1
|
Chagas DB, Santos FDS, de Oliveira NR, Bohn TLO, Dellagostin OA. Recombinant Live-Attenuated Salmonella Vaccine for Veterinary Use. Vaccines (Basel) 2024; 12:1319. [PMID: 39771981 PMCID: PMC11680399 DOI: 10.3390/vaccines12121319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Vaccination is essential for maintaining animal health, with priority placed on safety and cost effectiveness in veterinary use. The development of recombinant live-attenuated Salmonella vaccines (RASVs) has enabled the construction of balanced lethal systems, ensuring the stability of plasmid vectors encoding protective antigens post-immunization. These vaccines are particularly suitable for production animals, providing long-term immunity against a range of bacterial, viral, and parasitic pathogens. This review summarizes the progress made in this field, with a focus on clinical trials demonstrating the efficacy and commercial potential of RASVs in veterinary medicine.
Collapse
Affiliation(s)
- Domitila Brzoskowski Chagas
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| | - Francisco Denis Souza Santos
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
- Faculdade de Medicina, Universidade Federal do Rio Grande, Rio Grande 96200-400, Rio Grande do Sul, Brazil
| | - Natasha Rodrigues de Oliveira
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| | - Thaís Larré Oliveira Bohn
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| | - Odir Antônio Dellagostin
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, Rio Grande do Sul, Brazil (T.L.O.B.)
| |
Collapse
|
2
|
Liu J, Zhang Z, Pu W, Pan X, Li P, Bai Q, Liang S, Li C, Yu Y, Yao H, Ma J. A multi-epitope subunit vaccine providing broad cross-protection against diverse serotypes of Streptococcus suis. NPJ Vaccines 2024; 9:216. [PMID: 39543108 PMCID: PMC11564553 DOI: 10.1038/s41541-024-01015-7] [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: 08/08/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Streptococcus suis infection represents a major challenge in pig farming and public health due to its zoonotic potential and diverse serotypes, while existing vaccines lack effective cross-protection. This study employed reverse vaccinology and immunoinformatics to identify 8 conserved proteins across 11 prevalent serotypes of S. suis. 16 candidate epitopes were selected to design three multi-epitope antigens against S. suis (designated as MEASs), which fused with a dendritic cell-targeting peptide to improve antigen presentation in host. Purified MEASs displayed favorable cross-reactogenicity against 29 serotype-specific antiserums. Robust humoral and cellular immune responses can be induced by MEAS 1 and MEAS 3 in a mouse model, which provided substantial protection against virulent strains from two different serotypes. In particular, their immune serums exhibited positive opsonization effects within bloodstream and macrophage phagocytosis. Taken together, we identified two promising MEASs with excellent cross-protection, offering potential in preventing S. suis infections in a mouse model.
Collapse
Affiliation(s)
- Jianan Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Zhen Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Wanxia Pu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Xinming Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Pei Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Qiankun Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Song Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Caiying Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Yong Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| | - Jiale Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| |
Collapse
|
3
|
Yan A, Tian J, Ye J, Gao C, Ye L, Zhang D, Song Q. Construction of Toxoplasma gondii SRS29C nucleic acid vaccine and comparative immunoprotective study of an SRS29C and SAG1 combination. Mol Biochem Parasitol 2024; 259:111630. [PMID: 38795969 DOI: 10.1016/j.molbiopara.2024.111630] [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: 12/22/2023] [Revised: 03/29/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Toxoplasma gondii is an intracellular protozoan parasite that infects all nucleated cells except the red blood cells. Currently, nucleic acid vaccines are being widely investigated in Toxoplasma gondii control, and several nucleic acid vaccine candidate antigens have shown good protection in various studies. The aim of this study was to construct a nucleic acid vaccine with Toxoplasma gondii SRS29C as the target gene. We explored the nucleic acid vaccine with Toxoplasma surface protein SRS29C and the combined gene of SRS29C and SAG1 and evaluated its immunoprotective effect against Toxoplasma gondii. To amplify the gene fragment and clone it to the expression vector, the recombinant plasmid pEGFP-SRS29C was constructed by PCR. Eukaryotic cells were transfected with the plasmid, and the expression of the target protein was assessed using the Western blot method. The level of serum IgG was determined via ELISA, and the splenic lymphocyte proliferation ability was detected using the CCK-8 method. The percentages of CD4+ and CD8+ T cells were measured by flow cytometry. Mice were immunised three times with single-gene nucleic acid vaccine and combination vaccine. Splenic lymphocytokine expression was determined using ELISA kits. The mice's survival time was monitored and recorded during an in vivo insect assault experiment, and the vaccine's protective power was assessed. The outcomes showed that PCR-amplification of an SRS29C gene fragment was successful. The 4,733-bp vector fragment and the 1,119-bp target segment were both recognised by double digestion. Additionally, after transfection of the recombinant plasmid pEGFP-SRS29C, Western blot examination of the extracted protein revealed the presence of a target protein strip at 66 kDa. The test results demonstrated that the IgG content in the serum of the pEGFP-SRS29C group and the co-immunization group was significantly higher than that of the PBS group and the empty vector group. The IgG potency induced by the co-immunization group was higher than that of the pEGFP-SRS29C group and the pEGFP-SAG1 group, the number of splenic lymphocyte proliferation number was higher than that of the PBS group and the empty vector group. The CD4+/CD8+ T ratio was higher than that of the PBS group and the empty vector group. The expression of IFN-γ and TNF-α in the splenocytes of the pEGFP-SRS29C group and the combined immunisation group was significantly higher following antigen stimulation. In the worm attack experiments, mice in the PBS and empty vector groups perished within 9 days of the worm attack, whereas mice in the pEGFP-SRS29C group survived for 18 days, mice in the pEGFP-SAG1 group survived for 21 days, and mice in the co-immunization group survived for 24 days. This demonstrates that the constructed Toxoplasma gondii nucleic acid vaccine pEGFP-SRS29C and the combined gene vaccine can induce mice to develop certain humoral and cellular immune responses, and enhance their ability to resist Toxoplasma gondii infection.
Collapse
MESH Headings
- Animals
- Toxoplasma/immunology
- Toxoplasma/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/administration & dosage
- Protozoan Proteins/immunology
- Protozoan Proteins/genetics
- Protozoan Vaccines/immunology
- Protozoan Vaccines/genetics
- Mice
- Antibodies, Protozoan/blood
- Antibodies, Protozoan/immunology
- Antigens, Protozoan/immunology
- Antigens, Protozoan/genetics
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Female
- Toxoplasmosis, Animal/prevention & control
- Toxoplasmosis, Animal/immunology
- Mice, Inbred BALB C
- CD8-Positive T-Lymphocytes/immunology
- Spleen/immunology
- Spleen/parasitology
- Cell Proliferation
- Plasmids/genetics
- Plasmids/immunology
- Cytokines/metabolism
Collapse
Affiliation(s)
- An Yan
- Agricultural Animal Breeding and Healthy Breeding Main Laboratory in Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, China
| | - Jing Tian
- Agricultural Animal Breeding and Healthy Breeding Main Laboratory in Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, China
| | - Jianjun Ye
- Agricultural Animal Breeding and Healthy Breeding Main Laboratory in Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, China
| | - Chuanliang Gao
- Agricultural Animal Breeding and Healthy Breeding Main Laboratory in Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, China
| | - Liying Ye
- Agricultural Animal Breeding and Healthy Breeding Main Laboratory in Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, China
| | - Dongchao Zhang
- Agricultural Animal Breeding and Healthy Breeding Main Laboratory in Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, China
| | - Qiqi Song
- Agricultural Animal Breeding and Healthy Breeding Main Laboratory in Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, China.
| |
Collapse
|
4
|
Xu H, Zhu S, Govinden R, Chenia HY. Multiple Vaccines and Strategies for Pandemic Preparedness of Avian Influenza Virus. Viruses 2023; 15:1694. [PMID: 37632036 PMCID: PMC10459121 DOI: 10.3390/v15081694] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/14/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Avian influenza viruses (AIV) are a continuous cause of concern due to their pandemic potential and devasting effects on poultry, birds, and human health. The low pathogenic avian influenza virus has the potential to evolve into a highly pathogenic avian influenza virus, resulting in its rapid spread and significant outbreaks in poultry. Over the years, a wide array of traditional and novel strategies has been implemented to prevent the transmission of AIV in poultry. Mass vaccination is still an economical and effective approach to establish immune protection against clinical virus infection. At present, some AIV vaccines have been licensed for large-scale production and use in the poultry industry; however, other new types of AIV vaccines are currently under research and development. In this review, we assess the recent progress surrounding the various types of AIV vaccines, which are based on the classical and next-generation platforms. Additionally, the delivery systems for nucleic acid vaccines are discussed, since these vaccines have attracted significant attention following their significant role in the fight against COVID-19. We also provide a general introduction to the dendritic targeting strategy, which can be used to enhance the immune efficiency of AIV vaccines. This review may be beneficial for the avian influenza research community, providing ideas for the design and development of new AIV vaccines.
Collapse
Affiliation(s)
- Hai Xu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Roshini Govinden
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Hafizah Y. Chenia
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Durban 4001, South Africa;
| |
Collapse
|
5
|
Madlala T, Adeleke VT, Okpeku M, Tshilwane SI, Adeniyi AA, Adeleke MA. Screening of apical membrane antigen-1 (AMA1), dense granule protein-7 (GRA7) and rhoptry protein-16 (ROP16) antigens for a potential vaccine candidate against Toxoplasma gondii for chickens. Vaccine X 2023; 14:100347. [PMID: 37519774 PMCID: PMC10384181 DOI: 10.1016/j.jvacx.2023.100347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 06/23/2023] [Accepted: 07/06/2023] [Indexed: 08/01/2023] Open
Abstract
Toxoplasmosis is a zoonotic disease caused by the protozoan parasite, Toxoplasma gondii known to infect almost all animals, including birds and humans globally. This disease has impacted the livestock industry and public health, where infection of domestic animals increases the zoonotic risk of transmission of infection to humans, threatening public health. Hence the need to discover novel and safe vaccines to fight against toxoplasmosis. In the current study, a novel multiepitope vaccine was designed using immunoinformatics techniques targeting T. gondii AMA1, GRA7 and ROP16 antigens, consisting of antigenic, immunogenic, non-allergenic and cytokine inducing T-cell (9 CD8+ and 15 CD4+) epitopes and four (4) B-cell epitopes fused together using AAY, KK and GPGPG linkers. The tertiary model of the proposed vaccine was predicted and validated to confirm the structural quality of the vaccine. The designed vaccine was highly antigenic (antigenicity = 0.6645), immunogenic (score = 2.89998), with molecular weight of 73.35 kDa, instability and aliphatic index of 28.70 and 64.10, respectively; and GRAVY of -0.363. The binding interaction, stability and flexibility were assessed with molecular docking and dynamics simulation, which revealed the proposed vaccine to have good structural interaction (binding affinity = -106.882 kcal/mol) and stability when docked with Toll like receptor-4 (TLR4). The results revealed that the Profilin-adjuvanted vaccine is promising, as it predicted induction of enhanced immune responses through the production of cytokines and antibodies critical in blocking host invasion.
Collapse
Affiliation(s)
- Thabile Madlala
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, P/Bag X54001, Durban 4000, South Africa
| | - Victoria T. Adeleke
- Department of Chemical Engineering, Mangosuthu University of Technology, Durban 4031, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, P/Bag X54001, Durban 4000, South Africa
| | - Selaelo I. Tshilwane
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort 0110, South Africa
| | - Adebayo A. Adeniyi
- Department of Industrial Chemistry, Federal University, Oye-Ekiti, P.O Box 370111, Nigeria
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, South Africa
| | - Matthew A. Adeleke
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, P/Bag X54001, Durban 4000, South Africa
| |
Collapse
|
6
|
Du J, Wang T, Xu L, Wang C, Liu Y, Pan C, Chen X, Zhu Z, Luo Y, Yin C. Clostridium perfringens epsilon prototoxin mutant rpETX Y30A/Y71A/H106P/Y196A as a vaccine candidate against enterotoxemia. Vaccine 2023:S0264-410X(23)00719-3. [PMID: 37357076 DOI: 10.1016/j.vaccine.2023.06.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023]
Abstract
Epsilon toxin (ETX) is secreted by Clostridium perfringens (C. perfringens)as a relatively inactive prototoxin (pETX), which is enzymatically activated to ETX by removing carboxy-terminal and amino-terminal peptides. Genetically engineered ETX mutants have been shown to function as potential vaccine candidates in the prevention of the enterotoxemia caused by C. perfringens. In the present study, two recombinant site-directed mutants of pETX, rpETXY30A/Y71A/H106P/Y196A (rpETXm41) and rpETXY30A/H106P/Y196A/F199E (rpETXm42), were synthesized by mutating four essential amino acid residues (Tyr30, Tyr71, His106, Tyr196 or Phe199). Compared to recombinant pETX (rpETX), both rpETXm41 and rpETXm42 lacked the detectable toxicity in MDCK cells and mice, which suggested that both rpETXm41 and rpETXm42 are sufficiently safe to be vaccine candidates. Despite the fact that rpETXm41 and rpETXm42 were reactogenic with polyclonal antibodies against crude ETX, both single- and double-dose vaccination (Vs and Vd, respectively) of rpETXm41 induced a higher level of IgG titer and protection in mice than that of rpETXm42. Therefore, we selected rpETXm41 for the further study. Sheep received Vs of 150 μg rpETXm41 developed significant levels of toxin-neutralizing antibodies persisting for at least 6 months, which conferred protection against crude ETX challenge without microscopic lesions. These data suggest that genetically detoxified rpETXY30A/Y71A/H106P/Y196A could form the basis of a next-generation enterotoxemia vaccine.
Collapse
Affiliation(s)
- Jige Du
- China Institute of Veterinary Drug Control, Beijing, PR China.
| | - Tuanjie Wang
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Lei Xu
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Cong Wang
- China Animal Husbandry Industry Co., Ltd., Beijing 100070, PR China
| | - Ying Liu
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Chenfan Pan
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Xiaoyun Chen
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Zhen Zhu
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Yufeng Luo
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Chunsheng Yin
- China Institute of Veterinary Drug Control, Beijing, PR China.
| |
Collapse
|
7
|
Zhang X, Yuan H, Mahmmod YS, Yang Z, Zhao M, Song Y, Luo S, Zhang XX, Yuan ZG. Insight into the current Toxoplasma gondii DNA vaccine: a review article. Expert Rev Vaccines 2023; 22:66-89. [PMID: 36508550 DOI: 10.1080/14760584.2023.2157818] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Toxoplasma gondii (T.gondii) is a widespread protozoan with significant economic losses and public health importance. But so far, the protective effect of reported DNA-based vaccines fluctuates widely, and no study has demonstrated complete protection. AREAS COVERED This review provides an inclusive summary of T. gondii DNA vaccine antigens, adjuvants, and some other parameters. A total of 140 articles from 2000 to 2021 were collected from five databases. By contrasting the outcomes of acute and chronic challenges, we aimed to investigate and identify viable immunological strategies for optimum protection. Furthermore, we evaluated and discussed the impact of several parameters on challenge outcomes in the hopes of developing some recommendations to assist better future horizontal comparisons among research. EXPERT OPINION In the coming five years of research, the exploration of vaccine cocktails combining invasion antigens and metabolic antigens with genetic adjuvants or novel DNA delivery methods may offer us desirable protection against this multiple stage of life parasite. In addition to finding a better immune strategy, developing better in silico prediction methods, solving problems posed by variables in practical applications, and gaining a more profound knowledge of T.gondii-host molecular interaction is also crucial towards a successful vaccine.
Collapse
Affiliation(s)
- Xirui Zhang
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, PR China
| | - Hao Yuan
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, PR China
| | - Yasser S Mahmmod
- Veterinary Sciences Division, Faculty of Health Sciences, Higher Colleges of Technology, 17155, Abu Dhabi, United Arab Emirates
| | - Zipeng Yang
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, PR China
| | - Mengpo Zhao
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, PR China
| | - Yining Song
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, PR China
| | - Shengjun Luo
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, PR China
| | - Xiu-Xiang Zhang
- College of Agriculture, South China Agricultural University, 510642, Guangzhou, PR China
| | - Zi-Guo Yuan
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, PR China.,Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, PR China
| |
Collapse
|
8
|
Debnath N, Thakur M, Khushboo, Negi NP, Gautam V, Kumar Yadav A, Kumar D. Insight of oral vaccines as an alternative approach to health and disease management: An innovative intuition and challenges. Biotechnol Bioeng 2021; 119:327-346. [PMID: 34755343 DOI: 10.1002/bit.27987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022]
Abstract
Vaccination is the most suitable and persuasive healthcare program for the prohibition of various deadly diseases. However, the higher production cost and purification strategies are out of reach for the developing nations. In this scenario, development of edible vaccine turns out to be the most promising alternative for remodeling the pharmaceutical industry with reduced production and purification costs. Generally, oral route of vaccination is mostly preferred due to its safety, compliance, low manufacturing cost and most importantly the ability to induce immunity in both systemic and mucosal sites. Genetically modified microorganisms and plants could efficiently be used as vehicles for edible vaccines. Edible vaccines are supposed to reduce the risk associated with traditional vaccines. Currently, oral vaccines are available in the market for several viral and bacterial diseases like cholera, hepatitis B, malaria, rabies etc. Herein, the review focuses on the breakthrough events in the area of edible vaccines associated with dietary microbes and plants for better control over diseases.
Collapse
Affiliation(s)
- Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir (UT), India
| | - Mony Thakur
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Khushboo
- Department of Biotechnology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Neelam P Negi
- Department of Biotechnology, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashok Kumar Yadav
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir (UT), India
| | - Deepak Kumar
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
9
|
Warner RC, Chapman RC, Davis BN, Davis PH. REVIEW OF DNA VACCINE APPROACHES AGAINST THE PARASITE TOXOPLASMA GONDII. J Parasitol 2021; 107:882-903. [PMID: 34852176 DOI: 10.1645/20-157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Toxoplasma gondii is an apicomplexan parasite that affects both humans and livestock. Transmitted to humans through ingestion, it is the second-leading cause of foodborne illness-related death. Currently, there exists no approved vaccine for humans or most livestock against the parasite. DNA vaccines, a type of subunit vaccine which uses segments of the pathogen's DNA to generate immunity, have shown varying degrees of experimental efficacy against infection caused by the parasite. This review compiles DNA vaccine efforts against Toxoplasma gondii, segmenting the analysis by parasite antigen, as well as a review of concomitant adjuvant usage. No single antigenic group was consistently more effective within in vivo trials relative to others.
Collapse
Affiliation(s)
- Rosalie C Warner
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| | - Ryan C Chapman
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| | - Brianna N Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| | - Paul H Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, 68182
| |
Collapse
|
10
|
Wang N, Wang JY, Pan TX, Jiang YL, Huang HB, Yang WT, Shi CW, Wang JZ, Wang D, Zhao DD, Sun LM, Yang GL, Wang CF. Oral vaccination with attenuated Salmonella encoding the Trichinella spiralis 43-kDa protein elicits protective immunity in BALB/c mice. Acta Trop 2021; 222:106071. [PMID: 34331898 DOI: 10.1016/j.actatropica.2021.106071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 07/02/2021] [Accepted: 07/22/2021] [Indexed: 12/23/2022]
Abstract
A vaccine against Trichinella spiralis infection is urgently needed to interrupt its transmission from domestic animals to humans. However, no vaccine against T. spiralis is currently available. Our previous study demonstrated that the use of the 43-kDa glycoprotein present in excretory-secretory (ES) proteins of muscle larvae (ML) as an intramuscular DNA vaccine led to a 52.1% protection rate against T. spiralis infection. Attenuated Salmonella strains have the advantage of eliciting mucosal immunity, which is important for controlling T. spiralis infections at the intestinal stage and can be provided as vaccines via oral or intranasal routes. Therefore, in this study, complete 43-kDa glycoprotein (Ts43) sequences of T. spiralis were cloned into the vector pYA3681, and the recombinant plasmid pYA3681-Ts43 was transformed into the attenuated Salmonella typhimurium strain χ11802. The results showed that oral vaccination of mice with attenuated Salmonella carrying the recombinant plasmid pYA3681-Ts43 induced an evident elevation of the local intestinal mucosal sIgA and serum IgG antibody responses. The flow cytometry results showed that the percentages of CD4+ T cells and secreted IFN-γ, IL-4, and IL-17A in CD4+ T cells were significantly increased in the spleen and mesenteric lymph node (MLN) lymphocytes of the vaccinated groups. In addition, increased levels of the IFN-γ, IL-4, and IL-17A cytokines were also observed in the serum of the immunized groups. The above immune response results in the immunized groups demonstrated that protective immunity was elicited in this study. Finally, vaccinated mice demonstrated a significant 45.9% reduction in ML burden after infection with T. spiralis. This study demonstrated that oral vaccination with Ts43 delivered by attenuated Salmonella elicited local and systemic concurrent Th1/Th2/Th17 immune responses and provided partial protection against T. spiralis infection in BALB/c mice. This is a prospective strategy for the prevention and control of trichinellosis.
Collapse
|
11
|
Ghaffari AD, Dalimi A, Ghaffarifar F, Pirestani M. Structural predication and antigenic analysis of ROP16 protein utilizing immunoinformatics methods in order to identification of a vaccine against Toxoplasma gondii: An in silico approach. Microb Pathog 2020; 142:104079. [PMID: 32084578 DOI: 10.1016/j.micpath.2020.104079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/25/2020] [Accepted: 02/18/2020] [Indexed: 01/20/2023]
Abstract
Toxoplasmosis, caused by Toxoplasma gondii, is a common parasitic disease, affecting almost one-third of the world's population. Currently, there are no effective treatments for inhibiting the formation of chronic tissue cysts in infected hosts. Thus, the production of appropriate vaccines against this pathogen is an important goal to avoid toxoplasmosis. considering the role of rhoptry antigens like ROP16 in virulence and satisfactory immunogenicity, they can be used as promising vaccine candidates against T. gondii. In the present study, an in silico approach was used to analyze various aspects of the ROP16 protein, including physicochemical characteristics, the potential epitopes of B and T-cells, the secondary and tertiary structure, the subcellular localization, the transmembrane domain, and other important features of this protein using several bioinformatics tools to design a proper vaccine against T. gondii. The results showed that ROP16 protein includes 93 potential post-translational modification sites. The secondary structure of the ROP16 protein comprises 34.23% alpha-helix, 54.46% random coil, and 11.32% extended strand. Moreover, several potential B- and T-cell epitopes were identified for ROP16. Based on the results of Ramachandran plot, 84.64% of the amino acid residues were located in the favored, 10.34% in allowed, and 5.02% in outlier regions. Furthermore, the results of the antigenicity and allergenicity assessment noted that this protein was immunogenic and non-allergenic. Our findings suggested that structural and functional predictions applied to ROP16 protein using in silico tools can reduce the failure risk of the laboratory studies. This research provided an important basis for further studies and also developed an effective vaccine against acute and chronic toxoplasmosis by various strategies. Further studies are needed on the development of vaccines in vivo using ROP16 alone or in combination with other antigens in the future.
Collapse
Affiliation(s)
- Ali Dalir Ghaffari
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Abdolhossein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Majid Pirestani
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
12
|
Pagheh AS, Sarvi S, Sharif M, Rezaei F, Ahmadpour E, Dodangeh S, Omidian Z, Hassannia H, Mehrzadi S, Daryani A. Toxoplasma gondii surface antigen 1 (SAG1) as a potential candidate to develop vaccine against toxoplasmosis: A systematic review. Comp Immunol Microbiol Infect Dis 2020; 69:101414. [PMID: 31958746 DOI: 10.1016/j.cimid.2020.101414] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022]
Abstract
Toxoplasma gondii is an intracellular parasite that infects a broad range of animal species and humans. As the main surface antigen of the tachyzoite, SAG1 is involved in the process of recognition, adhesion and invasion of host cells. The aim of the current systematic review study is to clarify the latest status of studies in the literature regarding SAG1-associated recombinant proteins or SAG1-associated recombinant DNAs as potential vaccines against toxoplasmosis. Data were systematically collected from six databases including PubMed, Science Direct, Web of Science, Google Scholar, EBSCO and Scopus, up to 1st of January 2019. A total of 87 articles were eligible for inclusion criteria in the current systematic review. The most common antigens used for experimental cocktail vaccines together with SAG1 were ROP2 and SAG2. In addition, the most parasite strains used were RH and ME49. Freund's adjuvant and cholera toxin have been predominantly utilized. Furthermore, regarding the animal models, route and dose of vaccination, challenge methods, measurement of immune responses and cyst burden have been discussed in the text. Most of these experimental vaccines induce immune responses and have a high degree of protection against parasite infections, increase survival rates and duration and reduce cyst burdens. The data demonstrated that SAG1 antigen has a high potential for use as a vaccine and provided a promising approach for protecting humans and animals against toxoplasmosis.
Collapse
Affiliation(s)
- Abdol Sattar Pagheh
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehdi Sharif
- Department of Parasitology, School of Medicine, Sari Branch, Islamic AZAD University, Sari, Iran
| | - Fatemeh Rezaei
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ehsan Ahmadpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Dodangeh
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Omidian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Hadi Hassannia
- Immunonogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran..
| |
Collapse
|
13
|
Javadi Mamaghani A, Fathollahi A, Spotin A, Ranjbar MM, Barati M, Aghamolaie S, Karimi M, Taghipour N, Ashrafi M, Tabaei SJS. Candidate antigenic epitopes for vaccination and diagnosis strategies of Toxoplasma gondii infection: A review. Microb Pathog 2019; 137:103788. [DOI: 10.1016/j.micpath.2019.103788] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/05/2019] [Accepted: 10/08/2019] [Indexed: 12/28/2022]
|
14
|
Habibi P, Daniell H, Soccol CR, Grossi‐de‐Sa MF. The potential of plant systems to break the HIV-TB link. PLANT BIOTECHNOLOGY JOURNAL 2019; 17:1868-1891. [PMID: 30908823 PMCID: PMC6737023 DOI: 10.1111/pbi.13110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/13/2019] [Accepted: 03/21/2019] [Indexed: 06/09/2023]
Abstract
Tuberculosis (TB) and human immunodeficiency virus (HIV) can place a major burden on healthcare systems and constitute the main challenges of diagnostic and therapeutic programmes. Infection with HIV is the most common cause of Mycobacterium tuberculosis (Mtb), which can accelerate the risk of latent TB reactivation by 20-fold. Similarly, TB is considered the most relevant factor predisposing individuals to HIV infection. Thus, both pathogens can augment one another in a synergetic manner, accelerating the failure of immunological functions and resulting in subsequent death in the absence of treatment. Synergistic approaches involving the treatment of HIV as a tool to combat TB and vice versa are thus required in regions with a high burden of HIV and TB infection. In this context, plant systems are considered a promising approach for combatting HIV and TB in a resource-limited setting because plant-made drugs can be produced efficiently and inexpensively in developing countries and could be shared by the available agricultural infrastructure without the expensive requirement needed for cold chain storage and transportation. Moreover, the use of natural products from medicinal plants can eliminate the concerns associated with antiretroviral therapy (ART) and anti-TB therapy (ATT), including drug interactions, drug-related toxicity and multidrug resistance. In this review, we highlight the potential of plant system as a promising approach for the production of relevant pharmaceuticals for HIV and TB treatment. However, in the cases of HIV and TB, none of the plant-made pharmaceuticals have been approved for clinical use. Limitations in reaching these goals are discussed.
Collapse
Affiliation(s)
- Peyman Habibi
- Department of BiochemistrySchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Bioprocess Engineering and BiotechnologyFederal University of ParanáCuritibaPRBrazil
- Embrapa Genetic Resources and BiotechnologyBrasíliaDFBrazil
| | - Henry Daniell
- Department of BiochemistrySchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Maria Fatima Grossi‐de‐Sa
- Embrapa Genetic Resources and BiotechnologyBrasíliaDFBrazil
- Catholic University of BrasíliaBrasíliaDFBrazil
- Post Graduation Program in BiotechnologyUniversity PotiguarNatalRNBrazil
| |
Collapse
|
15
|
Kaushik H, Deshmukh SK, Solanki AK, Bhatia B, Tiwari A, Garg LC. Immunization with recombinant fusion of LTB and linear epitope (40-62) of epsilon toxin elicits protective immune response against the epsilon toxin of Clostridium perfringens type D. AMB Express 2019; 9:105. [PMID: 31300915 PMCID: PMC6626085 DOI: 10.1186/s13568-019-0824-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/26/2019] [Indexed: 11/10/2022] Open
Abstract
Epsilon toxin (Etx) produced by Clostridium perfringens types B and D, a major causative agent of enterotoxaemia causes significant economic losses to animal industry. Conventional vaccines against these pathogens generally employ formalin-inactivated culture supernatants. However, immunization with the culture supernatant and full length toxin subjects the animal to antigenic load and often have adverse effect due to incomplete inactivation of the toxins. In the present study, an epitope-based vaccine against Clostridium perfringens Etx, comprising 40-62 amino acid residues of the toxin in translational fusion with heat labile enterotoxin B subunit (LTB) of E. coli, was evaluated for its protective potential. The ability of the fusion protein rLTB.Etx40-62 to form pentamers and biologically active holotoxin with LTA of E. coli indicated that the LTB present in the fusion protein retained its biological activity. Antigenicity of both the components in the fusion protein was retained as anti-fusion protein antisera detected both the wild type Etx and LTB in Western blot analysis. Immunization of BALB/c mice with the fusion protein resulted in a significant increase in all isotypes, predominantly IgG1, IgG2a and IgG2b. Anti-fusion protein antisera neutralized the cytotoxicity of epsilon toxin both in vitro and in vivo. Thus, the results demonstrate the potential of rLTB.Etx40-62 as a candidate vaccine against C. perfringens.
Collapse
|
16
|
Loh FK, Nathan S, Chow SC, Fang CM. Vaccination challenges and strategies against long-lived Toxoplasma gondii. Vaccine 2019; 37:3989-4000. [PMID: 31186188 DOI: 10.1016/j.vaccine.2019.05.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/05/2019] [Accepted: 05/21/2019] [Indexed: 01/03/2023]
Abstract
Since the discovery of Toxoplasma gondii in 1908, it is estimated that one-third of the global population has been exposed to this ubiquitous intracellular protozoan. The complex life cycle of T. gondii has enabled itself to overcome stress and transmit easily within a broad host range thus achieving a high seroprevalence worldwide. To date, toxoplasmosis remains one of the most prevalent HIV-associated opportunistic central nervous system infections. This review presents a comprehensive overview of different vaccination approaches ranging from traditional inactivated whole-T. gondii vaccines to the popular DNA vaccines. Extensive discussions are made to highlight the challenges in constructing these vaccines, selecting adjuvants as well as delivery methods, immunisation approaches and developing study models. Herein we also deliberate over the latest and promising enhancement strategies that can address the limitations in developing an effective T. gondii prophylactic vaccine.
Collapse
Affiliation(s)
- Fei-Kean Loh
- Division of Biomedical Sciences, School of Pharmacy, The University of Nottingham Malaysia Campus, 43500 Semenyih, Selangor, Malaysia
| | - Sheila Nathan
- School of Biosciences and Biotechnology, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia
| | - Sek-Chuen Chow
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, The University of Nottingham Malaysia Campus, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
17
|
Hajissa K, Zakaria R, Suppian R, Mohamed Z. Epitope-based vaccine as a universal vaccination strategy against Toxoplasma gondii infection: A mini-review. J Adv Vet Anim Res 2019; 6:174-182. [PMID: 31453188 PMCID: PMC6702889 DOI: 10.5455/javar.2019.f329] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/10/2019] [Accepted: 02/16/2019] [Indexed: 01/18/2023] Open
Abstract
Despite the significant progress in the recent efforts toward developing an effective vaccine against toxoplasmosis, the search for new protective vaccination strategy still remains a challenge and elusive goal because it becomes the appropriate way to prevent the disease. Various experimental approaches in the past few years showed that developing a potential vaccine against the disease can be achievable. The combination of multi-epitopes expressing different stages of the parasite life cycle has become an optimal strategy for acquiring a potent, safe, and effective vaccine. Epitope-based vaccines have gained attention as alternative vaccine candidates due to their ability of inducing protective immune responses. This mini-review highlights the current status and the prospects of Toxoplasma gondii vaccine development along with the application of epitope-based vaccine in the future parasite immunization as a novel under development and evaluation strategy.
Collapse
Affiliation(s)
- Khalid Hajissa
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Robaiza Zakaria
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rapeah Suppian
- Biomedicine Program, School of Health Sciences, Universiti Sains Malaysia 16150 Kubang Kerian, Kelantan, Malaysia
| | - Zeehaida Mohamed
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
18
|
Foroutan M, Ghaffarifar F, Sharifi Z, Dalimi A, Jorjani O. Rhoptry antigens as Toxoplasma gondii vaccine target. Clin Exp Vaccine Res 2019; 8:4-26. [PMID: 30775347 PMCID: PMC6369123 DOI: 10.7774/cevr.2019.8.1.4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/10/2018] [Accepted: 01/14/2019] [Indexed: 01/14/2023] Open
Abstract
Toxoplasmosis is a cosmopolitan zoonotic infection, caused by a unicellular protozoan parasite known as Toxoplasma gondii that belongs to the phylum Apicomplexa. It is estimated that over one-third of the world's population has been exposed and are latently infected with the parasite. In humans, toxoplasmosis is predominantly asymptomatic in immunocompetent persons, while among immunocompromised individuals may be cause severe and progressive complications with poor prognosis. Moreover, seronegative pregnant mothers are other risk groups for acquiring the infection. The life cycle of T. gondii is very complex, indicating the presence of a plurality of antigenic epitopes. Despite of great advances, recognize and construct novel vaccines for prevent and control of toxoplasmosis in both humans and animals is still remains a great challenge for researchers to select potential protein sequences as the ideal antigens. Notably, in several past years, constant efforts of researchers have made considerable advances to elucidate the different aspects of the cell and molecular biology of T. gondii mainly on microneme antigens, dense granule antigens, surface antigens, and rhoptry proteins (ROP). These attempts thereby provided great impetus to the present focus on vaccine development, according to the defined subcellular components of the parasite. Although, currently there is no commercial vaccine for use in humans. Among the main identified T. gondii antigens, ROPs appear as a putative vaccine candidate that are vital for invasion procedure as well as survival within host cells. Overall, it is estimated that they occupy about 1%–30% of the total parasite cell volume. In this review, we have summarized the recent progress of ROP-based vaccine development through various strategies from DNA vaccines, epitope or multi epitope-based vaccines, recombinant protein vaccines to vaccines based on live-attenuated vectors and prime-boost strategies in different mouse models.
Collapse
Affiliation(s)
- Masoud Foroutan
- Abadan School of Medical Sciences, Abadan, Iran.,Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zohreh Sharifi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Abdolhosein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ogholniaz Jorjani
- Laboratory Science Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
19
|
Bioinformatics analysis of ROP8 protein to improve vaccine design against Toxoplasma gondii. INFECTION GENETICS AND EVOLUTION 2018; 62:193-204. [DOI: 10.1016/j.meegid.2018.04.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 01/17/2023]
|
20
|
Yurina V. Live Bacterial Vectors-A Promising DNA Vaccine Delivery System. Med Sci (Basel) 2018; 6:E27. [PMID: 29570602 PMCID: PMC6024733 DOI: 10.3390/medsci6020027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
Vaccination is one of the most successful immunology applications that has considerably improved human health. The DNA vaccine is a new vaccine being developed since the early 1990s. Although the DNA vaccine is promising, no human DNA vaccine has been approved to date. The main problem facing DNA vaccine efficacy is the lack of a DNA vaccine delivery system. Several studies explored this limitation. One of the best DNA vaccine delivery systems uses a live bacterial vector as the carrier. The live bacterial vector induces a robust immune response due to its natural characteristics that are recognized by the immune system. Moreover, the route of administration used by the live bacterial vector is through the mucosal route that beneficially induces both mucosal and systemic immune responses. The mucosal route is not invasive, making the vaccine easy to administer, increasing the patient's acceptance. Lactic acid bacterium is one of the most promising bacteria used as a live bacterial vector. However, some other attenuated pathogenic bacteria, such as Salmonella spp. and Shigella spp., have been used as DNA vaccine carriers. Numerous studies showed that live bacterial vectors are a promising candidate to deliver DNA vaccines.
Collapse
Affiliation(s)
- Valentina Yurina
- Department of Pharmacy, Medical Faculty, Universitas Brawijaya, East Java 65145, Malang, Indonesia.
| |
Collapse
|
21
|
Foroutan M, Ghaffarifar F. Calcium-dependent protein kinases are potential targets for Toxoplasma gondii vaccine. Clin Exp Vaccine Res 2018; 7:24-36. [PMID: 29399577 PMCID: PMC5795042 DOI: 10.7774/cevr.2018.7.1.24] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/03/2018] [Accepted: 01/06/2018] [Indexed: 01/30/2023] Open
Abstract
Toxoplasma gondii belongs to the Apicomplexa phylum that caused a widespread zoonotic infection in wide range of intermediate hosts. Over one-third of the world's population are latently infected with T. gondii and carry it. The complex life cycle of T. gondii indicates the presence of a plurality of antigenic epitopes. During the recent years, continuous efforts of scientists have made precious advances to elucidate the different aspects of the cell and molecular biology of T. gondii. Despite of great progresses, the development of vaccine candidates for preventing of T. gondii infection in men and animals is still remains a challenge. The calcium-dependent protein kinases (CDPKs) belongs to the superfamily of kinases, which restricted to the apicomplexans, ciliates, and plants. It has been documented that they contribute several functions in the life cycle of T. gondii such as gliding motility, cell invasion, and egress as well as some other critical developmental processes. In current paper, we reviewed the recent progress concerning the development of CDPK-based vaccines against acute and chronic T. gondii.
Collapse
Affiliation(s)
- Masoud Foroutan
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
22
|
HAJISSA K, ZAKARIA R, SUPPIAN R, MOHAMED Z. Immunogenicity of Multiepitope Vaccine Candidate against Toxoplasma gondii Infection in BALB/c Mice. IRANIAN JOURNAL OF PARASITOLOGY 2018; 13:215-224. [PMID: 30069205 PMCID: PMC6068360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Toxoplasma gondii is a widely prevalent intracellular protozoan parasite which causes serious clinical and veterinary problems. Development of an effective vaccine for controlling toxoplasmosis is an extremely important aim. In the present study, the protective efficacy of recombinant multiepitope antigen (USM.TOXO1) expressing nine potential epitopes identified from SAG1, GRA2, and GRA7 of Toxoplasma gondii was evaluated in BALB/c mice. METHODS Mice were immunized subcutaneously with three doses of USM.TOXO1 antigen (10 μg/ml). Following the immunization, the IgG antibody, IgG subclass, IFN-γ and IL-4 production were evaluated using ELISA, the study was conducted at Animal Research and Service Center (ARASC), USM Health Campus in 2016. RESULTS Mice immunized with USM.TOXO1 significantly induced a mixed Th1/Th2 response polarized toward the IgG1 antibody isotype. While the cytokine analysis revealed a significant release of IFN-γ cytokines. CONCLUSION USM.TOXO1 is a potential vaccine candidate that elicits strong immunity in BALB/c mice. The proven immunogenicity of the generated antigen can serve as a premise for further use of epitope-based vaccine in the immunoprevention of human and animal toxoplasmosis.
Collapse
Affiliation(s)
- Khalid HAJISSA
- Dept. of Zoology, Faculty of Science and Technology, Omdurman Islamic University, Omdurman, Sudan, Dept. of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Ku-bang Kerian, Kelantan, Malaysia
| | - Robaiza ZAKARIA
- Dept. of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Ku-bang Kerian, Kelantan, Malaysia
| | - Rapeah SUPPIAN
- Biomedicine Program, School of Health Sciences, Universiti Sains Malaysia, 16150 Kuban Kerian, Kelantan, Malaysia
| | - Zeehaida MOHAMED
- Dept. of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Ku-bang Kerian, Kelantan, Malaysia,Correspondence
| |
Collapse
|
23
|
Hajissa K, Zakaria R, Suppian R, Mohamed Z. An evaluation of a recombinant multiepitope based antigen for detection of Toxoplasma gondii specific antibodies. BMC Infect Dis 2017; 17:807. [PMID: 29284420 PMCID: PMC5747131 DOI: 10.1186/s12879-017-2920-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 12/14/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The inefficiency of the current tachyzoite antigen-based serological assays for the serodiagnosis of Toxoplasma gondii infection mandates the need for acquirement of reliable and standard diagnostic reagents. Recently, epitope-based antigens have emerged as an alternative diagnostic marker for the achievement of highly sensitive and specific capture antigens. In this study, the diagnostic utility of a recombinant multiepitope antigen (USM.TOXO1) for the serodiagnosis of human toxoplasmosis was evaluated. METHODS An indirect enzyme-linked immunosorbent assay (ELISA) was developed to evaluate the usefulness of USM.TOXO1 antigen for the detection of IgG antibodies against Toxoplasma gondii in human sera. Whereas the reactivity of the developed antigen against IgM antibody was evaluated by western blot and Dot enzyme immunoassay (dot-EIA) analysis. RESULTS The diagnostic performance of the new antigens in IgG ELISA was achieved at the maximum values of 85.43% and 81.25% for diagnostic sensitivity and specificity respectively. The USM.TOXO1 was also proven to be reactive with anti- T. gondii IgM antibody. CONCLUSIONS This finding makes the USM.TOXO1 antigen an attractive candidate for improving the toxoplasmosis serodiagnosis and demonstrates that multiepitope antigens could be a potential and promising diagnostic marker for the development of high sensitive and accurate assays.
Collapse
Affiliation(s)
- Khalid Hajissa
- Department of Zoology, Faculty of Science and Technology, Omdurman Islamic University, B.O.Box, 382, Omdurman, Sudan
| | - Robaiza Zakaria
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Rapeah Suppian
- Biomedicine Program, School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Zeehaida Mohamed
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
24
|
Wang T, Yin H, Li Y, Zhao L, Sun X, Cong H. Vaccination with recombinant adenovirus expressing multi-stage antigens of Toxoplasma gondii by the mucosal route induces higher systemic cellular and local mucosal immune responses than with other vaccination routes. ACTA ACUST UNITED AC 2017; 24:12. [PMID: 28367800 PMCID: PMC5399536 DOI: 10.1051/parasite/2017013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/17/2017] [Indexed: 01/08/2023]
Abstract
Toxoplasmosis caused by Toxoplasma gondii, an obligate intracellular protozoan, is a cause of congenital disease and abortion in humans and animals. Various vaccination strategies against toxoplasmosis in rodent models have been used in the past few decades; however, effective vaccines remain a challenge. A recombinant adenovirus vaccine expressing ubiquitin-conjugated multi-stage antigen segments (Ad-UMAS) derived from different life-cycle stages of T. gondii was constructed previously. Here, we compared the immune responses and protection effects in vaccination of mice with Ad-UMAS by five vaccination routes including intramuscular (i.m.), intravenous (i.v.), subcutaneous (s.c.), intraoral (i.o.), and intranasal (i.n.). Much higher levels of T. gondii-specific IgG and IgA antibodies were detected in the sera of the intraoral and intranasal vaccination groups on day 49 compared with controls (p < 0.05). The percentages of CD8+ T-cells in mice immunized intranasally and intraorally were larger than in mice immunized intramuscularly (p < 0.05). The highest level of IL-2 and IFN-γ was detected in the group with nasal immunization, and splenocyte proliferation activity was significantly enhanced in mice immunized via the oral and nasal routes. Furthermore, the higher survival rate (50%) and lower cyst numbers observed in the intraoral and intranasal groups all indicate that Ad-UMAS is far more effective in protecting mice against T. gondii infection via the mucosal route. Ad-UMAS could be an effective and safe mucosal candidate vaccine to protect animals and humans against T. gondii infection.
Collapse
Affiliation(s)
- Ting Wang
- Department of Human Parasitology, Shandong University, School of Medicine, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, P.R. China
| | - Huiquan Yin
- Department of Human Parasitology, Shandong University, School of Medicine, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, P.R. China
| | - Yan Li
- Department of Human Parasitology, Shandong University, School of Medicine, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, P.R. China
| | - Lingxiao Zhao
- Shandong Xiehe University, No. 6277 Jiqing Road, Jinan, Shandong 250107, P.R. China
| | - Xiahui Sun
- Department of Human Parasitology, Shandong University, School of Medicine, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, P.R. China
| | - Hua Cong
- Department of Human Parasitology, Shandong University, School of Medicine, No. 44 Wenhuaxi Road, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
25
|
Ducournau C, Nguyen TT, Carpentier R, Lantier I, Germon S, Précausta F, Pisella PJ, Leroux H, Van Langendonck N, Betbeder D, Dimier-Poisson I. Synthetic parasites: a successful mucosal nanoparticle vaccine against Toxoplasma congenital infection in mice. Future Microbiol 2017; 12:393-405. [PMID: 28339296 DOI: 10.2217/fmb-2016-0146] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM Development of protein vaccine to prevent congenital infection is a major public health priority. Our goal is the design of mucosal synthetic pathogen inducing protective immune responses against congenital toxoplasmosis. MATERIALS & METHODS Mice were immunized intranasally, establishing pregnancy and challenging orally. Placental immune response, congenital infection, pup growth, parasitic load rates were studied. RESULTS Pups born to vaccinated infected dams had significantly fewer brain cysts, no intraocular inflammation and normal growth. Protection was associated with a placental cellular Th1 response downregulated by IL-6 and correlated with persistence of vaccine for few hours in the nose before being totally eliminated. CONCLUSION Our vaccine conferred high protection against congenital toxoplasmosis. These results provide support for future studies of other congenital vaccine.
Collapse
Affiliation(s)
- Céline Ducournau
- Immunologie Parasitaire et Vaccinologie, Biothérapies Anti-Infectieuses, Université de Tours-INRA, UMR1282 Infectiologie et Santé Publique, UFR Pharmacie, F-37000 Tours, France
| | - Thi Tl Nguyen
- Immunologie Parasitaire et Vaccinologie, Biothérapies Anti-Infectieuses, Université de Tours-INRA, UMR1282 Infectiologie et Santé Publique, UFR Pharmacie, F-37000 Tours, France
| | - Rodolphe Carpentier
- Centre International de Recherche sur l'Inflammation de Lille LIRIC -UMR 995 Inserm/Université Lille 2/CHRU Lille. Innovation thérapeutique ciblant l'inflammation. Groupe Nanomédecine, Faculté de Médecine, F-59045 Lille Cedex, France.,Université d'Artois, rue du Temple, 62030 ARRAS, France
| | - Isabelle Lantier
- Laboratoire d'Expertise en Infection Animale, INRA-Université de Tours, UMR1282 Infectiologie et Santé Publique, F-37380 Nouzilly, France
| | - Stéphanie Germon
- Immunologie Parasitaire et Vaccinologie, Biothérapies Anti-Infectieuses, Université de Tours-INRA, UMR1282 Infectiologie et Santé Publique, UFR Pharmacie, F-37000 Tours, France
| | - Flavien Précausta
- Immunologie Parasitaire et Vaccinologie, Biothérapies Anti-Infectieuses, Université de Tours-INRA, UMR1282 Infectiologie et Santé Publique, UFR Pharmacie, F-37000 Tours, France
| | - Pierre-Jean Pisella
- Immunologie Parasitaire et Vaccinologie, Biothérapies Anti-Infectieuses, Université de Tours-INRA, UMR1282 Infectiologie et Santé Publique, UFR Pharmacie, F-37000 Tours, France
| | - Hervé Leroux
- Laboratoire d'Expertise en Infection Animale, INRA-Université de Tours, UMR1282 Infectiologie et Santé Publique, F-37380 Nouzilly, France
| | | | - Didier Betbeder
- Centre International de Recherche sur l'Inflammation de Lille LIRIC -UMR 995 Inserm/Université Lille 2/CHRU Lille. Innovation thérapeutique ciblant l'inflammation. Groupe Nanomédecine, Faculté de Médecine, F-59045 Lille Cedex, France.,Université d'Artois, rue du Temple, 62030 ARRAS, France
| | - Isabelle Dimier-Poisson
- Immunologie Parasitaire et Vaccinologie, Biothérapies Anti-Infectieuses, Université de Tours-INRA, UMR1282 Infectiologie et Santé Publique, UFR Pharmacie, F-37000 Tours, France
| |
Collapse
|
26
|
Wang L, Wang X, Bi K, Sun X, Yang J, Gu Y, Huang J, Zhan B, Zhu X. Oral Vaccination with Attenuated Salmonella typhimurium-Delivered TsPmy DNA Vaccine Elicits Protective Immunity against Trichinella spiralis in BALB/c Mice. PLoS Negl Trop Dis 2016; 10:e0004952. [PMID: 27589591 PMCID: PMC5010209 DOI: 10.1371/journal.pntd.0004952] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/03/2016] [Indexed: 02/01/2023] Open
Abstract
Background Our previous studies showed that Trichinella spiralis paramyosin (TsPmy) is an immunomodulatory protein that inhibits complement C1q and C8/C9 to evade host complement attack. Vaccination with recombinant TsPmy protein induced protective immunity against T. spiralis larval challenge. Due to the difficulty in producing TsPmy as a soluble recombinant protein, we prepared a DNA vaccine as an alternative approach in order to elicit a robust immunity against Trichinella infection. Methods and Findings The full-length TsPmy coding DNA was cloned into the eukaryotic expression plasmid pVAX1, and the recombinant pVAX1/TsPmy was transformed into attenuated Salmonella typhimurium strain SL7207. Oral vaccination of mice with this attenuated Salmonella-delivered TsPmy DNA vaccine elicited a significant mucosal sIgA response in the intestine and a systemic IgG antibody response with IgG2a as the predominant subclass. Cytokine analysis also showed a significant increase in the Th1 (IFN-γ, IL-2) and Th2 (IL-4, 5, 6, 10) responses in lymphocytes from the spleen and MLNs of immunized mice upon stimulation with TsPmy protein. The expression of the homing receptors CCR9/CCR10 on antibody secreting B cells may be related to the translocation of IgA-secreted B cells to local intestinal mucosa. The mice immunized with Salmonella-delivered TsPmy DNA vaccine produced a significant 44.8% reduction in adult worm and a 46.6% reduction in muscle larvae after challenge with T. spiralis larvae. Conclusion Our results demonstrated that oral vaccination with TsPmy DNA delivered by live attenuated S. typhimurium elicited a significant local IgA response and a mixed Th1/Th2 immune response that elicited a significant protection against T. spiralis infection in mice. Trichinellosis is one of the most important food-borne parasitic zoonoses, and a serious public health issue worldwide. Developing a vaccine is an alternative approach to control the disease. TsPmy is a paramyosin expressed by Trichinella spiralis to bind and neutralize human complement and a vaccine antigen. We made a DNA vaccine of TsPmy orally delivered by attenuated Salmonella typhimurium that elicited a robust Th1/Th2 and mucosa IgA responses, and protected mice against T. spiralis infection with significant worm reduction against larval challenge. The attenuated Salmonella-delivered TsPmy DNA vaccine provides a feasible and promising approach for controlling trichinellosis in human and domestic animals.
Collapse
Affiliation(s)
- Lei Wang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
- Beijing Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Xiaohuan Wang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Kuo Bi
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Ximeng Sun
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Jing Yang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Yuan Gu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
- * E-mail:
| |
Collapse
|
27
|
Protective immunity against acute toxoplasmosis in BALB/c mice induced by a DNA vaccine encoding Toxoplasma gondii elongation factor 1-alpha. BMC Infect Dis 2015; 15:448. [PMID: 26497908 PMCID: PMC4619988 DOI: 10.1186/s12879-015-1220-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 10/14/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Toxoplasma gondii can infect almost all warm-blood animals including human beings. The high incidence and severe damage that can be caused by T. gondii infection clearly indicates the need for the development of a vaccine. T. gondii elongation factor 1-alpha (TgEF-1α) plays an important role in pathogenesis and host cell invasion for this parasite. The aim of this study was to evaluate the immune protective efficacy of a DNA vaccine encoding TgEF-1α gene against acute T. gondii infection in mice. METHODS A DNA vaccine (pVAX-EF-1α) encoding T. gondii EF-1a (TgEF-1α) gene was constructed and its immune response and protective efficacy against lethal challenge in BALB/c mice were evaluated. RESULTS Mice inoculated with the pVAX-EF-1α vaccine had a high level of specific anti-T. gondii antibodies and produced high levels of IFN-gamma, interleukin (IL)-4, and IL-17. The expression levels of MHC-I and MHC-II molecules as well as the percentages of both CD4(+) and CD8(+) T cells in mice vaccinated with pVAX-EF-1α were significantly increased (p < 0.05), compared with those in all the mice from control groups (blank control, PBS, and pVAXI). Immunization with pVAX-EF-1α significantly (p < 0.05) prolonged mouse survival time to 14.1 ± 1.7 days after challenge infection with the virulent T. gondii RH strain, compared with mice in the control groups which died within 8 days. CONCLUSIONS DNA vaccination with pVAX-EF-1α triggered strong humoral and cellular responses and induced effective protection in mice against acute T. gondii infection, indicating that TgEF-1α is a promising vaccine candidate against acute toxoplasmosis.
Collapse
|
28
|
Zhang NZ, Wang M, Xu Y, Petersen E, Zhu XQ. Recent advances in developing vaccines against Toxoplasma gondii: an update. Expert Rev Vaccines 2015; 14:1609-21. [PMID: 26467840 DOI: 10.1586/14760584.2015.1098539] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Toxoplasma gondii, a significant public health risk, is able to infect almost all warm-blooded animals including humans, and it results in economic losses in production animals. In the last three years, a large number of vaccination experiments have been performed to control T. gondii infection, with the target of limiting the acute infection and reducing or eliminating tissue cysts in the intermediate hosts. In this paper, we summarize the latest results of the veterinary vaccines against T. gondii infection since 2013. Immunization with live-attenuated whole organisms of non-reverting mutants has been shown to induce remarkably potent immune responses associated with control of acute and chronic toxoplasmosis. The non-cyst-forming mutants are promising new tools for the development of veterinary vaccines against T. gondii infection.
Collapse
Affiliation(s)
- Nian-Zhang Zhang
- a State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , PR China
| | - Meng Wang
- a State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , PR China
| | - Ying Xu
- a State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , PR China.,b Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Protozoa Laboratory, College of Veterinary Medicine , China Agricultural University , Beijing , PR China
| | - Eskild Petersen
- c Department of Infectious Diseases, Clinical Institute, Faculty of Health Sciences , Aarhus University , Aarhus , Denmark
| | - Xing-Quan Zhu
- a State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , PR China
| |
Collapse
|
29
|
A Toxoplasma gondii vaccine encoding multistage antigens in conjunction with ubiquitin confers protective immunity to BALB/c mice against parasite infection. Parasit Vectors 2015; 8:498. [PMID: 26420606 PMCID: PMC4588682 DOI: 10.1186/s13071-015-1108-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/22/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is a widely prevalent intracellular parasite which infects almost all warm-blooded animals including humans and causes serious zoonotic toxoplasmosis. DNA vaccines have proved effective in the protection against parasites. However, the problems of weak immunity and inefficient delivery of DNA vaccine remain major issues. Therefore, comprehensive antigens derived from all stages of the parasite, effective adjuvants and delivery systems should be considered in the vaccine construction. METHODS SAG3101-144,ROP18347-396, MIC6288-347, GRA7182-224, MAG158-125, BAG1156-211 and SPA142-200, derived from antigens in tachyzoite, bradyzoite and sporozoite stages of T. gondii were screened based on CD8(+) T cell epitope binding affinity to HLA and H-2. We constructed a recombinant DNA vaccine and an adenovirus vaccine encoding multi-stage antigen of T. gondii linked to ubiquitin molecules and vaccinated BALB/c mice with different strategies. Antibodies, cytokines, splenocytes proliferation, as well as the percentage of CD4(+) and CD8(+) T cells in immunized mouse were analyzed by the Enzyme-Linked Immunosorbent Assays (ELISA), Flow Cytometry (FCM). Protective efficacy was evaluated by challenging immunized mice with type I and type II parasite. RESULTS Our results indicated that the DNA vaccine had the advantage of inducing a stronger humoral response, whereas the adenovirus-vectored vaccine effectively improved the cellular immune response. Priming with DNA vaccine and boosting with adenovirus-vectored vaccine induced Th1-type immune responses with highest levels of IgG2a and secretion of cytokines IL-2 and IFN-γ. Effective protection against type I and type II parasite with an increase in survival rate and a decrease in brain cyst burden was achieved in immunized mice. CONCLUSIONS Priming vaccination with DNA vaccine and boosting with the recombinant adenovirus vaccine encoding ubiquitin conjugated multi-stage antigens of T. gondii was proved to be a potential strategy against the infection of type I and type II parasite.
Collapse
|
30
|
Wang S, Wang Y, Sun X, Zhang Z, Liu T, Gadahi JA, Xu L, Yan R, Song X, Li X. Protective immunity against acute toxoplasmosis in BALB/c mice induced by a DNA vaccine encoding Toxoplasma gondii 10 kDa excretory-secretory antigen (TgESA10). Vet Parasitol 2015; 214:40-8. [PMID: 26421596 DOI: 10.1016/j.vetpar.2015.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/05/2015] [Accepted: 09/11/2015] [Indexed: 12/31/2022]
Abstract
Toxoplasma gondii 10 kDa excretory-secretory antigen (TgESA10) is involved in the early stages of host invasion. The aim of this study was to evaluate the immune protective efficacy of a DNA vaccine encoding TgESA10 gene against acute T. gondii infection in mice. The gene sequence encoding TgESA10 was inserted into the eukaryotic expression vector pVAX I, and the efficacy of intramuscular vaccination of BALB/c mice with pVAX-ESA10 was analyzed. Mice immunized with pVAX-ESA10 elicited high titers of total IgG, IgG1, IgG2a, IgA and IgM antibodies, while IgE showed no changes. Analysis of cytokine profiles revealed significant increases of IFN-γ, IL-4 and IL-17, while no significant changes were detected in TGF-β1. Additionally, we found that pVAX-ESA10 enhanced the activation of CD4(+) and CD8(+) T cells and the expression of MHC-I and MHC-II molecules in spleen in mice. Immunization with pVAX-ESA10 significantly prolonged survival time (14.3 ± 1.7 days) after challenge infection with the virulent T. gondii RH strain, compared with the control groups which died within 8 days. These results suggested that TgESA10 DNA vaccine could trigger strong humoral and cellular responses and induce partial protection against acute toxoplasmosis.
Collapse
Affiliation(s)
- Shuai Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Yujian Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xiaoni Sun
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Zhenchao Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Tingqi Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Javaid Ali Gadahi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Lixin Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Ruofeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xiaokai Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xiangrui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China.
| |
Collapse
|
31
|
Song X, Xu L, Yan R, Huang X, Li X. Construction of Eimeria tenella multi-epitope DNA vaccines and their protective efficacies against experimental infection. Vet Immunol Immunopathol 2015; 166:79-87. [DOI: 10.1016/j.vetimm.2015.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/13/2015] [Accepted: 05/26/2015] [Indexed: 01/12/2023]
|
32
|
Khalili S, Rahbar MR, Dezfulian MH, Jahangiri A. In silico analyses of Wilms׳ tumor protein to designing a novel multi-epitope DNA vaccine against cancer. J Theor Biol 2015; 379:66-78. [DOI: 10.1016/j.jtbi.2015.04.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 02/25/2015] [Accepted: 04/20/2015] [Indexed: 02/06/2023]
|
33
|
Research progress on surface antigen 1 (SAG1) of Toxoplasma gondii. Parasit Vectors 2014; 7:180. [PMID: 24726014 PMCID: PMC3989796 DOI: 10.1186/1756-3305-7-180] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/04/2014] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasitic protozoan that has a wide host range and causes a zoonotic parasitosis called toxoplasmosis. This infection causes significant morbidity, costs for care and loss of productivity and suffering. The most effective measures to minimize this parasite’s harm to patients are prompt diagnosis and treatment and preventing infection. A parasite surface antigen, SAG1, is considered an important antigen for the development of effective diagnostic tests or subunit vaccines. This review covers several aspects of this antigen, including its gene structure, contribution to host invasion, mechanisms of the immune responses and its applications for diagnosis and vaccine development. This significant progress on this antigen provides foundations for further development of more effective and precise approaches to diagnose toxoplasmosis in the clinic, and also have important implications for exploring novel measures to control toxoplasmosis in the near future.
Collapse
|