1
|
Zhang H, Liu Z, Li Y, Tao Z, Shen L, Shang Y, Huang X, Liu Q. Adjuvants for Helicobacter pylori vaccines: Outer membrane vesicles provide an alternative strategy. Virulence 2024; 15:2425773. [PMID: 39501551 PMCID: PMC11583678 DOI: 10.1080/21505594.2024.2425773] [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: 01/29/2024] [Revised: 09/19/2024] [Accepted: 10/31/2024] [Indexed: 11/12/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a gram-negative, spiral-shaped bacterium that colonizes the human stomach, leading to various gastric diseases. The efficacy of traditional treatments, such as bismuth-based triple and quadruple therapies, has been reduced due to increasing antibiotic resistance and drug toxicity. As a result, the development of effective vaccines was proposed to control H. pylori-induced infections; however, one of the primary challenges is the lack of potent adjuvants. Although various adjuvants, both toxic (e.g. cholera toxin and Escherichia coli heat-labile toxin) and non-toxic (e.g. aluminum and propolis), have been tested for vaccine development, no clinically favorable adjuvants have been identified due to high toxicity, weak immunostimulatory effects, inability to elicit specific immune responses, or latent side effects. Outer membrane vesicles (OMVs), mainly secreted by gram-negative bacteria, have emerged as promising candidates for H. pylori vaccine adjuvants due to their potential applications. OMVs enhance mucosal immunity and Th1 and Th17 cell responses, which have been recognized to have protective effects and guarantee safety and efficacy. The development of an effective vaccine against H. pylori infection is ongoing, with clinical trials expected in the future.
Collapse
Affiliation(s)
- Hanchi Zhang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Zhili Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yi Li
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- The First Clinical Medical College, Nanchang University, Nanchang, China
| | - Ziwei Tao
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lu Shen
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yinpan Shang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
2
|
Yin J, Wu H, Li W, Wang Y, Li Y, Mo X, Li S, Ren Y, Pan H, Jiang P, Wang Q. Escherichia coli heat-labile enterotoxin B subunit as an adjuvant of mucosal immune combined with GCRV-II VP6 triggers innate immunity and enhances adaptive immune responses following oral vaccination of grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2024; 154:109969. [PMID: 39419132 DOI: 10.1016/j.fsi.2024.109969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
The grass carp reovirus (GCRV) is the most major pathogen that has threatened the grass carp (Ctenopharyngodon idella) industry of China for years. Though the oral vaccine has many advantages, the current vaccines still do not provide complete protection. Therefor the exploration of new preventive strategies is urgently needed. In this study, heat-labile enterotoxin B subunit of Escherichia coli (LTB) was combined with VP6 from GCRV type II (GCRV-II) via Lactococcus lactis expression system to form a potent oral vaccine and determines if fusion of LTB to the protective vaccine antigen can enhance protection in the fish. The expression of recombinant protein was confirmed by Western-blotting and enzyme-linked immunosorbent assay. The rare minnow was set as the model for the evaluation of the experiment administrated orally. The immune response including the antibody titer and the immune-related gene expression, and the protective efficacy which included the virus loaded and the relative protection, were thoroughly investigated after the trial. The results indicated that LTB can significantly elicit a higher neutralizing antibody responses and enhanced T-cell priming, activities and proliferation in mononuclear cells from intestine, spleen and kidney tissues when compared to the VP6 vaccine alone. Moreover, the combined adjuvant can significantly up-regulate type I interferon signaling in different immune organs, especially the mucosa associated lymphoidtissue which could not be induced by VP6 along, result in the contribution of the improvement in adaptive immune responses of the fish. In addition, challenge study showed that LTB combined VP6 could greatly improve the relative percent survival of the fish during the virus infection. These results highlight that LTB has the potential value to be a mucosal adjuvant of the fish, approaching for improving the efficacy of vaccination against GCRV-II, which does elicit both non-specific and specific immune responses.
Collapse
Affiliation(s)
- Jiyuan Yin
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Huiliang Wu
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Wei Li
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Yingying Wang
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China.
| | - Yingying Li
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Xubing Mo
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Siming Li
- Institute for Quality & Safety and Standards of Agricultural Products Research, Jiangxi Academy of Agricultural Sciences, Nanlian Road 602, Nanchang, 30200, China
| | - Yan Ren
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Houjun Pan
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Peng Jiang
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China
| | - Qing Wang
- Pearl River Fishery Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Fishery Drug Development, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Aquatic Animal Immunology and Sustainable Aquaculture, Guangzhou, 510380, China.
| |
Collapse
|
3
|
Ni X, Liu Y, Sun M, Jiang Y, Wang Y, Ke D, Guo G, Liu K. Oral Live-Carrier Vaccine of Recombinant Lactococcus lactis Inducing Prophylactic Protective Immunity Against Helicobacter pylori Infection. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10360-x. [PMID: 39251521 DOI: 10.1007/s12602-024-10360-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Helicobacter pylori infects the gastric mucosa and induces chronic gastritis, peptic ulcers, and gastric cancer. Research has demonstrated that vaccination can induce a protective immune response and prevent H. pylori infection. Oral administration of the Lactococcus lactis live-carrier vaccine is safe and easily complied with by the public. In this study, two recombinant L. lactis strains were constructed that expressed antigens of H. pylori urease subunit alpha (UreA) and UreA fused with Escherichia coli heat-labile toxin B subunit (LTB-UreA), named LL-UreA and LL-LTB-UreA, respectively. The expression of antigen proteins was confirmed by Western blotting analysis. Survival assessment indicated that the engineered L. lactis could colonize in the digestive tract of BALB/c mice up to 10 days after the last oral administration with our immunization protocol. The ability to induce immune response and immune protective efficacy of the L. lactis were confirmed. These results indicated that oral administration with LL-UreA or LL-LTB-UreA could induce UreA-specific mucosal secretory IgA (sIgA) and cellular immune response, significantly increasing the cytokines levels of interferon-gamma (IFN-γ), interleukin (IL)-17A, and IL-10, together with the proportion of CD4+IFN-γ+ T cells and CD4+IL17A+ T cells. More importantly, oral administration of LL-UreA and LL-LTB-UreA brought about effective protection in mice to prevent H. pylori infection, especially LL-UreA, resulting in 70% of mice showing no H. pylori colonization and the remaining 30% showing only low levels of colonization. These findings underscore the potential of using orally administered engineered L. lactis vaccines to prevent H. pylori infection.
Collapse
Affiliation(s)
- Xiumei Ni
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Yu Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Min Sun
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Yajun Jiang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Yi Wang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Dingxin Ke
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Gang Guo
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China.
| | - Kaiyun Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China.
| |
Collapse
|
4
|
Shi Q, Wang Q, Shen Y, Chen S, Gan S, Lin T, Song F, Ma Y. Escherichia coli LTB26 mutant enhances immune responses to rotavirus antigen VP8 in a mouse model. Mol Immunol 2024; 173:10-19. [PMID: 39004021 DOI: 10.1016/j.molimm.2024.07.001] [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: 06/10/2023] [Revised: 06/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Adjuvant is a major supplementary component of vaccines to boost adaptive immune responses. To select an efficient adjuvant from the heat-labile toxin B subunit (LTB) of E. coli, four LTB mutants (numbered LTB26, LTB34, LTB57, and LTB85) were generated by multi-amino acid random replacement. Mice have been intranasally vaccinated with human rotavirus VP8 admixed. Among the four mutants, enzyme-linked immunosorbent assay (ELISA) revealed that LTB26 had enhanced mucosal immune adjuvanticity compared to LTB, showing significantly enhanced immune responses in both serum IgG and mucosal sIgA levels. The 3D modeling analysis suggested that the enhanced immune adjuvanticity of LTB26 might be due to the change of the first LTB α-helix to a β-sheet. The molecular mechanism was studied using transcriptomic and flow cytometric (FCM) analysis. The transcriptomic data demonstrated that LTB26 enhanced immune response by enhancing B cell receptor (BCR) and major histocompatibility complex (MHC) II+-related pathways. Furthermore, LTB26 promoted Th1 and Th2-type immune responses which were confirmed by detecting IFN-γ and IL-4 expression levels. Immunohistochemical analysis demonstrated that LTB26 enhanced both Th1 and Th2 type immunity. Therefore, LTB26 was a potent mucosal immune adjuvant meeting the requirement for use in human clinics in the future.
Collapse
Affiliation(s)
- Qinlin Shi
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qiujuan Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yanxi Shen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijing Chen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijie Gan
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yongping Ma
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China.
| |
Collapse
|
5
|
de Oliveira NR, Santos FDS, Dos Santos VAC, Maia MAC, Oliveira TL, Dellagostin OA. Challenges and Strategies for Developing Recombinant Vaccines against Leptospirosis: Role of Expression Platforms and Adjuvants in Achieving Protective Efficacy. Pathogens 2023; 12:787. [PMID: 37375478 DOI: 10.3390/pathogens12060787] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
The first leptospiral recombinant vaccine was developed in the late 1990s. Since then, progress in the fields of reverse vaccinology (RV) and structural vaccinology (SV) has significantly improved the identification of novel surface-exposed and conserved vaccine targets. However, developing recombinant vaccines for leptospirosis faces various challenges, including selecting the ideal expression platform or delivery system, assessing immunogenicity, selecting adjuvants, establishing vaccine formulation, demonstrating protective efficacy against lethal disease in homologous challenge, achieving full renal clearance using experimental models, and reproducibility of protective efficacy against heterologous challenge. In this review, we highlight the role of the expression/delivery system employed in studies based on the well-known LipL32 and leptospiral immunoglobulin-like (Lig) proteins, as well as the choice of adjuvants, as key factors to achieving the best vaccine performance in terms of protective efficacy against lethal infection and induction of sterile immunity.
Collapse
Affiliation(s)
- Natasha Rodrigues de Oliveira
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | - Francisco Denis Souza Santos
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | | | - Mara Andrade Colares Maia
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | - Thaís Larré Oliveira
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| | - Odir Antônio Dellagostin
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-610, RS, Brazil
| |
Collapse
|
6
|
Liu D, Zhan Y, Wu X, Qiao H, Zhang Y, Li B. Design, preparation and characterization of octopus-like self-releasing intracellular protein transporter LEB5 based on Escherichia coli heat-labile enterotoxin. Int J Biol Macromol 2023; 237:124172. [PMID: 36966860 DOI: 10.1016/j.ijbiomac.2023.124172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Despite the great potential of protein drugs as intracellular therapeutic agents, the unmet challenge in breaking through the cell membrane barrier and delivering them to intracellular targets remains. Therefore, developing safe and effective delivery vehicles is critical for fundamental biomedical research and clinical applications. In this study, we designed an octopus-like self-releasing intracellular protein transporter, the LEB5, based on the heat-labile enterotoxin. This carrier comprises five identical units, each of which has three main components: a linker, a self-releasing enzyme sensitivity loop, and the LTB transport domain. The LEB5 comprises five purified monomers that self-assemble to create a pentamer with ganglioside GM1 binding capacity. The fluorescent protein EGFP was used as a reporter system to identify the LEB5 features. The high-purity fusion protein ELEB monomer was produced from modified bacteria carrying pET24a(+)-eleb recombinant plasmids. EGFP protein could effectively detach from LEB5 by low dosage trypsin, according to electrophoresis analysis. The transmission electron microscopy results indicate that both LEB5 and ELEB5 pentamers exhibit a relatively regularly spherical shape, and the differential scanning calorimetry measurements further suggest that these proteins possess excellent thermal stability. Fluorescence microscopy revealed that LEB5 translocated EGFP into different cell types. Flow cytometry showed cellular differences in the transport capacity of LEB5. According to the confocal microscopy, fluorescence analysis and western blotting data, EGFP was transferred to the endoplasmic reticulum by the LEB5 carrier, detached from LEB5 by cleavage of the enzyme-sensitive loop, and released into the cytoplasm. Within the dosage range of LEB5 10-80 μg/mL, cell counting kit-8 assay revealed no significant changes in cell viability. These results demonstrated that LEB5 is a safe and effective intracellular self-releasing delivery vehicle capable of transporting and releasing protein medicines into cells.
Collapse
Affiliation(s)
- Di Liu
- College of Biological Sciences and Technology and Center for Veterinary Medicine, Taiyuan Normal University, Jinzhong 030619, Shanxi, China.
| | - Yafen Zhan
- College of Biological Sciences and Technology and Center for Veterinary Medicine, Taiyuan Normal University, Jinzhong 030619, Shanxi, China
| | - Xiaoying Wu
- College of Biological Sciences and Technology and Center for Veterinary Medicine, Taiyuan Normal University, Jinzhong 030619, Shanxi, China
| | - Hongping Qiao
- College of Biological Sciences and Technology and Center for Veterinary Medicine, Taiyuan Normal University, Jinzhong 030619, Shanxi, China
| | - Yeli Zhang
- College of Biological Sciences and Technology and Center for Veterinary Medicine, Taiyuan Normal University, Jinzhong 030619, Shanxi, China
| | - Bo Li
- College of Biological Sciences and Technology and Center for Veterinary Medicine, Taiyuan Normal University, Jinzhong 030619, Shanxi, China; School of Environmental Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China.
| |
Collapse
|
7
|
He X, Chen X, Wang H, Du G, Sun X. Recent advances in respiratory immunization: A focus on COVID-19 vaccines. J Control Release 2023; 355:655-674. [PMID: 36787821 PMCID: PMC9937028 DOI: 10.1016/j.jconrel.2023.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
The development of vaccines has always been an essential task worldwide since vaccines are regarded as powerful weapons in protecting the global population. Although the vast majority of currently authorized human vaccinations are administered intramuscularly or subcutaneously, exploring novel routes of immunization has been a prominent area of study in recent years. This is particularly relevant in the face of pandemic diseases, such as COVID-19, where respiratory immunization offers distinct advantages, such as inducing systemic and mucosal responses to prevent viral infections in both the upper and lower respiratory tracts and also leading to higher patient compliance. However, the development of respiratory vaccines confronts challenges due to the physiological barriers of the respiratory tract, with most of these vaccines still in the research and development stage. In this review, we detail the structure of the respiratory tract and the mechanisms of mucosal immunity, as well as the obstacles to respiratory vaccination. We also examine the considerations necessary in constructing a COVID-19 respiratory vaccine, including the dosage form of the vaccines, potential excipients and mucosal adjuvants, and delivery systems and devices for respiratory vaccines. Finally, we present a comprehensive overview of the COVID-19 respiratory vaccines currently under clinical investigation. We hope this review can provide valuable insights and inspiration for the future development of respiratory vaccinations.
Collapse
Affiliation(s)
- Xiyue He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaoyan Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hairui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
8
|
Neupane DP, Ahn C, Yang YA, Lee GY, Song J. Malnutrition and maternal vaccination against typhoid toxin. PLoS Pathog 2022; 18:e1010731. [PMID: 35960787 PMCID: PMC9401117 DOI: 10.1371/journal.ppat.1010731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/24/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022] Open
Abstract
Children are particularly susceptible to typhoid fever caused by the bacterial pathogen Salmonella Typhi. Typhoid fever is prevalent in developing countries where diets can be less well-balanced. Here, using a murine model, we investigated the role of the macronutrient composition of the diet in maternal vaccination efficacies of two subunit vaccines targeting typhoid toxin: ToxoidVac and PltBVac. We found that maternal vaccinations protected all offspring against a lethal-dose typhoid toxin challenge in a balanced, normal diet (ND) condition, but the declined protection in a malnourished diet (MD) condition was observed in the PltBVac group. Despite the comparable antibody titers in both MD and ND mothers, MD offspring had a significantly lower level of typhoid toxin neutralizing antibodies than their ND counterparts. We observed a lower expression of the neonatal Fc receptor on the yolk sac of MD mothers than in ND mothers, agreeing with the observed lower antibody titers in MD offspring. Protein supplementation to MD diets, but not fat supplementation, increased FcRn expression and protected all MD offspring from the toxin challenge. Similarly, providing additional typhoid toxin-neutralizing antibodies to MD offspring was sufficient to protect all MD offspring from the toxin challenge. These results emphasize the significance of balanced/normal diets for a more effective maternal vaccination transfer to their offspring.
Collapse
Affiliation(s)
- Durga P. Neupane
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Changhwan Ahn
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Yi-An Yang
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Gi Young Lee
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Jeongmin Song
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
9
|
Abstract
AB toxins are protein virulence factors secreted by many bacterial pathogens, contributing to the pathogenicity of the cognate bacteria. AB toxins consist of two functionally distinct components: the enzymatic "A" component for pathogenicity and the receptor-binding "B" component for toxin delivery. Consistently, unlike other virulence factors such as effectors, AB toxins do not require additional systems to deliver them to the target host cells. Target host cells are located in the infection site and/or located distantly from infected host cells. The first part of this review discusses the structural and functional features of single-peptide and multiprotein AB toxins in the context of host-microbe interactions, using several well-characterized examples. The second part of this review discusses toxin neutralization strategies, as well as applications of AB toxins relevant to developing intervention strategies against diseases.
Collapse
Affiliation(s)
- Jeongmin Song
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
10
|
Evaluation of Immunogenicity and Efficacy of Fasciola hepatica Tetraspanin 2 (TSP2) Fused to E. coli Heat-Labile Enterotoxin B Subunit LTB Adjuvant Following Intranasal Vaccination of Cattle. Vaccines (Basel) 2021; 9:vaccines9111213. [PMID: 34835144 PMCID: PMC8623123 DOI: 10.3390/vaccines9111213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 11/16/2022] Open
Abstract
Fasciolosis, caused by the liver flukes Fasciola hepatica and F. gigantica, is an economically important and globally distributed zoonotic disease. Liver fluke infections in livestock cause significant losses in production and are of particular concern to regions where drug resistance is emerging. Antigens of the F. hepatica surface tegument represent promising vaccine candidates for controlling this disease. Tetraspanins are integral tegumental antigens that have shown partial protection as vaccine candidates against other trematode species. The Escherichia coli heat-labile enterotoxin's B subunit (LTB) is a potent mucosal adjuvant capable of inducing an immune response to fused antigens. This study investigates the potential of F. hepatica tetraspanin 2 extracellular loop 2 (rFhTSP2) as a protective vaccine antigen and determines if fusion of FhTSP2 to LTB can enhance protection in cattle. Cattle were immunised subcutaneously with rFhTSP2 mixed in the Freund's adjuvant and intranasally with rLTB-FhTSP2 in saline, accounting for equal molar ratios of tetraspanin in both groups. Vaccination with rFhTSP2 stimulated a strong specific serum IgG response, whereas there was no significant serum IgG response following rLTB-FhTSP2 intranasal vaccination. There was no substantial antigen specific serum IgA generated in all groups across the trial. Contrastingly, after the fluke challenge, a rise in antigen specific saliva IgA was observed in both vaccination groups on Day 42, with the rLTB-FhTSP2 vaccination group showing significant mucosal IgA production at Day 84. However, neither vaccine group showed a significant reduction of fluke burden nor faecal egg output. These results suggest that intranasal vaccination with rLTB-FhTSP2 does elicit a humoral mucosal response but further work is needed to evaluate if mucosal delivery of liver fluke antigens fused to LTB is a viable vaccine strategy.
Collapse
|
11
|
Guerrero Manriquez GG, Tuero I. Adjuvants: friends in vaccine formulations against infectious diseases. Hum Vaccin Immunother 2021; 17:3539-3550. [PMID: 34288795 DOI: 10.1080/21645515.2021.1934354] [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] [Indexed: 12/15/2022] Open
Abstract
Infectious diseases represent a major cause of deaths worldwide. No vaccine or effective treatment exists nowadays, especially against intracellular pathogens. The increase in multiple drug and superbug antibiotic resistance strains, excessive medication, or misuse of drugs has prompted the search for other safe and effective alternatives. Consistent with this, adjuvants (Latin word "adjuvare": "help or aid") co-administered (Exo) in vaccines have emerged as a promising alternative to initiate and boost an innate, downstream signal that led to adaptative immune response. Nowadays, a promising model of strong immunogens and adjuvants at mucosal sites are the microbial bacterial toxins. Other adjuvants that are also used and might successfully replace aluminum salts in combination with nanotechnology are CpG-ODN, poly IC, type I IFNs, mRNA platforms. Therefore, in the present review, we focused to revisit the old to the new adjuvants compounds, the properties that make them friends in vaccine formulations against infectious diseases.
Collapse
Affiliation(s)
| | - I Tuero
- Faculty of Science and Phylosophy, Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
12
|
Zerna G, Spithill TW, Beddoe T. Current Status for Controlling the Overlooked Caprine Fasciolosis. Animals (Basel) 2021; 11:1819. [PMID: 34207215 PMCID: PMC8235714 DOI: 10.3390/ani11061819] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 01/17/2023] Open
Abstract
The disease fasciolosis is caused by the liver flukes Fasciola hepatica and F. gigantica, which infect a wide range of mammals and production livestock, including goats. These flatworm parasites are globally distributed and predicted to cost the livestock industry a now conservative USD 3 billion per year in treatment and lowered on-farm productivity. Infection poses a risk to animal welfare and results in lowered fertility rates and reduced production yields of meat, milk and wool. This zoonotic disease is estimated to infect over 600 million animals and up to 2.4 million humans. Current and future control is threatened with the global emergence of flukes resistant to anthelmintics. Drug resistance calls for immediate on-farm parasite management to ensure treatments are effective and re-infection rates are kept low, while a sustainable long-term control method, such as a vaccine, is being developed. Despite the recent expansion of the goat industry, particularly in developing countries, there are limited studies on goat-focused vaccine control studies and the effectiveness of drug treatments. There is a requirement to collate caprine-specific fasciolosis knowledge. This review will present the current status of liver fluke caprine infections and potential control methods for application in goat farming.
Collapse
Affiliation(s)
| | | | - Travis Beddoe
- Department of Animal, Plant and Soil Sciences and Centre for AgriBioscience, La Trobe University, Bundoora, Victoria 3083, Australia; (G.Z.); (T.W.S.)
| |
Collapse
|
13
|
Immunization with recombinant E rns-LTB fusion protein elicits protective immune responses against bovine viral diarrhea virus. Vet Microbiol 2021; 259:109084. [PMID: 34153721 DOI: 10.1016/j.vetmic.2021.109084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/18/2021] [Indexed: 02/07/2023]
Abstract
Bovine viral diarrhea virus (BVDV), a major infectious pathogen and is associated with major economic losses and significant impact on animal welfare worldwide. Here, recombinant Erns-LTB protein vaccine containing MF59 adjuvant was prepared and assessed using a mouse model. The recombinant plasmid (pET32a-Erns-LTB) was constructed and transformed into BL21 (DE3) cells to produce Erns-LTB protein. The Erns-LTB protein was formulated with MF59 adjuvant, when delivered intraperitoneally in mice, exhibited higher immunogenic and induced superior levels of anti-BVDV IgG compared with the MF59 adjuvanted Erns protein. Importantly, after challenged with different BVDV BJ175170 and BJ1305 isolate strains, mice inoculated with Erns-LTB protein displayed alleviated pathological damage and decreased plasma virus shedding compared with mice inoculated with Erns protein. The enhanced protection from Erns-LTB protein is mediated by T cell immunity and primarily based on CD4+ T helper (Th) and CD8+ cytotoxic T lymphocyte (CTL), these results suggest that Erns-LTB protein has potential to protect against a broad range of BVDV strains thereby providing a novel direction for developing broadly protective vaccines.
Collapse
|
14
|
Li Z, Zhao Y, Li Y, Chen X. Adjuvantation of Influenza Vaccines to Induce Cross-Protective Immunity. Vaccines (Basel) 2021; 9:75. [PMID: 33494477 PMCID: PMC7911902 DOI: 10.3390/vaccines9020075] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/22/2022] Open
Abstract
Influenza poses a huge threat to global public health. Influenza vaccines are the most effective and cost-effective means to control influenza. Current influenza vaccines mainly induce neutralizing antibodies against highly variable globular head of hemagglutinin and lack cross-protection. Vaccine adjuvants have been approved to enhance seasonal influenza vaccine efficacy in the elderly and spare influenza vaccine doses. Clinical studies found that MF59 and AS03-adjuvanted influenza vaccines could induce cross-protective immunity against non-vaccine viral strains. In addition to MF59 and AS03 adjuvants, experimental adjuvants, such as Toll-like receptor agonists, saponin-based adjuvants, cholera toxin and heat-labile enterotoxin-based mucosal adjuvants, and physical adjuvants, are also able to broaden influenza vaccine-induced immune responses against non-vaccine strains. This review focuses on introducing the various types of adjuvants capable of assisting current influenza vaccines to induce cross-protective immunity in preclinical and clinical studies. Mechanisms of licensed MF59 and AS03 adjuvants to induce cross-protective immunity are also introduced. Vaccine adjuvants hold a great promise to adjuvant influenza vaccines to induce cross-protective immunity.
Collapse
Affiliation(s)
| | | | | | - Xinyuan Chen
- Biomedical & Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, RI 02881, USA; (Z.L.); (Y.Z.); (Y.L.)
| |
Collapse
|
15
|
Dos Santos NFB, da Silva LR, Costa FJMD, de Mattos DM, de Carvalho E, Ferreira LCDS, Ferreira RDCC. Immunization with a recombinant BibA surface protein confers immunity and protects mice against group B Streptococcus (GBS) vaginal colonization. Vaccine 2020; 38:5286-5296. [PMID: 32571719 DOI: 10.1016/j.vaccine.2020.05.076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/30/2020] [Accepted: 05/27/2020] [Indexed: 11/29/2022]
Abstract
Streptococcus agalactiae or group B Streptococcus (GBS) is a Gram-positive bacterium divided into ten distinct serotypes that colonizes the vaginal and rectal tracts of approximately 30% of women worldwide. GBS is the leading cause of invasive infection in newborns, causing sepsis, pneumoniae and meningitis. The main strategy to prevent GSB infection in newborns includes the use of intrapartum antibiotic therapy, which does not prevent late-onset diseases and may select resistant bacterial strains. We still do not have a vaccine formulation specific for this pathogen approved for human use. Conserved surface proteins are potential antigens that could be targets for recognition by antibodies and activation of cell opsonization. We used a serotype V GBS (GBS-V)-derived recombinant surface protein, rBibA, and evaluated the potential protective role of the induced antigen-specific antibodies after parenteral or mucosal immunizations in C57BL/6 mice. In vitro and in vivo assays demonstrated that vaccine formulations containing BibA combined with different adjuvants induced serum IgG and/or secreted IgA antibodies, leading to enhanced opsonophagocytosis of GBS-V cells and reduced invasion of epithelial cells. One BibA-based vaccine formulation adjuvanted with a nontoxic derivative of the heat-labile toxin produced by enterotoxigenic Escherichia coli (ETEC) strains was capable of inducing protection against vaginal colonization and lethal parenteral challenge with GBS-V. Serum collected from vaccinated mice conferred passive protection against vaginal colonization in naïve mice challenged with GBS-V. Taken together, the present data demonstrate that the BibA protein is a promising antigen for development of a vaccine to protect against GBS infection.
Collapse
Affiliation(s)
- Nayara Fernanda Barros Dos Santos
- Laboratory of Vaccine Development, Department of Microbiology, Biomedical Science Institute, University of São Paulo, 1374 Prof. Lineu Prestes Avenue, São Paulo, SP 05508-000, Brazil.
| | - Lukas Raposo da Silva
- Laboratory of Vaccine Development, Department of Microbiology, Biomedical Science Institute, University of São Paulo, 1374 Prof. Lineu Prestes Avenue, São Paulo, SP 05508-000, Brazil.
| | - Fagner James Martins Dantas Costa
- Laboratory of Vaccine Development, Department of Microbiology, Biomedical Science Institute, University of São Paulo, 1374 Prof. Lineu Prestes Avenue, São Paulo, SP 05508-000, Brazil.
| | - Daniely Maranhão de Mattos
- Laboratory of Vaccine Development, Department of Microbiology, Biomedical Science Institute, University of São Paulo, 1374 Prof. Lineu Prestes Avenue, São Paulo, SP 05508-000, Brazil.
| | - Enéas de Carvalho
- Laboratory of Molecular Biotechnology I, Biotechnology Center, Butantan Institute, 1500 Vital Brasil Avenue, São Paulo, SP 03178-200, Brazil.
| | - Luís Carlos de Souza Ferreira
- Laboratory of Vaccine Development, Department of Microbiology, Biomedical Science Institute, University of São Paulo, 1374 Prof. Lineu Prestes Avenue, São Paulo, SP 05508-000, Brazil.
| | - Rita de Cássia Café Ferreira
- Laboratory of Vaccine Development, Department of Microbiology, Biomedical Science Institute, University of São Paulo, 1374 Prof. Lineu Prestes Avenue, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
16
|
Different types of adjuvants in prophylactic and therapeutic human papillomavirus vaccines in laboratory animals: a systematic review. Arch Virol 2019; 165:263-284. [PMID: 31802228 DOI: 10.1007/s00705-019-04479-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 10/23/2019] [Indexed: 01/06/2023]
Abstract
Human papillomavirus (HPV) causes cervical carcinoma, which and is the third most common cancer, accounting for 275,000 deaths annually worldwide. Adjuvants have a key role in promotion of vaccine efficacy; therefore, using prophylactic and therapeutic vaccines combined with adjuvant could be of great benefit in prevention and treatment of cervical cancer. There are different types of adjuvants, including MF59TM adjuvants, RNA-based, JY (interleukin2/chitosan), cholera toxin (CT), heat-labile enterotoxin (LT), Freund's adjuvant, alum, SA-4-1BBL, λ-carrageenan (λ-CGN), heat shock proteins (HSPs), juzen-taiho-to (JTT) and hochu-ekki-to (HET), ISCOM and ISCOMATRIX™, very small size proteoliposomes (VSSPs), granulocyte macrophage colony-stimulating factor (GM-CSF), and Toll-like receptors (TLRs). Adjuvants have various functions, especially in therapeutic vaccines, and they lead to an increase in cytotoxic T lymphocytes (CTLs), so they are important in the design of vaccines. Here, we review the currently used adjuvants and their combinations with HPV protein vaccines in order to introduce an appropriate adjuvant for HPV vaccines.
Collapse
|
17
|
Evaluation of the reactogenicity, adjuvanticity and antigenicity of LT(R192G) and LT(R192G/L211A) by intradermal immunization in mice. PLoS One 2019; 14:e0224073. [PMID: 31682624 PMCID: PMC6827915 DOI: 10.1371/journal.pone.0224073] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
The development of an effective subunit vaccine is frequently complicated by the difficulty of eliciting protective immune responses, often requiring the co-administration of an adjuvant. Heat-labile toxin (LT), an enterotoxin expressed by enterotoxigenic E. coli (ETEC) with an AB5 structure similar to cholera toxin, is a strong adjuvant. While the mucosa represents the natural route of exposure to LT and related toxins, the clinical utility of LT and similar adjuvants given by mucosal routes has been limited by toxicity, as well as the association between intranasal delivery of LT and Bell's palsy. Single and double amino acid mutants of LT, LT(R192G)/mLT and LT(R192G/L211A)/dmLT respectively, have been proposed as alternatives to reduce the toxicity associated with the holotoxin. In the present study, we compared mLT and dmLT given via a non-mucosal route (i.e. intradermally) to investigate their adjuvanticity when co-administrated with an enterotoxigenic E. coli vaccine candidate, CfaEB. Antigenicity (i.e. ability to elicit response against LT) and reactogenicity at the injection site were also evaluated. BALB/c mice were immunized by the intradermal route with CfaEB plus increasing doses of either mLT or dmLT (0.01 to 2.5 μg). Both adjuvants induced dose-dependent skin reactogenicity, with dmLT being less reactogenic than mLT. Both adjuvants significantly boosted the anti-CfaE IgG and functional hemagglutination inhibiting (HAI) antibody responses, compared to the antigen alone. In addition to inducing anti-LT responses, even at the lowest dose tested (0.01 μg), the adjuvants also prompted in vitro cytokine responses (IFN-γ, IL-4, IL-5, IL-10 and IL-17) that followed different patterns, depending on the protein used for stimulation (CfaE or LTB) and/or the dose used for immunization. The two LT mutants evaluated here, mLT and dmLT, are potent adjuvants for intradermal immunization and should be further investigated for the intradermal delivery of subunit ETEC vaccines.
Collapse
|
18
|
Carvalho Dos Santos C, Rodriguez D, Kanno Issamu A, Cezar De Cerqueira Leite L, Pereira Nascimento I. Recombinant BCG expressing the LTAK63 adjuvant induces increased early and long-term immune responses against Mycobacteria. Hum Vaccin Immunother 2019; 16:673-683. [PMID: 31665996 PMCID: PMC7227645 DOI: 10.1080/21645515.2019.1669414] [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] [Indexed: 12/26/2022] Open
Abstract
The development of more effective vaccines against Mycobacterium tuberculosis has become a world priority. Previously, we have shown that a recombinant BCG expressing the LTAK63 adjuvant (rBCG-LTAK63) displayed higher protection than BCG against tuberculosis challenge in mice. In order to elucidate the immune effector mechanisms induced by rBCG-LTAK63, we evaluated the immune response before and after challenge. The potential to induce an innate immune response was investigated by intraperitoneal immunization with BCG or rBCG-LTAK63: both displayed increased cellular infiltration in the peritoneum with high numbers of neutrophils at 24 h and macrophages at 7 d. The rBCG-LTAK63-immunized mice displayed increased production of Nitric Oxide at 24 h and Hydrogen Peroxide at 7 d. The number of lymphocytes was higher in the rBCG-LTAK63 group when compared to BCG. Immunophenotyping of lymphocytes showed that rBCG-LTAK63 immunization increased CD4+ and CD8+ T cells. An increased long-term Th1/Th17 cytokine profile was observed 90 d after subcutaneous immunization with rBCG-LTAK63. The evaluation of immune responses at 15 d after challenge showed that rBCG-LTAK63-immunized mice displayed increased TNF-α-secreting CD4+ T cells and multifunctional IL-2+ TNF-α+ CD4+ T cells as compared to BCG-immunized mice. Our results suggest that immunization with rBCG-LTAK63 induces enhanced innate and long-term immune responses as compared to BCG. These results can be correlated with the superior protection induced against TB.
Collapse
Affiliation(s)
- Carina Carvalho Dos Santos
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil.,Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo, Brazil
| | - Dunia Rodriguez
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Alex Kanno Issamu
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil
| | - Luciana Cezar De Cerqueira Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo, Brazil.,Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
19
|
Lei Y, Zhao F, Shao J, Li Y, Li S, Chang H, Zhang Y. Application of built-in adjuvants for epitope-based vaccines. PeerJ 2019; 6:e6185. [PMID: 30656066 PMCID: PMC6336016 DOI: 10.7717/peerj.6185] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Several studies have shown that epitope vaccines exhibit substantial advantages over conventional vaccines. However, epitope vaccines are associated with limited immunity, which can be overcome by conjugating antigenic epitopes with built-in adjuvants (e.g., some carrier proteins or new biomaterials) with special properties, including immunologic specificity, good biosecurity and biocompatibility, and the ability to vastly improve the immune response of epitope vaccines. When designing epitope vaccines, the following types of built-in adjuvants are typically considered: (1) pattern recognition receptor ligands (i.e., toll-like receptors); (2) virus-like particle carrier platforms; (3) bacterial toxin proteins; and (4) novel potential delivery systems (e.g., self-assembled peptide nanoparticles, lipid core peptides, and polymeric or inorganic nanoparticles). This review primarily discusses the current and prospective applications of these built-in adjuvants (i.e., biological carriers) to provide some references for the future design of epitope-based vaccines.
Collapse
Affiliation(s)
- Yao Lei
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Furong Zhao
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Junjun Shao
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yangfan Li
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifang Li
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Huiyun Chang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
20
|
Bernstein DI, Pasetti MF, Brady R, Buskirk AD, Wahid R, Dickey M, Cohen M, Baughman H, El-Khorazaty J, Maier N, Sztein MB, Baqar S, Bourgeois AL. A Phase 1 dose escalating study of double mutant heat-labile toxin LTR192G/L211A (dmLT) from Enterotoxigenic Escherichia coli (ETEC) by sublingual or oral immunization. Vaccine 2018; 37:602-611. [PMID: 30563789 DOI: 10.1016/j.vaccine.2018.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND The public health burden of Enterotoxigenic Escherichia coli (ETEC) is high but no vaccine is specifically approved to prevent ETEC infections. METHODS We performed a Phase 1, dose escalation study (1-50 µg) evaluating the sublingual (SL) delivery of the double mutant heat-labile toxin LTR192G/L211A (dmLT) in 80 healthy adult volunteers. The primary objective was safety and the secondary was the immunogenicity of the dmLT. Subjects received 3 doses of dmLT at days 1, 15, and 29. Subjects receiving the first dose at each dosage level were observed overnight in a research facility. The second and third doses were administered on an outpatient basis. Data from cohorts 1-4 were used to select the cohort 5 dose (25 µg), comparing SL and oral routes. RESULTS The vaccine appeared safe and well tolerated with only rare development of vomiting or diarrhea. The serum anti-dmLT IgA and IgG and neutralizing antibody responses were modest after any of the SL immunizations. Serum IgA and IgG titers were increased at the higher antigen doses (25 or 50 µg) but the percent with 4-fold increases was at best 38% for both IgA and IgG. The 4-fold increase among subjects receiving all 3 doses was 43% for both IgA and IgG. Antibody titers following oral administration were, in general, significantly higher than after SL. The frequency of IgA- or IgG-ASCs in circulation were somewhat vaccine dose dependent and were detected at a moderate level. However, antibodies in saliva or stool were rarely detected. Post-vaccination increases in T cells or cytokine production were also infrequent. CONCLUSION The dmLT vaccine formulation evaluated here was safe but only moderately immunogenic at doses up to 50 µg when administered by the SL or oral route. Studies at higher doses with better formulations appear warranted.
Collapse
Affiliation(s)
- David I Bernstein
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rebecca Brady
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Amanda D Buskirk
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rezwanul Wahid
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Michelle Dickey
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Mitchell Cohen
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | | | | | | | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shahida Baqar
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | |
Collapse
|
21
|
Stubljar D, Jukic T, Ihan A. How far are we from vaccination against Helicobacter pylori infection? Expert Rev Vaccines 2018; 17:935-945. [PMID: 30238819 DOI: 10.1080/14760584.2018.1526680] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Helicobacter pylori infection results in chronic gastritis, peptic ulcer, or gastric cancer; therefore, eradication of this bacterium is essential. The strategy for developing effective vaccines against H. pylori entails immunization of mice with a combination of classical and recombinant H. pylori antigens, but this has proven to be onerous in all cases. AREAS COVERED We have reviewed literature databases in PubMed and Scopus using the key words H. pylori, vaccine, and vaccination and have conducted a systematic review of published clinical trials and animal model studies on vaccines against H. pylori and have tried to summarize why the vaccines are not effective or only partially effective. EXPERT COMMENTARY This is the perfect time to review vaccine development against H. pylori as, after several failed attempts, promising results were reported by Zeng et al. in 2015. Successful vaccine development requires knowledge of both the immune mechanisms active during natural infection by H. pylori, owing to the complicated host response against the pathogen, and the factors that allow the persistence of bacteria, such as genetic diversity of H. pylori. Moreover, various clinical trials are needed to prove vaccine efficacy.
Collapse
Affiliation(s)
- David Stubljar
- a Department of Research & Development , In-Medico , Metlika , Slovenia
| | - Tomislav Jukic
- b Department of Biomedicine and Public Health , Faculty of Medicine Osijek , Osijek , Croatia
| | - Alojz Ihan
- c Medical Faculty of Ljubljana , Institute of Microbiology and Immunology , Ljubljana , Slovenia
| |
Collapse
|
22
|
Ren Z, Zhao Y, Liu J, Ji X, Meng L, Wang T, Sun W, Zhang K, Sang X, Yu Z, Li Y, Feng N, Wang H, Yang S, Yang Z, Wang Z, Gao Y, Xia X. Inclusion of membrane-anchored LTB or flagellin protein in H5N1 virus-like particles enhances protective responses following intramuscular and oral immunization of mice. Vaccine 2018; 36:5990-5998. [PMID: 30172635 DOI: 10.1016/j.vaccine.2018.08.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/10/2018] [Accepted: 08/18/2018] [Indexed: 12/24/2022]
Abstract
We previously demonstrated that intramuscular immunization with virus-like particles (VLPs) composed of the haemagglutinin (HA), neuraminidase (NA), and matrix (M1) proteins of A/meerkat/Shanghai/SH-1/2012 (clade 2.3.2.1) protected mice from lethal challenge with viruses from other H5 HPAI clades. The inclusion of additional proteins that can serve as immunological adjuvants in VLPs may enhance adaptive immune responses following vaccination, and oral vaccines may represent the safest choice. Here, we report the generation of H5N1 VLPs composed of the viral HA, NA, and M1 proteins and membrane-anchored forms of the Escherichia coli heat-labile enterotoxin B subunit protein (LTB) or the Toll-like receptor 5 ligand flagellin (Flic). Mice intramuscularly or orally immunized with VLPs containing LTB or Flic generated greater humoural and cellular immune responses than those administered H5N1 VLPs without LTB or Flic. Intramuscular immunization with VLPs protected mice from lethal challenge with homologous or heterologous H5N1 viruses irrespective of whether the VLPs additionally included LTB or Flic. In contrast, oral immunization of mice with LTB- or Flic-VLPs conferred substantial protection against lethal challenge with both homologous and heterologous H5N1 influenza viruses, whereas mice immunized orally with VLPs lacking LTB and Flic universally succumbed to infection. Mice immunized orally with LTB- or Flic-VLPs showed 10-fold higher virus-specific IgG titres than mice immunized with H5N1-VLPs lacking LTB or Flic. Collectively, these results indicate that the inclusion of immunostimulatory proteins, such as LTB and Flic, in VLP-based vaccines may represent a promising new approach for the control of current H5N1 HPAI outbreaks by eliciting higher humoural and cellular immune responses and conferring improved cross-clade protection.
Collapse
Affiliation(s)
- Zhiguang Ren
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China.
| | - Yongkun Zhao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Jing Liu
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianliang Ji
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Lingnan Meng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Tiecheng Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Weiyang Sun
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Kun Zhang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Xiaoyu Sang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Zhijun Yu
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Yuanguo Li
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Na Feng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Hualei Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Songtao Yang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Zhengyan Yang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China
| | - Zhizeng Wang
- Joint National Laboratory for Antibody Drug Engineering, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China; Key Lab of Cellular and Molecular Immunology, Henan University, School of Basic Medical Sciences, Kaifeng, Henan Province, China
| | - Yuwei Gao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| | - Xianzhu Xia
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| |
Collapse
|
23
|
Lee CH, Hajishengallis G, Connell TD. Dendritic Cell-Mediated Mechanisms Triggered by LT-IIa-B 5, a Mucosal Adjuvant Derived from a Type II Heat-Labile Enterotoxin of Escherichia coli. J Microbiol Biotechnol 2017; 27:709-717. [PMID: 28144014 DOI: 10.4014/jmb.1611.11072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Mucosal tissues are the initial site through which most pathogens invade. As such, vaccines and adjuvants that modulate mucosal immune functions have emerged as important agents for disease prevention. Herein, we investigated the immunomodulatory mechanisms of the B subunit of Escherichia coli heat-labile enterotoxin type IIa (LT-IIa-B5), a potent non-toxic mucosal adjuvant. Alternations in gene expression in response to LT-IIa-B5 were identified using a genome-wide transcriptional microarray that focused on dendritic cells (DC), a type of cell that broadly orchestrates adaptive and innate immune responses. We found that LT-IIa-B5 enhanced the homing capacity of DC into the lymph nodes and selectively regulated transcription of pro-inflammatory cytokines, chemokines, and cytokine receptors. These data are consistent with a model in which directional activation and differentiation of immune cells by LT-IIa-B5 serve as a critical mechanism whereby this potent adjuvant amplifies mucosal immunity to co-administered antigens.
Collapse
Affiliation(s)
- Chang Hoon Lee
- Bio & Drug Discovery Division, Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea.,Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Daejeon 34114, Republic of Korea
| | - George Hajishengallis
- Department of Microbiology, School of Dental Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Terry D Connell
- Department of Microbiology & Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, The Jacob's School of Medicine and Biomedical Sciences, The University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| |
Collapse
|
24
|
Maciel L, Magalhães C, Moraes C, Rosa M, Mendonça M, Moreira A, Dummer L, Finger P, Ferreira M, Leite F, Conceição F. Imunogenicidade da proteína M recombinante de Streptococcus equi subsp. equi coadministrada com um adjuvante molecular. ARQ BRAS MED VET ZOO 2017. [DOI: 10.1590/1678-4162-9288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | | | - M.C. Rosa
- Universidade Federal de Pelotas, Brazil
| | - M. Mendonça
- Universidade Federal Rural de Pernambuco, Brazil
| | | | | | | | | | | | | |
Collapse
|
25
|
Maeda DLNF, Batista MT, Pereira LR, de Jesus Cintra M, Amorim JH, Mathias-Santos C, Pereira SA, Boscardin SB, Silva SDR, Faquim-Mauro EL, Silveira VB, Oliveira DBL, Johnston SA, Ferreira LCDS, Rodrigues JF. Adjuvant-Mediated Epitope Specificity and Enhanced Neutralizing Activity of Antibodies Targeting Dengue Virus Envelope Protein. Front Immunol 2017; 8:1175. [PMID: 28993770 PMCID: PMC5622152 DOI: 10.3389/fimmu.2017.01175] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022] Open
Abstract
The heat-labile toxins (LT) produced by enterotoxigenic Escherichia coli display adjuvant effects to coadministered antigens, leading to enhanced production of serum antibodies. Despite extensive knowledge of the adjuvant properties of LT derivatives, including in vitro-generated non-toxic mutant forms, little is known about the capacity of these adjuvants to modulate the epitope specificity of antibodies directed against antigens. This study characterizes the role of LT and its non-toxic B subunit (LTB) in the modulation of antibody responses to a coadministered antigen, the dengue virus (DENV) envelope glycoprotein domain III (EDIII), which binds to surface receptors and mediates virus entry into host cells. In contrast to non-adjuvanted or alum-adjuvanted formulations, antibodies induced in mice immunized with LT or LTB showed enhanced virus-neutralization effects that were not ascribed to a subclass shift or antigen affinity. Nonetheless, immunosignature analyses revealed that purified LT-adjuvanted EDIII-specific antibodies display distinct epitope-binding patterns with regard to antibodies raised in mice immunized with EDIII or the alum-adjuvanted vaccine. Notably, the analyses led to the identification of a specific EDIII epitope located in the EF to FG loop, which is involved in the entry of DENV into eukaryotic cells. The present results demonstrate that LT and LTB modulate the epitope specificity of antibodies generated after immunization with coadministered antigens that, in the case of EDIII, was associated with the induction of neutralizing antibody responses. These results open perspectives for the more rational development of vaccines with enhanced protective effects against DENV infections.
Collapse
Affiliation(s)
| | - Milene Tavares Batista
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Center for Innovation in Medicine, Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Lennon Ramos Pereira
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana de Jesus Cintra
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jaime Henrique Amorim
- Center of Biological and Health Sciences, Federal University of Western Bahia, Bahia, Brazil
| | - Camila Mathias-Santos
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sara Araújo Pereira
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Vanessa Barbosa Silveira
- Clinical and Molecular Virology Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Danielle Bruna Leal Oliveira
- Clinical and Molecular Virology Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Stephen Albert Johnston
- Center for Innovation in Medicine, Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Luís Carlos de Souza Ferreira
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Juliana Falcão Rodrigues
- Vaccine Development Laboratory, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Feng H, Li X, Song W, Duan M, Chen H, Wang T, Dong J. Oral Administration of a Seed-based Bivalent Rotavirus Vaccine Containing VP6 and NSP4 Induces Specific Immune Responses in Mice. FRONTIERS IN PLANT SCIENCE 2017; 8:910. [PMID: 28620404 PMCID: PMC5449476 DOI: 10.3389/fpls.2017.00910] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/15/2017] [Indexed: 06/07/2023]
Abstract
Rotavirus is the leading cause of severe diarrheal disease among newborns. Plant-based rotavirus vaccines have been developed in recent years and have been proven to be effective in animal models. In the present study, we report a bivalent vaccine candidate expressing rotavirus subunits VP6 and NSP4 fused with the adjuvant subunit B of E. coli heat-labile enterotoxin (LTB) in maize seeds. The RT-PCR and Western blot results showed that VP6 and LTB-NSP4 antigens were expressed and accumulated in maize seeds. The expression levels were as high as 0.35 and 0.20% of the total soluble protein for VP6 and LTB-NSP4, respectively. Oral administration of transgenic maize seeds successfully stimulated systemic and mucosal responses, with high titers of serum IgG and mucosal IgA antibodies, even after long-term storage. This study is the first to use maize seeds as efficient generators for the development of a bivalent vaccine against rotavirus.
Collapse
Affiliation(s)
- Hao Feng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural UniversityBeijing, China
| | - Xin Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural UniversityBeijing, China
| | - Weibin Song
- State Key Laboratory of Agrobiotechnology and National Maize Improvement Center of China, Department of Plant Genetics and Breeding, China Agricultural UniversityBeijing, China
| | - Mei Duan
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural UniversityBeijing, China
| | - Hong Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural UniversityBeijing, China
| | - Tao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural UniversityBeijing, China
| | - Jiangli Dong
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural UniversityBeijing, China
| |
Collapse
|
27
|
Márquez-Escobar VA. Current developments and prospects on human metapneumovirus vaccines. Expert Rev Vaccines 2017; 16:419-431. [PMID: 28116910 DOI: 10.1080/14760584.2017.1283223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Human metapneumovirus (hMPV) has become one of the major pathogens causing acute respiratory infections (ARI) mainly affecting young children, immunocompromised patients, and the elderly. Currently there are no licensed vaccines against this virus. Areas covered: Since the discovery of hMPV in 2001, many groups have focused on developing vaccines against this pathogen. This review presents the outcomes and perspectives derived from preclinical studies performed in cell cultures and animals as well as the only candidate that has reached evaluation in a clinical trial. Limitations of the current vaccine candidates are discussed and perspectives for the development of plant-based vaccines are analyzed. Expert commentary: Several hMPV vaccine candidates are under development with the potential to progress into clinical trials. In parallel, the molecular farming field offers new opportunities to generate innovative vaccines that will offer several advantages in the fight against hMPV.
Collapse
Affiliation(s)
- Verónica Araceli Márquez-Escobar
- a Facultad de Ciencias Químicas , Universidad Autónoma de San Luis Potosí , Av. Dr. Manuel Nava 6, San Luis Potosí 78210 , SLP , Mexico
| |
Collapse
|
28
|
Killingbeck SS, Ge MQ, Haczku A. Patching it together: epicutaneous vaccination with heat-labile Escherichia coli toxin against birch pollen allergy. Allergy 2017; 72:5-8. [PMID: 27716934 PMCID: PMC6279960 DOI: 10.1111/all.13064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - M. Q. Ge
- University of California, Davis, CA, USA
| | - A. Haczku
- University of California, Davis, CA, USA
| |
Collapse
|
29
|
Yu M, Qi R, Chen C, Yin J, Ma S, Shi W, Wu Y, Ge J, Jiang Y, Tang L, Xu Y, Li Y. Immunogenicity of recombinantLactobacillus casei-expressing F4 (K88) fimbrial adhesin FaeG in conjunction with a heat-labile enterotoxin A (LTAK63) and heat-labile enterotoxin B (LTB) of enterotoxigenicEscherichia colias an oral adjuvant in mice. J Appl Microbiol 2016; 122:506-515. [DOI: 10.1111/jam.13352] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/01/2016] [Accepted: 11/13/2016] [Indexed: 12/01/2022]
Affiliation(s)
- M. Yu
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - R. Qi
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - C. Chen
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - J. Yin
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - S. Ma
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - W. Shi
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - Y. Wu
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - J. Ge
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - Y. Jiang
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - L. Tang
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - Y. Xu
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| | - Y. Li
- College of Veterinary Medicine; Northeast Agricultural University; Harbin China
| |
Collapse
|
30
|
Márquez-Escobar VA, Rosales-Mendoza S, Beltrán-López JI, González-Ortega O. Plant-based vaccines against respiratory diseases: current status and future prospects. Expert Rev Vaccines 2016; 16:137-149. [PMID: 27599605 DOI: 10.1080/14760584.2017.1232167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Respiratory infections have an enormous, worldwide epidemiologic impact on humans and animals. Among the prophylactic measures, vaccination has the potential to neutralize this impact. New technologies for vaccine production and delivery are of importance in this field since they offer the potential to develop new immunization approaches overriding the current limitations that comprise high cost, safety issues, and limited efficacy. Areas covered: In the present review, the state of the art in developing plant-based vaccines against respiratory diseases is presented. The review was based on the analysis of current biomedical literature. Expert commentary: Preclinical and clinical evaluations of several vaccine candidates against influenza, tuberculosis, respiratory syncytial virus, pneumonia, anthrax and asthma are discussed and placed in perspective.
Collapse
Affiliation(s)
| | - Sergio Rosales-Mendoza
- a Facultad de Ciencias Químicas , Universidad Autonoma de San Luis Potosi , San Luis Potosi , Mexico
| | - Josué I Beltrán-López
- a Facultad de Ciencias Químicas , Universidad Autonoma de San Luis Potosi , San Luis Potosi , Mexico
| | - Omar González-Ortega
- a Facultad de Ciencias Químicas , Universidad Autonoma de San Luis Potosi , San Luis Potosi , Mexico
| |
Collapse
|
31
|
Quantitative Proteomic Analysis of Escherichia coli Heat-Labile Toxin B Subunit (LTB) with Enterovirus 71 (EV71) Subunit VP1. Int J Mol Sci 2016; 17:ijms17091419. [PMID: 27618897 PMCID: PMC5037698 DOI: 10.3390/ijms17091419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
The nontoxic heat-labile toxin (LT) B subunit (LTB) was used as mucosal adjuvant experimentally. However, the mechanism of LTB adjuvant was still unclear. The LTB and enterovirus 71 (EV71) VP1 subunit (EVP1) were constructed in pET32 and expressed in E. coli BL21, respectively. The immunogenicity of purified EVP1 and the adjuvanticity of LTB were evaluated via intranasal immunization EVP1 plus LTB in Balb/c mice. In order to elucidate the proteome change triggered by the adjuvant of LTB, the proteomic profiles of LTB, EVP1, and LTB plus EVP1 were quantitatively analyzed by iTRAQ-LC-MS/MS (isobaric tags for relative and absolute quantitation; liquid chromatography-tandem mass spectrometry) in murine macrophage RAW264.7. The proteomic data were analyzed by bioinformatics and validated by western blot analysis. The predicted protein interactions were confirmed using LTB pull-down and the LTB processing pathway was validated by confocal microscopy. The results showed that LTB significantly boosted EVP1 specific systematic and mucosal antibodies. A total of 3666 differential proteins were identified in the three groups. Pathway enrichment of proteomic data predicted that LTB upregulated the specific and dominant MAPK (mitogen-activated protein kinase) signaling pathway and the protein processing in endoplasmic reticulum (PPER) pathway, whereas LTB or EVP1 did not significantly upregulate these two signaling pathways. Confocal microscopy and LTB pull-down assays confirmed that the LTB adjuvant was endocytosed and processed through endocytosis (ENS)-lysosomal-endoplasmic reticulum (ER) system.
Collapse
|
32
|
Kazemi R, Akhavian A, Amani J, Salimian J, Motamedi MJ, Mousavi A, Jafari M, Salmanian AH. Immunogenic properties of trivalent recombinant protein composed of B-subunits of LT, STX-2, and CT toxins. Microbes Infect 2016; 18:421-429. [DOI: 10.1016/j.micinf.2016.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 01/31/2023]
|
33
|
Liu D, Guo H, Zheng W, Zhang N, Wang T, Wang P, Ma X. Discovery of the cell-penetrating function of A2 domain derived from LTA subunit of Escherichia coli heat-labile enterotoxin. Appl Microbiol Biotechnol 2016; 100:5079-88. [PMID: 26960316 DOI: 10.1007/s00253-016-7423-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/21/2016] [Accepted: 02/24/2016] [Indexed: 10/22/2022]
Abstract
Heat-labile enterotoxin (LT) is a protein toxin produced by enterotoxigenic Escherichia coli (ETEC). As a bacterial toxin, LT holotoxin can enter intestinal epithelial cells and cause diarrhea. In addition, LT is also a powerful mucosal adjuvant capable of enhancing the strong immune responses to co-administered antigens. However, the LT immunological mechanism is still not clear in some aspects, especially with the respect to how the LTA subunit functions alone. Here, we discovered that the A2 domain of LTA could carry a fluorescent protein into cells, whose function is similar to a cell-penetrating peptide. The transmembrane-transporting ability of the A2 domain is non-specific in its cell-penetrating function, which was shown through testing with different cell types. Moreover, the LTA2 fusion protein penetrated a fluorescently labeled cell membrane that identified LTA2 internalization through membrane transport pathways, and showed it finally localized in the endoplasmic reticulum. Furthermore, low-temperature stress and pharmacological agent treatments showed that the LTA2 internalization route is a temperature-dependent process involving the clathrin-mediated endocytosis and the macropinocytosis pathways. These results could explain the internalization of the LTA subunit alone without the LTB pentamer, contributing to a better understanding of LTA working as a mucosal adjuvant; they also suggest that the A2 domain could be used as a novel transport vehicle for research and treatment of disease.
Collapse
Affiliation(s)
- Di Liu
- School of Biotechnology and State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Hua Guo
- School of Biotechnology and State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Wenyun Zheng
- School of Pharmacy, Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai, China
| | - Na Zhang
- School of Biotechnology and State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Tianwen Wang
- School of Biotechnology and State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Ping Wang
- School of Biotechnology and State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Xingyuan Ma
- School of Biotechnology and State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
34
|
Ma Y. Recent advances in nontoxicEscherichia coliheat-labile toxin and its derivative adjuvants. Expert Rev Vaccines 2016; 15:1361-1371. [DOI: 10.1080/14760584.2016.1182868] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
35
|
A Novel Method for Efficient Preparation of Mucosal Adjuvant Escherichia coli Heat-Labile Enterotoxin Mutant (LTm) by Artificially Assisted Self-Assembly In Vitro. Appl Biochem Biotechnol 2016; 179:33-45. [PMID: 26879977 DOI: 10.1007/s12010-015-1977-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/30/2015] [Indexed: 10/22/2022]
Abstract
As well-known powerful mucosal adjuvant proteins, Escherichia coli heat-labile enterotoxin (LT) and its non-toxic or low-toxic mutants (LTm) are capable of promoting strong mucosal immune responses to co-administered antigens in various types of vaccines. However, due to the complex composition and special structure, the yield of LTm directly from the recombinant genetic engineering strains is quite low. Here, we put forward a novel method to prepare LTm protein which designed, expressed, and purified three kinds of component subunits respectively and assembled them into a hexamer structure in vitro by two combination modes. In addition, by simulated in vivo environment of polymer protein assembly, the factors of the protein solution system which include environment temperature, pH, ionic strength of the solution, and ratio between each subunit were taken into consideration. Finally, we confirmed the optimal conditions of two assembly strategies and prepared the hexamer holotoxin in vitro. These results are not only an important significance in promoting large-scale preparation of the mucosal adjuvant LTm but also an enlightening to produce other multi-subunit proteins.
Collapse
|
36
|
Zhang J, Fan HY, Zhang Z, Zhang J, Zhang J, Huang JN, Ye Y, Liao M. Recombinant baculovirus vaccine containing multiple M2e and adjuvant LTB induces T cell dependent, cross-clade protection against H5N1 influenza virus in mice. Vaccine 2016; 34:622-629. [DOI: 10.1016/j.vaccine.2015.12.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 10/19/2015] [Accepted: 12/15/2015] [Indexed: 12/01/2022]
|
37
|
Zeng M, Mao XH, Li JX, Tong WD, Wang B, Zhang YJ, Guo G, Zhao ZJ, Li L, Wu DL, Lu DS, Tan ZM, Liang HY, Wu C, Li DH, Luo P, Zeng H, Zhang WJ, Zhang JY, Guo BT, Zhu FC, Zou QM. Efficacy, safety, and immunogenicity of an oral recombinant Helicobacter pylori vaccine in children in China: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015; 386:1457-64. [PMID: 26142048 DOI: 10.1016/s0140-6736(15)60310-5] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Helicobacter pylori is one of the most common gastric pathogens, affecting at least half the world's population, and is strongly associated with gastritis, peptic ulcer, gastric adenocarcinoma, and lymphoma. We aimed to assess the efficacy, safety, and immunogenicity of a three-dose oral recombinant H pylori vaccine in children in China. METHODS We did this randomised, double-blind, placebo-controlled, phase 3 trial at one centre in Ganyu County, Jiangsu Province, China. Healthy children aged 6-15 years without past or present H pylori infection were randomly assigned (1:1), via computer-generated randomisation codes in blocks of ten, to receive the H pylori vaccine or placebo. Participants, their guardians, and study investigators were masked to treatment allocation. The primary efficacy endpoint was the occurrence of H pylori infection within 1 year after vaccination. We did analysis in the per-protocol population. This trial is registered with ClinicalTrials.gov, number NCT02302170. FINDINGS Between Dec 2, 2004, and March 19, 2005, we randomly assigned 4464 participants to either the vaccine group (n=2232) or the placebo group (n=2232), of whom 4403 (99%) participants completed the three-dose vaccination schedule and were included in the per-protocol efficacy analysis. We extended follow-up to 3 years. We recorded 64 events of H pylori infection within the first year (14 events in 2074·3 person-years at risk in the vaccine group vs 50 events in 2089·6 person-years at risk in the placebo group), resulting in a vaccine efficacy of 71·8% (95% CI 48·2-85·6). 157 (7%) participants in the vaccine group and 161 (7%) participants in the placebo group reported at least one adverse reaction. Serious adverse events were reported in five (<1%) participants in the vaccine group and seven (<1%) participants in the placebo group, but none was considered to be vaccination related. INTERPRETATION The oral recombinant H pylori vaccine was effective, safe, and immunogenic in H pylori-naive children. This vaccine could substantially reduce the incidence of H pylori infection; however, follow up over a longer period is needed to confirm the protection of the vaccine against H pylori-associated diseases. FUNDING Chongqing Kangwei Biological Technology.
Collapse
Affiliation(s)
- Ming Zeng
- National Institute for Food and Drug Control, Beijing, China
| | - Xu-Hu Mao
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jing-Xin Li
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Wen-De Tong
- Kangwei Biological Technology, Chongqing, China
| | - Bin Wang
- National Institute for Food and Drug Control, Beijing, China
| | - Yi-Ju Zhang
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Gang Guo
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Zhi-Jing Zhao
- National Institute for Food and Drug Control, Beijing, China
| | - Liang Li
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - De-Lin Wu
- Ganyu County Center for Disease Control and Prevention, Lianyungang, Jiangsu Province, China
| | - Dong-Shui Lu
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Zhong-Ming Tan
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China
| | - Hao-Yu Liang
- National Institute for Food and Drug Control, Beijing, China
| | - Chao Wu
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Da-Han Li
- Ganyu County Center for Disease Control and Prevention, Lianyungang, Jiangsu Province, China
| | - Ping Luo
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wei-Jun Zhang
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jin-Yu Zhang
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Bo-Tao Guo
- College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Feng-Cai Zhu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu Province, China.
| | - Quan-Ming Zou
- College of Pharmacy, Third Military Medical University, Chongqing, China.
| |
Collapse
|
38
|
Marciani DJ. Alzheimer's disease vaccine development: A new strategy focusing on immune modulation. J Neuroimmunol 2015; 287:54-63. [PMID: 26439962 DOI: 10.1016/j.jneuroim.2015.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/31/2022]
Abstract
Despite significant advances in the development of Alzheimer's disease (AD) vaccines effective in animal models, these prototypes have been clinically unsuccessful; apparently the result of using immunogens modified to prevent inflammation. Hence, a new paradigm is needed that uses entire AD-associated immunogens, a notion supported by recent successful passive immunotherapy results, with adjuvants that induce Th2-only while inhibiting without abrogating Th1 immunity. Here, we discuss the obstacles to AD vaccine development and Th2-adjuvants that by acting on dendritic and T cells, would elicit regardless of the antigen a safe and effective antibody response, while preventing damaging neuroinflammation and ameliorating immunosenescence.
Collapse
Affiliation(s)
- Dante J Marciani
- Qantu Therapeutics, Inc., 612 E. Main Street, Lewisville, TX 75057, USA.
| |
Collapse
|
39
|
Nizard M, Diniz MO, Roussel H, Tran T, Ferreira LC, Badoual C, Tartour E. Mucosal vaccines: novel strategies and applications for the control of pathogens and tumors at mucosal sites. Hum Vaccin Immunother 2015; 10:2175-87. [PMID: 25424921 DOI: 10.4161/hv.29269] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mucosal immune system displays several adaptations reflecting the exposure to the external environment. The efficient induction of mucosal immune responses also requires specific approaches, such as the use of appropriate administration routes and specific adjuvants and/or delivery systems. In contrast to vaccines delivered via parenteral routes, experimental, and clinical evidences demonstrated that mucosal vaccines can efficiently induce local immune responses to pathogens or tumors located at mucosal sites as well as systemic response. At least in part, such features can be explained by the compartmentalization of mucosal B and T cell populations that play important roles in the modulation of local immune responses. In the present review, we discuss molecular and cellular features of the mucosal immune system as well as novel immunization approaches that may lead to the development of innovative and efficient vaccines targeting pathogens and tumors at different mucosal sites.
Collapse
Affiliation(s)
- Mevyn Nizard
- a INSERM U970; Universite Paris Descartes; Sorbonne Paris-Cité; Paris, France
| | | | | | | | | | | | | |
Collapse
|
40
|
Ji J, Griffiths KL, Milburn PJ, Hirst TR, O'Neill HC. The B subunit of Escherichia coli heat-labile toxin alters the development and antigen-presenting capacity of dendritic cells. J Cell Mol Med 2015; 19:2019-31. [PMID: 26130503 PMCID: PMC4549052 DOI: 10.1111/jcmm.12599] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/25/2015] [Indexed: 02/05/2023] Open
Abstract
Escherichia coli's heat-labile enterotoxin (Etx) and its non-toxic B subunit (EtxB) have been characterized as adjuvants capable of enhancing T cell responses to co-administered antigen. Here, we investigate the direct effect of intravenously administered EtxB on the size of the dendritic and myeloid cell populations in spleen. EtxB treatment appears to enhance the development and turnover of dendritic and myeloid cells from precursors within the spleen. EtxB treatment also gives a dendritic cell (DC) population with higher viability and lower activation status based on the reduced expression of MHC-II, CD80 and CD86. In this respect, the in vivo effect of EtxB differs from that of the highly inflammatory mediator lipopolysaccharide. In in vitro bone marrow cultures, EtxB treatment was also found to enhance the development of DC from precursors dependent on Flt3L. In terms of the in vivo effect of EtxB on CD4 and CD8 T cell responses in mice, the interaction of EtxB directly with DC was demonstrated following conditional depletion of CD11c(+) DC. In summary, all results are consistent with EtxB displaying adjuvant ability by enhancing the turnover of DC in spleen, leading to newly mature myeloid and DC in spleen, thereby increasing DC capacity to perform as antigen-presenting cells on encounter with T cells.
Collapse
Affiliation(s)
- Jing Ji
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Kristin L Griffiths
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Peter J Milburn
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Timothy R Hirst
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Helen C O'Neill
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4229, Australia
| |
Collapse
|
41
|
Bonavita R, Isticato R, Maurano F, Ricca E, Rossi M. Mucosal immunity induced by gliadin-presenting spores of Bacillus subtilis in HLA-DQ8-transgenic mice. Immunol Lett 2015; 165:84-9. [PMID: 25944582 DOI: 10.1016/j.imlet.2015.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/17/2015] [Accepted: 04/22/2015] [Indexed: 11/16/2022]
Abstract
The induction of mucosal immunity requires efficient antigen delivery and adjuvant systems. Probiotic bacterial strains are considered to be very promising tools to address both of these needs. In particular, Bacillus subtilis spores are currently under investigation as a long-lived, protease-resistant adjuvant system for different antigens. Furthermore, a non-recombinant approach has been developed based on the stable adsorption of antigen on the spore surface. In the present study, we explored this strategy as a means of modulating the immune response to wheat gliadin, the triggering agent of celiac disease (CD), an enteropathy driven by inflammatory CD4(+) T cells. Gliadin adsorption was tested on untreated or autoclaved wild-type (wt) and mutant (cotH or cotE) spores. We found that gliadin was stably and maximally adsorbed by autoclaved wt spores. We then tested the immune properties of the spore-adsorbed gliadin in HLA-DQ8-transgenic mice, which express one of the two HLA heterodimers associated with CD. In vitro, spore-adsorbed gliadin was efficiently taken up by mouse dendritic cells (DCs). Interestingly, gliadin-pulsed DCs efficiently stimulated splenic CD4(+) T cells from mice immunised with spore-adsorbed gliadin. Nasal pre-dosing with spore-adsorbed gliadin failed to down-regulate the ongoing cellular response in gliadin-sensitised DQ8 mice. Notably, naïve mice inoculated intranasally with multiple doses of spore-adsorbed gliadin developed an intestinal antigen-specific CD4(+) T cell-mediated response. In conclusion, our data highlight the ability of spore-adsorbed gliadin to elicit a T-cell response in the gut that could be exploitable for developing immune strategies in CD.
Collapse
Affiliation(s)
| | | | | | - Ezio Ricca
- Department of Biology, Federico II University, Naples, Italy
| | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy.
| |
Collapse
|
42
|
Newsted D, Fallahi F, Golshani A, Azizi A. Advances and challenges in mucosal adjuvant technology. Vaccine 2015; 33:2399-405. [PMID: 25865473 DOI: 10.1016/j.vaccine.2015.03.096] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/17/2015] [Accepted: 03/26/2015] [Indexed: 12/23/2022]
Abstract
Adjuvants play attractive roles in enhancement of immune response during vaccination; however, due to several challenges, only a limited number of adjuvants are licensed by health authorities. The lack of an effective mucosal adjuvant is even more significant as none of the licensed adjuvants revealed a strong enhancement in immune system after mucosal administration. Over the past two decades, several mucosal adjuvants have been developed to deliver antigens to the target cells in the mucosal immune system and increase specific immune responses. However, the safety and efficacy of these adjuvants for testing in human trials is still an important issue, requiring further study. In this article, we briefly review the challenges associated with most common mucosal adjuvants and discuss potential strategies for targeting the mucosal immune system.
Collapse
Affiliation(s)
- Daniel Newsted
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, Canada
| | | | - Ashkan Golshani
- Department of Biology, Carleton University, 1125 Colonel by Drive, Ottawa, ON, Canada
| | - Ali Azizi
- Department of Pathology and Laboratory Medicine, University of Ottawa, 451 Smyth Rd, Ottawa, ON, Canada; Department of Biotechnology, University of Ontario Institute of Technology, Toronto, ON, Canada.
| |
Collapse
|
43
|
Li HS, Piao DC, Jiang T, Bok JD, Cho CS, Lee YS, Kang SK, Choi YJ. Recombinant interleukin 6 with M cell-targeting moiety produced in Lactococcus lactis IL1403 as a potent mucosal adjuvant for peroral immunization. Vaccine 2015; 33:1959-67. [DOI: 10.1016/j.vaccine.2015.02.061] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 02/11/2015] [Accepted: 02/19/2015] [Indexed: 12/29/2022]
|
44
|
Rosales-Mendoza S, Ríos-Huerta R, Angulo C. An overview of tuberculosis plant-derived vaccines. Expert Rev Vaccines 2015; 14:877-89. [PMID: 25683476 DOI: 10.1586/14760584.2015.1015996] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tuberculosis (TB) is a leading fatal infectious disease to which the current BCG vaccine has a questionable efficacy in adults. Thus, the development of improved vaccines against TB is needed. In addition, decreasing the cost of vaccine formulations is required for broader vaccination coverage through global vaccination programs. In this regard, the use of plants as biofactories and delivery vehicles of TB vaccines has been researched over the last decade. These studies are systematically analyzed in the present review and placed in perspective. It is considered that substantial preclinical trials are still required to address improvements in expression levels as well as immunological data. Approaches for testing additional antigenic configurations with higher yields and improved immunogenic properties are also discussed.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, México
| | | | | |
Collapse
|
45
|
Salazar-González JA, Bañuelos-Hernández B, Rosales-Mendoza S. Current status of viral expression systems in plants and perspectives for oral vaccines development. PLANT MOLECULAR BIOLOGY 2015; 87:203-17. [PMID: 25560432 DOI: 10.1007/s11103-014-0279-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/22/2014] [Indexed: 05/23/2023]
Abstract
During the last 25 years, the technology to produce recombinant vaccines in plant cells has evolved from modest proofs of the concept to viable technologies adopted by some companies due to significant improvements in the field. Viral-based expression strategies have importantly contributed to this success owing to high yields, short production time (which is in most cases free of tissue culture steps), and the implementation of confined processes for production under GMPs. Herein the distinct expression systems based on viral elements are analyzed. This review also presents the outlook on how these technologies have been successfully applied to the development of plant-based vaccines, some of them being in advanced stages of development. Perspectives on how viral expression systems could allow for the development of innovative oral vaccines constituted by minimally-processed plant biomass are discussed.
Collapse
Affiliation(s)
- Jorge A Salazar-González
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, 78210, San Luis Potosí, SLP, Mexico
| | | | | |
Collapse
|
46
|
Immunogenicity and in vitro and in vivo protective effects of antibodies targeting a recombinant form of the Streptococcus mutans P1 surface protein. Infect Immun 2014; 82:4978-88. [PMID: 25225243 DOI: 10.1128/iai.02074-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Streptococcus mutans is a major etiologic agent of dental caries, a prevalent worldwide infectious disease and a serious public health concern. The surface-localized S. mutans P1 adhesin contributes to tooth colonization and caries formation. P1 is a large (185-kDa) and complex multidomain protein considered a promising target antigen for anticaries vaccines. Previous observations showed that a recombinant P1 fragment (P1(39-512)), produced in Bacillus subtilis and encompassing a functional domain, induces antibodies that recognize the native protein and interfere with S. mutans adhesion in vitro. In the present study, we further investigated the immunological features of P1(39-512) in combination with the following different adjuvants after parenteral administration to mice: alum, a derivative of the heat-labile toxin (LT), and the phase 1 flagellin of S. Typhimurium LT2 (FliCi). Our results demonstrated that recombinant P1(39-512) preserves relevant conformational epitopes as well as salivary agglutinin (SAG)-binding activity. Coadministration of adjuvants enhanced anti-P1 serum antibody responses and affected both epitope specificity and immunoglobulin subclass switching. Importantly, P1(39-512)-specific antibodies raised in mice immunized with adjuvants showed significantly increased inhibition of S. mutans adhesion to SAG, with less of an effect on SAG-mediated bacterial aggregation, an innate defense mechanism. Oral colonization of mice by S. mutans was impaired in the presence of anti-P1(39-512) antibodies, particularly those raised in combination with adjuvants. In conclusion, our results confirm the utility of P1(39-512) as a potential candidate for the development of anticaries vaccines and as a tool for functional studies of S. mutans P1.
Collapse
|
47
|
Marchioro SB, Sácristan RDP, Michiels A, Haesebrouck F, Conceição FR, Dellagostin OA, Maes D. Immune responses of a chimaeric protein vaccine containing Mycoplasma hyopneumoniae antigens and LTB against experimental M. hyopneumoniae infection in pigs. Vaccine 2014; 32:4689-94. [DOI: 10.1016/j.vaccine.2014.05.072] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 05/13/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
|
48
|
Marchioro SB, Fisch A, Gomes CK, Jorge S, Galli V, Haesebrouck F, Maes D, Dellagostin O, Conceição FR. Local and systemic immune responses induced by a recombinant chimeric protein containing Mycoplasma hyopneumoniae antigens fused to the B subunit of Escherichia coli heat-labile enterotoxin LTB. Vet Microbiol 2014; 173:166-71. [PMID: 25091529 DOI: 10.1016/j.vetmic.2014.07.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 11/24/2022]
Abstract
A multi-antigen chimera composed of three antigens of Mycoplasma hyopneumoniae (R1, P42, and NrdF) and the mucosal adjuvant Escherichia coli heat-labile enterotoxin B subunit (LTB) was constructed, and its antigenic and immunogenic properties were evaluated in mice and pigs. In addition, we compared the effect of the fusion and co-administration of these proteins in mice. Antibodies against each subunit recognized the chimeric protein. Intranasal and intramuscular immunization of mice with the chimeric protein significantly increased IgG and IgA levels in the serum and tracheobronchial lavages, respectively, against some of the antigens present in the chimeric. Swine immunized with the chimeric protein developed an immune response against all M. hyopneumoniae antigens present in the fusion with a statistically significant difference (P<0.05). The adjuvant rLTB enhanced the immune response in both fused and co-administered antigens; however, better results were obtained with the chimeric protein. This multi-antigen is a promising vaccine candidate that may help control M. hyopneumoniae infection.
Collapse
Affiliation(s)
- Silvana Beutinger Marchioro
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, RS, Brazil.
| | - Andressa Fisch
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, RS, Brazil
| | - Charles K Gomes
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, RS, Brazil
| | - Sérgio Jorge
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, RS, Brazil
| | - Vanessa Galli
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, RS, Brazil
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Faculty of Veterinary Medicine, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Dominiek Maes
- Unit of Porcine Health Management, Department of Reproduction, Obstetrics and Herd Health, Ghent University, Faculty of Veterinary Medicine, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - Odir Dellagostin
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, RS, Brazil
| | - Fabricio R Conceição
- Núcleo de Biotecnologia, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, RS, Brazil
| |
Collapse
|
49
|
Ying S, He J, Yu M, Zhang Y, Deng S, Zhang L, Xie M, Hu S. Recombinant Neisseria surface protein A is a potential vaccine candidate against Neisseria meningitides serogroup B. Mol Med Rep 2014; 10:1619-25. [PMID: 24926810 DOI: 10.3892/mmr.2014.2325] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 04/14/2014] [Indexed: 11/06/2022] Open
Abstract
Neisseria meningitidis is the pathogen of epidemic encephalomyelitis and is responsible for permanent damage to the brain and nervous system. In the present study, the prokaryotic expression vector pGEX-6p-1/neisseria surface protein A (NspA) was constructed and the immune protective effect was investigated with the purified recombinant rNspA. Female BALB/c mice were immunized by intraperitoneal inoculation of rNspA, glutathione S-transferase (GST) or phosphate-buffered saline (PBS). The protection experiment in mice demonstrated that the protection rate of the rNspA group was 85% against the N. meningitidis strain MC58, and a serum bactericidal assay in vitro revealed that the serum bactericidal titer of the rNspA group reached 1:64 following three immunizations. The levels of specific immunoglobulin (Ig) A (SIgA), IgG, IgG1, IgG2a, IgG2b and IgG3 of mice in the rNspA group peaked at week six and were higher than those in the mice in the GST and PBS groups. The levels of stimulation index, interleukin-4 and interferon-γ in the culture supernatant of the spleen lymphocytes of the rNspA group increased in a time-dependent manner and were higher than those of the mice in the GST and PBS groups over the same period. The results suggested that rNspA may induce increased specific humoral and cellular immune responses, and that it is effectively protective against N. meningitidis serogroup B in mice. The present study offered novel evidence that may lead to the development of a novel effective N. meningitidis serogroup B vaccine.
Collapse
Affiliation(s)
- Shangyun Ying
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jun He
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Minjun Yu
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yukuai Zhang
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Suhong Deng
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lusi Zhang
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Meihua Xie
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Sihai Hu
- Pathogenic Biology Institute, College of Basic Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
50
|
Bagheri S, Mousavi Gargari SL, Rasooli I, Nazarian S, Alerasol M. A CssA, CssB and LTB chimeric protein induces protection against Enterotoxigenic Escherichia coli. Braz J Infect Dis 2014; 18:308-14. [PMID: 24389278 PMCID: PMC9427529 DOI: 10.1016/j.bjid.2013.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 06/29/2013] [Accepted: 07/16/2013] [Indexed: 12/26/2022] Open
Abstract
Objectives Enterotoxigenic Escherichia coli (ETEC), a major cause of diarrhea in children under 5, is an important agent for traveler's diarrhea. Heat-labile enterotoxin (LT) and colonization factors (CFs) are two main virulence mechanisms in ETEC. CS6 is one of the most prevalent CFs consisting of two structural subunits viz., CssA, CssB, necessary for attachment to the intestinal cells. Methods In the present research, a chimeric trivalent protein composed of CssB, CssA and LTB was constructed. The chimeric gene was synthesized with codon bias of E. coli for enhanced expression of the protein. Recombinant proteins were expressed and purified. Mice were immunized with the recombinant protein. The antibody titer and specificity of the immune sera were analyzed by ELISA and Western blotting. Efficiency of the immune sera against ETEC was evaluated. Results Antibody induction was followed by immunization of mice with the chimeric protein. Pretreatment of the ETEC cells with immunized animal antisera remarkably decreased their adhesion to Caco-2 cells. Discussion The results indicate efficacy of the recombinant chimeric protein as an effective immunogen, which induces strong humoral response as well as protection against ETEC adherence and toxicity.
Collapse
|