most exciting developments in the field of vaccine research in recent years has been DNA vaccines, with which immune responses are induced subsequent to the in vivo expression of antigen from directly introduced plasmid DNA. Strong immune responses have been demonstrated in a number of animal models against many viral, bacterial and parasitic pathogens, and several human clinical trials have been undertaken. The strong and long-lasting antigen-specific humoral (antibodies) and cell-mediated (T help, other cytokine functions and cytotoxic T cells) immune responses induced by DNA vaccines appear to be due to the sustained in vivo expression of antigen, efficient antigen presentation and the presence of stimulatory CpG motifs. These features are desirable for the development of prophylactic vaccines against numerous infectious agents. Furthermore, the strong cellular responses are also very desirable for the development of therapeutic DNA vaccines to treat chronic viral infections or cancer. Efforts are now focusing on understanding the mechanisms for the induction of these immune responses, which in turn should aid in the optimization of DNA vaccines. This review will focus on the role of CpG motifs in DNA vaccines.
Collapse
Affiliation(s)
- M J McCluskie
- Loeb Health Research Institute at the Ottawa Hospital, Canada
| | | | | |
Collapse
|
46
|
Abstract
DNA vaccines can induce potent humoral and cellular immune responses in numerous animal models. Most DNA vaccines have been administered parenterally; however, more effective protection against mucosal pathogens could be achieved with mucosal immunization. This review concentrates on the use of DNA vaccines for the induction of mucosal immunity.
Collapse
Affiliation(s)
- M J McCluskie
- Loeb Health Research Institute at the Ottawa Hospital, 725 Parkdale Avenue, Ottawa K1Y 4E9, Canada
| | | |
Collapse
|
47
|
Route and Method of Delivery of DNA Vaccine Influence Immune Responses in Mice and Non-Human Primates. Mol Med 1999. [DOI: 10.1007/bf03402065] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|