1
|
Liu B, Li M, Li X, Yang J, Yan H. An optimized caries model of Streptococcus mutans in rats and its application for evaluating prophylactic vaccines. Hum Vaccin Immunother 2024; 20:2345943. [PMID: 38757492 PMCID: PMC11110711 DOI: 10.1080/21645515.2024.2345943] [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: 01/22/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Dental caries is a prevalent oral disease that mainly results from Streptococcus mutans. Susceptibility to S. mutans decreased rapidly after weaning in a well-known rat model. However, owing to the lack of time to establish protective immunity ahead of challenge, the weaning rat model is suboptimal for assessing prophylactic vaccines against S. mutans infection. In this study, we found that, in adult rats, S. mutans cultured under air-restricted conditions showed dramatically increased colonization efficacy and accelerated development of dental caries compared with those cultured under air-unrestricted conditions. We propose that S. mutans cultured under air-restricted conditions can be used to develop an optimal caries model, especially for the evaluation of prophylactic efficacy against S. mutans. Therefore, we used the anti-caries vaccine, KFD2-rPAc, to reevaluate the protection against the challenge of S. mutans. In immunized rats, rPAc-specific protective antibodies were robustly elicited by KFD2-rPAc before the challenge. In addition to inhibiting the initial and long-term colonization of S. mutans in vivo, KFD2-rPAc immunization showed an 83% inhibitory efficacy against the development of caries, similar to that previously evaluated in a weaning rat model. These results demonstrate that culturing under air-restricted conditions can promote S. mutans infection in adult rats, thereby helping establish a rat infection model to evaluate the prophylactic efficacy of vaccines and anti-caries drugs.
Collapse
Affiliation(s)
- Bowen Liu
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Min Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xian Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jingyi Yang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
2
|
Al-Alawi FZM, Kariminik A, Tajbakhsh E. Toll-like receptors and Streptococcus mutans: An updated review article. Allergol Immunopathol (Madr) 2024; 52:79-84. [PMID: 38186197 DOI: 10.15586/aei.v52i1.935] [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: 06/11/2023] [Accepted: 10/10/2023] [Indexed: 01/09/2024]
Abstract
It has been reported that toll-like receptors (TLRs) are the main innate immune receptors that recognize gram-positive pathogen-associated molecular patterns (PAMPs). The molecules can induce expression of the innate immune-related molecules that are essential against the bacteria. Streptococcus mutans (S. mutans) is a potential caries-associated pathogen, and innate immunity plays a key role in inhibiting its development and the progression of inflammatory responses. Recently, the roles played by TLRs against S. mutans and the induction of inflammatory responses were evaluated by several investigations. This review article discusses updated information regarding the roles played by TLRs and their potential therapeutic effects against S. mutans.
Collapse
Affiliation(s)
| | - Ashraf Kariminik
- Department of Microbiology, Kerman Branch, Islamic Azad University, Kerman, Iran;
| | - Elaheh Tajbakhsh
- Department of Microbiology, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
3
|
Yu YB, Liu Y, Liang H, Dong X, Yang XY, Li S, Guo Z. A Nanoparticle-Based Anticaries Vaccine Enhances the Persistent Immune Response To Inhibit Streptococcus mutans and Prevent Caries. Microbiol Spectr 2023; 11:e0432822. [PMID: 36976019 PMCID: PMC10100722 DOI: 10.1128/spectrum.04328-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
Caries vaccines have been identified as a good strategy for the prevention of caries through the mechanism of inoculation against Streptococcus mutans, which is the main etiological bacterium causing caries. Protein antigen c (PAc) of S. mutans has been administered as an anticaries vaccine but shows relatively weak immunogenicity to elicit a low-level immune response. Here, we report a zeolitic imidazolate framework-8 nanoparticle (ZIF-8 NP)-based adjuvant with good biocompatibility, pH responsiveness, and high loading performance for PAc that was used as an anticaries vaccine. In this study, we prepared a ZIF-8@PAc anticaries vaccine and investigated the immune responses and anticaries efficacy induced by this vaccine in vitro and in vivo. ZIF-8 NPs substantially improved the internalization of PAc in lysosomes for further processing and presentation to T lymphocytes. In addition, significantly higher IgG antibody titers, cytokine levels, splenocyte proliferation indices, and percentages of mature dendritic cells (DCs) and central memory T cells were detected in mice subcutaneously immunized with ZIF-8@PAc than in mice subcutaneously immunized with PAc alone. Finally, rats were immunized with ZIF-8@PAc, and ZIF-8@PAc elicited a strong immune response to inhibit colonization by S. mutans and improve prophylactic efficacy against caries. Based on the results, ZIF-8 NPs are promising as an adjuvant for anticaries vaccine development. IMPORTANCE Streptococcus mutans is the main etiologic bacterium of dental caries, whose protein antigen c (PAc) has been administered as an anticaries vaccine. However, the immunogenicity of PAc is relatively weak. To improve the immunogenicity of PAc, ZIF-8 NP was used as an adjuvant, and the immune responses and protective effect elicited by ZIF-8@PAc anticaries vaccine were evaluated in vitro and in vivo. The findings will help in prevention of dental caries and provide new insight for the development of anticaries vaccine in the future.
Collapse
Affiliation(s)
- You-Bo Yu
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong, China
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Ying Liu
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Hangeri Liang
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Xianxin Dong
- Instrumentation and Service Center for Science and Technology, Beijing Normal University, Zhuhai, Guangdong, China
| | - Xiao-Yan Yang
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Sha Li
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Zhong Guo
- Center for Biological Science and Technology, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, Guangdong, China
| |
Collapse
|
4
|
Yu YB, Liu Y, Li S, Yang XY, Guo Z. The pH-responsive zeolitic imidazolate framework nanoparticle as a promising immune-enhancing adjuvant for anti-caries vaccine. J Dent 2023; 130:104413. [PMID: 36634754 DOI: 10.1016/j.jdent.2023.104413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Streptococcus mutans (S. mutans) is the main aetiologic bacterium of dental caries, whose protein antigen (PAc) has been administered as an anti-caries vaccine. In addition, several fusion proteins or PAc combined with adjuvants were used as anti-caries vaccines to improve the relatively weak immunogenicity of PAc. However, there are no nanoparticle-based adjuvants with good biocompatibility, excellent biodegradability, or the high loading performance of antigens used for anti-caries vaccines. This study aimed to prepare an innovative nanoparticle-based anti-caries vaccine and evaluate immune responses elicited by this vaccine in vitro and in vivo. METHODS In this study, an anti-caries vaccine was prepared by an antigen of recombinant protein PAc from S. mutans and an adjuvant of zeolitic imidazolate framework-8 nanoparticles (ZIF-8 NPs) synthesized using a hydrothermal method. Then, mice were administrated intranasally by ZIF-8@PAc vaccine, and immune responses were evaluated. RESULTS ZIF-8 NPs not only greatly improved the internalization of the antigen but also released the PAc protein after degradation of ZIF-8 NPs in lysosomes for the further processing and presentation of antigen-presenting cells. In addition, ZIF-8@PAc induced significantly more potent PAc-specific serum IgG and saliva IgA antibodies, a higher splenocyte proliferation index, higher levels of the cytokines IL-4, IL-6, IL-10, IL-17A and IFN-γ, and a higher percentage of mature DCs and CD4+ memory T cells in vivo. CONCLUSIONS The ZIF-8 NPs, as an anti-caries vaccine adjuvant-assisted antigen PAc, elicit significantly potent immune responses, aiding in the further prevention of dental caries. CLINICAL SIGNIFICANCE Vaccine immunotherapy is an attractive strategy for prevention and treatment of dental caries. The ZIF-8@PAc vaccine can induce significantly high level of immune responses in this study, which indicates great potential for prevention and treatment of caries.
Collapse
Affiliation(s)
- You-Bo Yu
- Center for Biological Science and Technology, Guangdong Zhuhai-Macao Joint Biotech Laboratory, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China; Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Ying Liu
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Sha Li
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Xiao-Yan Yang
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Zhong Guo
- Center for Biological Science and Technology, Guangdong Zhuhai-Macao Joint Biotech Laboratory, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
5
|
Jiang W, Shi L, Cai L, Wang X, Li J, Li H, Liang J, Gu Q, Ji G, Li J, Liu L, Sun M. A two-adjuvant multiantigen candidate vaccine induces superior protective immune responses against SARS-CoV-2 challenge. Cell Rep 2021; 37:110112. [PMID: 34863353 PMCID: PMC8610932 DOI: 10.1016/j.celrep.2021.110112] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/06/2021] [Accepted: 11/18/2021] [Indexed: 01/22/2023] Open
Abstract
An ideal vaccine against SARS-CoV-2 is expected to elicit broad immunity to prevent viral infection and disease, with efficient viral clearance in the upper respiratory tract (URT). Here, the N protein and prefusion-full S protein (SFLmut) are combined with flagellin (KF) and cyclic GMP-AMP (cGAMP) to generate a candidate vaccine, and this vaccine elicits stronger systemic and mucosal humoral immunity than vaccines containing other forms of the S protein. Furthermore, the candidate vaccine administered via intranasal route can enhance local immune responses in the respiratory tract. Importantly, human ACE2 transgenic mice given the candidate vaccine are protected against lethal SARS-CoV-2 challenge, with superior protection in the URT compared with that in mice immunized with an inactivated vaccine. In summary, the developed vaccine can elicit a multifaceted immune response and induce robust viral clearance in the URT, which makes it a potential vaccine for preventing disease and infection of SARS-CoV-2.
Collapse
Affiliation(s)
- Wenwen Jiang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Li Shi
- Laboratory of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Lukui Cai
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Xiaoyu Wang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Jingyan Li
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Heng Li
- Laboratory of Respiratory Infection, Kunming National High-level Biosafety Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Jiangli Liang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Qin Gu
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Guang Ji
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Jing Li
- Laboratory of Respiratory Infection, Kunming National High-level Biosafety Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China
| | - Longding Liu
- Laboratory of Respiratory Infection, Kunming National High-level Biosafety Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China.
| | - Mingbo Sun
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China; Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, Yunnan, China.
| |
Collapse
|
6
|
Zhao B, Yang J, He B, Li X, Yan H, Liu S, Yang Y, Zhou D, Liu B, Fan X, Zhong M, Zhang E, Zhang F, Zhang Y, Chen YQ, Jiang S, Yan H. A safe and effective mucosal RSV vaccine in mice consisting of RSV phosphoprotein and flagellin variant. Cell Rep 2021; 36:109401. [PMID: 34289371 DOI: 10.1016/j.celrep.2021.109401] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/03/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of serious acute lower respiratory tract infection in infants and the elderly. The lack of a licensed RSV vaccine calls for the development of vaccines with other targets and vaccination strategies. Here, we construct a recombinant protein, designated P-KFD1, comprising RSV phosphoprotein (P) and the E.-coli-K12-strain-derived flagellin variant KFD1. Intranasal immunization with P-KFD1 inhibits RSV replication in the upper and lower respiratory tract and protects mice against lung disease without vaccine-enhanced disease (VED). The P-specific CD4+ T cells provoked by P-KFD1 intranasal (i.n.) immunization either reside in or migrate to the respiratory tract and mediate protection against RSV infection. Single-cell RNA sequencing (scRNA-seq) and carboxyfluorescein succinimidyl ester (CFSE)-labeled cell transfer further characterize the Th1 and Th17 responses induced by P-KFD1. Finally, we find that anti-viral protection depends on either interferon-γ (IFN-γ) or interleukin-17A (IL-17A). Collectively, P-KFD1 is a promising safe and effective mucosal vaccine candidate for the prevention of RSV infection.
Collapse
Affiliation(s)
- Bali Zhao
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jingyi Yang
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Bing He
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Xian Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Hu Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shuning Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Dihan Zhou
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Bowen Liu
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xuxu Fan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Maohua Zhong
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ejuan Zhang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fan Zhang
- The Core Facility and Technical Support, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yue Zhang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yao-Qing Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Shibo Jiang
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Huimin Yan
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
7
|
The Combinations Chitosan-Pam 3CSK 4 and Chitosan-Monophosphoryl Lipid A: Promising Immune-Enhancing Adjuvants for Anticaries Vaccine PAc. Infect Immun 2019; 87:IAI.00651-19. [PMID: 31527122 DOI: 10.1128/iai.00651-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 09/02/2019] [Indexed: 11/20/2022] Open
Abstract
We previously demonstrated that recombinant protein PAc could be administered as an anticaries vaccine. However, the relatively weak immunogenicity of PAc limits its application. In the present study, we investigated the effect of two adjuvant combinations of chitosan plus Pam3CSK4 (chitosan-Pam3CSK4) and of chitosan plus monophosphoryl lipid A (chitosan-MPL) in the immune responses to the PAc protein in vivo and in vitro PAc-chitosan-Pam3CSK4 or PAc-chitosan-MPL promoted significantly higher PAc-specific antibody titers in serum and saliva, inhibited Streptococcus mutans colonization onto the tooth surfaces, and endowed better protection effect with significantly less caries activities than PAc alone. Chitosan-Pam3CSK4 and chitosan-MPL showed no statistically significant differences. In conclusion, our study demonstrated that the chitosan-Pam3CSK4 and chitosan-MPL combinations are promising for anticaries vaccine development.
Collapse
|
8
|
Patel M. Dental caries vaccine: are we there yet? Lett Appl Microbiol 2019; 70:2-12. [PMID: 31518435 DOI: 10.1111/lam.13218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/21/2022]
Abstract
Dental caries, caused by Streptococcus mutans, is a common infection. Caries vaccine has been under investigation for the last 40 years. Many in vitro and in vivo studies and some human clinical trials have determined many pertinent aspects regarding vaccine development. The virulence determinants of Strep. mutans, such as Ag I/II, responsible for adherence to surfaces, glucosyltransferase, responsible for the production of glucan, and the glucan-binding protein, responsible for the attachment of glucan to surfaces, have been known to elicit an antigen-specific immune response. It is also known that more than one antigen or a functional part of the genome responsible for these virulence determinants provide a better host response compared with the monogenic vaccine or complete genome of a specific antigen. To enhance the host response, the use of adjuvants has been studied and the routes of antigen administration have been investigated. In recent years, some promising vaccines such as pGJA-P/VAX, LT derivative/Pi39-512 , KFD2-rPAc and SBR/GBR-CMV-nirB have been developed and tested in animals. New virulence targets need to be explored. Multicentre collaborative studies and human clinical trials are required and some interest from funders and public health experts should be generated to overcome this hurdle. SIGNIFICANCE AND IMPACT OF THE STUDY: Dental caries is an irreversible, multifactorial opportunistic infection. The treatment is costly, making it a public health problem. Despite many years of promising laboratory research, animal studies and clinical trials, there is no commercially available vaccine today. The research objectives have become more refined from lessons learnt over the years. Multigenic DNA/recombinant vaccines, using the best proved adjuvants with a delivery system for the nasal or sublingual route, should be developed and researched with multicentre collaborative efforts. In addition, new vaccine targets can be identified. To overcome the economic hurdle, funders and public health interest should be stimulated.
Collapse
Affiliation(s)
- M Patel
- Department of Oral Biological Sciences, School of Oral Health Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
9
|
Bai G, Tian Y, Wu J, Gu Y, Chen Z, Zeng F, Liu J. Construction of a fusion anti-caries DNA vaccine in transgenic tomato plants for PAcA gene and cholera toxin B subunit. Biotechnol Appl Biochem 2019; 66:924-929. [PMID: 31434162 DOI: 10.1002/bab.1806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/20/2019] [Indexed: 11/10/2022]
Abstract
Chronic bacterial infections in the oral cavity influence the development of dental caries. Mutans streptococci are the major pathogenic cause of dental caries. The World Health Organization (WHO) ranks dental caries, cancer, and cardiovascular diseases as the three major global diseases that need urgent preventative and curative measures. However, substantial evidence suggests that traditional prevention and treatment strategies are inefficient in reducing the prevalence of dental caries. For protection against caries, it is important to develop effective vaccines that induce anticolonizing immunity against Streptococcus mutans infections. In the present investigation, we constructed a fusion anti-caries DNA vaccine (PAcA-ctxB) through fusing A region of cell surface protein PAc (PAcA) coding gene of mutans streptococci with cholera toxin B subunit coding gene (CTB). Afterward, the plasmids were integrated into tomato genomes through agrobacterium-mediated plant transformation technology. The presence of transgenes in the tomato genome was confirmed by PCR, β-glucuronidase gene (GUS), and western blot. The expression of genes was confirmed at transcription and protein level. Altogether, the results presented herein showed that transgenic tomatoes may provide a useful system for the production of human caries antigen.
Collapse
Affiliation(s)
- Guohui Bai
- Special Key Laboratory of Oral Diseases Research, Higher Education Institution in Guizhou Province, Zunyi Medical University, Zunyi, People's Republic of China
| | - Yuan Tian
- Special Key Laboratory of Oral Diseases Research, Higher Education Institution in Guizhou Province, Zunyi Medical University, Zunyi, People's Republic of China.,Hospital of Stomatology, Zunyi Medical University, Zunyi, People's Republic of China
| | - Jiayuan Wu
- Special Key Laboratory of Oral Diseases Research, Higher Education Institution in Guizhou Province, Zunyi Medical University, Zunyi, People's Republic of China.,Hospital of Stomatology, Zunyi Medical University, Zunyi, People's Republic of China
| | - Yu Gu
- Special Key Laboratory of Oral Diseases Research, Higher Education Institution in Guizhou Province, Zunyi Medical University, Zunyi, People's Republic of China
| | - Zhu Chen
- Guiyang Hospital of Stomatology, Guiyang, People's Republic of China
| | - Fengjiao Zeng
- Special Key Laboratory of Oral Diseases Research, Higher Education Institution in Guizhou Province, Zunyi Medical University, Zunyi, People's Republic of China.,Hospital of Stomatology, Zunyi Medical University, Zunyi, People's Republic of China
| | - Jianguo Liu
- Special Key Laboratory of Oral Diseases Research, Higher Education Institution in Guizhou Province, Zunyi Medical University, Zunyi, People's Republic of China
| |
Collapse
|
10
|
Zhang T, Chen X, Liu H, Bao Q, Wang Z, Liao G, Xu X. A rationally designed flagellin-L2 fusion protein induced serum and mucosal neutralizing antibodies against multiple HPV types. Vaccine 2019; 37:4022-4030. [PMID: 31213378 DOI: 10.1016/j.vaccine.2019.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/12/2019] [Accepted: 06/03/2019] [Indexed: 12/27/2022]
Abstract
The amino terminus of human papillomavirus (HPV) minor capsid protein L2 harbors several conserved neutralizing epitopes, including aa.17-36 (RG-1 epitope) and aa.65-85 consensus epitope (cL2 epitope), which are considered to be promising for the construction of cost-effective pan-HPV vaccine candidates. However, the immunogenicity of L2 epitope/peptide is rather weak, and the neutralizing spectrum induced by single type of L2 antigen is suboptimal. In this study, we constructed L2 concatemer with HPV18/33/58/59 RG-1 epitopes and 16L2 aa.11-88 peptide, and fused it with flagellin, a strong systemic and mucosal adjuvant, by hypervariable region replacement. A copy of cL2 epitope was also introduced to the C-terminus of the recombinant protein. The resultant Fla-5PcL2 protein can be produced in E. coli expression system with high yield and good stability. We assessed the immunogenicity of Fla-5PcL2 in mouse model via systemic and mucosal route, and found that subcutaneous immunization with Fla-5PcL2 induced robust serum neutralizing antibodies against divergent HPV types, while intranasal immunization with Fla-5PcL2 induced remarkable L2-specific IgA and cross-neutralizing antibodies in mucosal secretions, and medium titers of cross-neutralizing antibodies in sera. Moreover, Fla-5PcL2 induced full protection against vaginal HPV challenges. As mucosal antibodies provide the first-line defense at infection sites, and needle-free immunizations may increase vaccine compliance and require less public health resources, our results demonstrate that Fla-5PcL2 is a promising vaccine candidate which possibly meet the need in low-resource regions.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xue Chen
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Hongyang Liu
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Qifeng Bao
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Zhirong Wang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Guoyang Liao
- The Fifth Department of Biological Products, Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Yunnan, China.
| | - Xuemei Xu
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| |
Collapse
|
11
|
Barnowski C, Kadzioch N, Damm D, Yan H, Temchura V. Advantages and Limitations of Integrated Flagellin Adjuvants for HIV-Based Nanoparticle B-Cell Vaccines. Pharmaceutics 2019; 11:E204. [PMID: 31052410 PMCID: PMC6572692 DOI: 10.3390/pharmaceutics11050204] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 11/29/2022] Open
Abstract
The great advantage of virus-like particle (VLP) nano-vaccines is their structural identity to wild-type viruses, ensuring that antigen-specific B-cells encounter viral proteins in their natural conformation. "Wild-type" viral nanoparticles can be further genetically or biochemically functionalized with biomolecules (antigens and adjuvants). Flagellin is a potent inducer of innate immunity and it has demonstrated adjuvant effectiveness due to its affinity for toll-like receptor 5 (TLR5). In contrast to most TLR ligands, flagellin is a protein and can induce an immune response against itself. To avoid side-effects, we incorporated a less inflammatory and less immunogenic form of flagellin as an adjuvant into HIV-based nanoparticle B-cell-targeting vaccines that display either the HIV-1 envelope protein (Env) or a model antigen, hen egg lysozyme (HEL). While flagellin significantly enhanced HEL-specific IgG responses, anti-Env antibody responses were suppressed. We demonstrated that flagellin did not activate B-cells directly in vitro, but might compete for CD4+ T-cell help in vivo. Therefore, we hypothesize that in the context of VLP-based B-cell nano-vaccines, flagellin serves as an antigen itself and may outcompete a less immunogenic antigen with its antibody response. In contrast, in combination with a strong immunogen, the adjuvant activity of flagellin may dominate over its immunogenicity.
Collapse
Affiliation(s)
- Cornelia Barnowski
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
- Institute of Virology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| | - Nicole Kadzioch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
- Division of Experimental Clinical Research, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland.
| | - Dominik Damm
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Vladimir Temchura
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
12
|
Flores-Langarica A, Cook C, Müller Luda K, Persson EK, Marshall JL, Beristain-Covarrubias N, Yam-Puc JC, Dahlgren M, Persson JJ, Uematsu S, Akira S, Henderson IR, Lindbom BJ, Agace W, Cunningham AF. Intestinal CD103 +CD11b + cDC2 Conventional Dendritic Cells Are Required for Primary CD4 + T and B Cell Responses to Soluble Flagellin. Front Immunol 2018; 9:2409. [PMID: 30386346 PMCID: PMC6199373 DOI: 10.3389/fimmu.2018.02409] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Systemic immunization with soluble flagellin (sFliC) from Salmonella Typhimurium induces mucosal responses, offering potential as an adjuvant platform for vaccines. Moreover, this engagement of mucosal immunity is necessary for optimal systemic immunity, demonstrating an interaction between these two semi-autonomous immune systems. Although TLR5 and CD103+CD11b+ cDC2 contribute to this process, the relationship between these is unclear in the early activation of CD4+ T cells and the development of antigen-specific B cell responses. In this work, we use TLR5-deficient mice and CD11c-cre.Irf4 fl/fl mice (which have reduced numbers of cDC2, particularly intestinal CD103+CD11b+ cDCs), to address these points by studying the responses concurrently in the spleen and the mesenteric lymph nodes (MLN). We show that CD103+CD11b+ cDC2 respond rapidly and accumulate in the MLN after immunization with sFliC in a TLR5-dependent manner. Furthermore, we identify that whilst CD103+CD11b+ cDC2 are essential for the induction of primary T and B cell responses in the mucosa, they do not play such a central role for the induction of these responses in the spleen. Additionally, we show the involvement of CD103+CD11b+ cDC2 in the induction of Th2-associated responses. CD11c-cre.Irf4 fl/fl mice showed a reduced primary FliC-specific Th2-associated IgG1 responses, but enhanced Th1-associated IgG2c responses. These data expand our current understanding of the mucosal immune responses promoted by sFliC and highlights the potential of this adjuvant for vaccine usage by taking advantage of the functionality of mucosal CD103+CD11b+ cDC2.
Collapse
Affiliation(s)
- Adriana Flores-Langarica
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Charlotte Cook
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katarzyna Müller Luda
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Emma K Persson
- VIB-Ugent Center for Inflammation Research, Ghent, Belgium
| | - Jennifer L Marshall
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Nonantzin Beristain-Covarrubias
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Juan Carlos Yam-Puc
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Madelene Dahlgren
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jenny J Persson
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Satoshi Uematsu
- International Research and Development Centre for Mucosal Vaccine, Institute for Medical Science, The University of Tokyo, Tokyo, Japan.,Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shizuo Akira
- World Premier International Immunology Frontier Research Centre, Osaka University, Suita, Japan
| | - Ian R Henderson
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Bengt Johansson Lindbom
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden.,Section of Biology and Chemistry, Department for Micro- and Nanotechnology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - William Agace
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden.,Section of Biology and Chemistry, Department for Micro- and Nanotechnology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
13
|
Cao XX, Li YH, Ye QL, Hu X, Wang TF, Fan MW. Self-assembling anticaries mucosal vaccine containing ferritin cage nanostructure and glucan-binding region of S. mutans glucosyltransferase effectively prevents caries formation in rodents. Hum Vaccin Immunother 2018; 13:2332-2340. [PMID: 28759297 DOI: 10.1080/21645515.2017.1349046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Anticaries protein vaccines that induce a mucosal immune response are not effective. Therefore, development of effective and convenient anticaries vaccines is a priority of dental research. Here we generated self-assembling nanoparticles by linking the glucan-binding region of Streptococcus mutans glucosyltransferase (GLU) to the N-terminal domain of ferritin to determine whether these novel nanoparticles enhanced the immunogenicity of an anticaries protein vaccine against GLU in rodents. We constructed the expression plasmid pET28a-GLU-FTH and purified the proteins from bacteria using size-exclusion chromatography. BALB/c mice were used to evaluate the ability of GLU-ferritin (GLU-FTH) nanoparticles to induce GLU-specific mucosal and systemic responses. The protective efficiency of GLU-FTH nanoparticles was compared with that of GLU alone or a mixture of GLU and poly(I:C) after administering an intranasal infusion to Wistar rats. The phagocytosis and maturation of dendritic cells (DCs) exposed in vitro to the nanoparticles were assessed using flow cytometry. The GLU-FTH nanoparticle vaccine elicited significantly higher levels of GLU-specific antibodies compared with GLU or a mixture of GLU and poly(I:C). Immunization with GLU-FTH achieved lower caries scores compared with those of the other vaccines. Administration of GLU-FTH nanoparticles enhanced phagocytosis by DCs and their maturation. Thus, self-assembling GLU-FTH is a highly effective anticaries mucosal vaccine that enhanced antibody production and inhibited S. mutans infection in rodents.
Collapse
Affiliation(s)
- Xi-Xi Cao
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM) , School & Hospital of Stomatology, Wuhan University , Wuhan , China
| | - Yu-Hong Li
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM) , School & Hospital of Stomatology, Wuhan University , Wuhan , China.,b Department of Endodontics , School and Hospital of Stomatology, Wuhan University , Wuhan , China
| | - Qian-Lin Ye
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM) , School & Hospital of Stomatology, Wuhan University , Wuhan , China
| | - Xuan Hu
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM) , School & Hospital of Stomatology, Wuhan University , Wuhan , China
| | - Tian-Feng Wang
- c Department of Oral Radiology , Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , Amsterdam , The Netherlands
| | - Ming-Wen Fan
- a The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM) , School & Hospital of Stomatology, Wuhan University , Wuhan , China
| |
Collapse
|
14
|
Flores-Langarica A, Müller Luda K, Persson EK, Cook CN, Bobat S, Marshall JL, Dahlgren MW, Hägerbrand K, Toellner KM, Goodall MD, Withers DR, Henderson IR, Johansson Lindbom B, Cunningham AF, Agace WW. CD103 +CD11b + mucosal classical dendritic cells initiate long-term switched antibody responses to flagellin. Mucosal Immunol 2018; 11:681-692. [PMID: 29346347 PMCID: PMC5912514 DOI: 10.1038/mi.2017.105] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/23/2017] [Indexed: 02/04/2023]
Abstract
Antibody responses induced at mucosal and nonmucosal sites demonstrate a significant level of autonomy. Here, we demonstrate a key role for mucosal interferon regulatory factor-4 (IRF4)-dependent CD103+CD11b+ (DP), classical dendritic cells (cDCs) in the induction of T-dependent immunoglobulin G (IgG) and immunoglobulin A (IgA) responses in the mesenteric lymph node (MLN) following systemic immunization with soluble flagellin (sFliC). In contrast, IRF8-dependent CD103+CD11b- (SP) are not required for these responses. The lack of this response correlated with a complete absence of sFliC-specific plasma cells in the MLN, small intestinal lamina propria, and surprisingly also the bone marrow (BM). Many sFliC-specific plasma cells accumulating in the BM of immunized wild-type mice expressed α4β7+, suggesting a mucosal origin. Collectively, these results suggest that mucosal DP cDC contribute to the generation of the sFliC-specific plasma cell pool in the BM and thus serve as a bridge linking the mucosal and systemic immune system.
Collapse
Affiliation(s)
- A Flores-Langarica
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - K Müller Luda
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - E K Persson
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - C N Cook
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - S Bobat
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - J L Marshall
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - M W Dahlgren
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - K Hägerbrand
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
| | - K M Toellner
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - M D Goodall
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - D R Withers
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - I R Henderson
- Institute of Microbiology and Infection, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - B Johansson Lindbom
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU). Kongens Lyngby, Denmark
| | - A F Cunningham
- Institute of Immunology & Immunotherapy, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Institute of Microbiology and Infection, College of Medical & Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - W W Agace
- Immunology Section, Lund University, BMC D14 Sölvegatan 19, S-221 84. Lund 22184, Sweden
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU). Kongens Lyngby, Denmark
| |
Collapse
|
15
|
Schülke S, Kuttich K, Wolfheimer S, Duschek N, Wangorsch A, Reuter A, Briza P, Pablos I, Gadermaier G, Ferreira F, Vieths S, Toda M, Scheurer S. Conjugation of wildtype and hypoallergenic mugwort allergen Art v 1 to flagellin induces IL-10-DC and suppresses allergen-specific TH2-responses in vivo. Sci Rep 2017; 7:11782. [PMID: 28924222 PMCID: PMC5603567 DOI: 10.1038/s41598-017-11972-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/16/2017] [Indexed: 11/09/2022] Open
Abstract
Allergies to weed pollen including members of the Compositae family, such as mugwort, ragweed, and feverfew are spreading worldwide. To efficiently treat these newly arising allergies, allergen specific immunotherapy needs to be improved. Therefore, we generated novel vaccine candidates consisting of the TLR5-ligand Flagellin A from Listeria and the major mugwort allergen Art v 1 including either the wild type Art v 1 sequence (rFlaA:Artv1) or a hypoallergenic variant (rFlaA:Artv1hyp) with reduced IgE-binding capacity. Immune modulating capacity of these constructs and respective controls was evaluated in vitro and in vivo. Incorporation of hypoallergenic Art v 1 derivative did not interfere with the resulting fusion proteins’ immune stimulatory capacity. Both rFlaA:Artv1 and rFlaA:Artv1hyp induced a prominent, mTOR-dependent, IL-10 secretion from murine dendritic cells, and suppressed allergen-specific TH2-cytokine secretion in vitro and in vivo. Both conjugates retained the capacity to induce rFlaA-specific antibody responses while efficiently inducing production of Art v 1-specific IgG1 and IgG2a antibodies in mice. Interestingly, only the suppression of TH2-cytokine secretion by rFlaA:Artv1 (but not rFlaA:Artv1hyp) was paralleled by a strong secretion of IFN-γ. In summary, we provided evidence that incorporating hypoallergens into flagellin:allergen fusion proteins is a suitable strategy to further improve these promising vaccine candidates.
Collapse
Affiliation(s)
- Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany.
| | - Kirsten Kuttich
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Sonja Wolfheimer
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Nadine Duschek
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Andrea Wangorsch
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Andreas Reuter
- Division of Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Peter Briza
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Isabel Pablos
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Gabriele Gadermaier
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Fatima Ferreira
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Stefan Vieths
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Masako Toda
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Stephan Scheurer
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| |
Collapse
|
16
|
Yang J, Sun Y, Bao R, Zhou D, Yang Y, Cao Y, Yu J, Zhao B, Li Y, Yan H, Zhong M. Second-generation Flagellin-rPAc Fusion Protein, KFD2-rPAc, Shows High Protective Efficacy against Dental Caries with Low Potential Side Effects. Sci Rep 2017; 7:11191. [PMID: 28894188 PMCID: PMC5593867 DOI: 10.1038/s41598-017-10247-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022] Open
Abstract
Dental caries is one of the most common global chronic diseases affecting all ages of the population; thus a vaccine against caries is urgently needed. Our previous studies demonstrated that a fusion protein, KF-rPAc, in which rPAc of S. mutans is directly fused to the C-terminal of E. coli-derived flagellin (KF), could confer high prophylactic and therapeutic efficiency against caries. However, possible side effects, including the high antigenicity of flagellin and possible inflammatory injury induced by flagellin, may restrict its clinical usage. Here, we produced a second-generation flagellin-rPAc fusion protein, KFD2-rPAc, by replacing the main antigenicity region domains D2 and D3 of KF with rPAc. Compared with KF-rPAc, KFD2-rPAc has lower TLR5 agonist efficacy and induces fewer systemic inflammatory responses in mice. After intranasal immunization, KFD2-rPAc induces significantly lower flagellin-specific antibody responses but a comparable level of rPAc-specific antibody responses in mice. More importantly, in rat challenge models, KFD2-rPAc induces a robust rPAc-specific IgA response, and confers efficient prophylactic and therapeutic efficiency against caries as does KF-rPAc, while the flagellin-specific antibody responses are highly reduced. In conclusion, low side effects and high protective efficiency against caries makes the second-generation flagellin-rPAc fusion protein, KFD2-rPAc, a promising vaccine candidate against caries.
Collapse
Affiliation(s)
- Jingyi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Ying Sun
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Rong Bao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.,Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, Hubei, 430071, China
| | - Dihan Zhou
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Yi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Yuan Cao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Jie Yu
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Bali Zhao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Yaoming Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China
| | - Maohua Zhong
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, 430071, China.
| |
Collapse
|
17
|
Flagellin: a unique microbe-associated molecular pattern and a multi-faceted immunomodulator. Cell Mol Immunol 2017; 14:862-864. [PMID: 28845044 DOI: 10.1038/cmi.2017.78] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/08/2017] [Indexed: 01/07/2023] Open
|
18
|
Sun Y, Yang Y, Zhou D, Cao Y, Yu J, Zhao B, Zhong M, Li Y, Yang J, Yan H. Flagellin-rPAc vaccine inhibits biofilm formation but not proliferation of S. mutans. Hum Vaccin Immunother 2016; 12:2847-2854. [PMID: 27392114 DOI: 10.1080/21645515.2016.1203496] [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: 12/19/2022] Open
Abstract
As the main etiologic bacterium of dental caries, Streptococcus mutans (S. mutans) has been considered as the primary object of vaccine research. We previously constructed a recombinant flagellin-rPAc fusion protein (KF-rPAc) that consists of an alanine-rich region to proline-rich region fragment of PAc (rPAc) from S. mutans and flagellin KF from E.coli K12 strain. Intranasal (i.n) immunization of KF-rPAc could induce high level of rPAc-specific antibody responses and offer robust protection against dental caries. In caries development, biofilm formation was considered as the necessary process involved. As PAc possesses other activities besides affecting adherence of S. mutans to salivary glycoproteins, we wondered whether rPAc-specific antibody responses induced by KF-rPAc could inhibit biofilm formation. Hence, in the present study, a simple and convenient in vitro biofilm model of S. mutans was constructed without saliva pre-coated. Both serum and saliva from KF-rPAc immunized rats significantly inhibited biofilm formation. Moreover, with the presence of serum or saliva, the biofilm formation is negatively correlated with the level of rPAc-specific antibody, and positively correlated with caries scores in rat. Moreover, in immunized mice, the level of rPAc-specific antibody also negatively correlated with the biofilm formation. Unlike ampicillin, serum of KF-rPAc immunized mice only inhibited biofilm formation but not proliferation. All together, we discovered that besides the well known blocking adherence of S. mutans to salivary glycoproteins by rPAc-specific antibody, flagellin-rPAc vaccine could also protects tooth from caries by inhibiting biofilm structure formation in between bacteria.
Collapse
Affiliation(s)
- Ying Sun
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Yi Yang
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Dihan Zhou
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Yuan Cao
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Jie Yu
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Bali Zhao
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Maohua Zhong
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Yaoming Li
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Jingyi Yang
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| | - Huimin Yan
- a Mucosal Immunity Research Group , State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences , Wuhan , Hubei , China
| |
Collapse
|
19
|
Generic determinants of Streptococcus colonization and infection. INFECTION GENETICS AND EVOLUTION 2015; 33:361-70. [DOI: 10.1016/j.meegid.2014.09.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/10/2014] [Accepted: 09/14/2014] [Indexed: 11/20/2022]
|
20
|
Bao R, Yang JY, Sun Y, Zhou DH, Yang Y, Li YM, Cao Y, Xiao Y, Li W, Yu J, Zhao BL, Zhong MH, Yan HM. Flagellin-PAc Fusion Protein Inhibits Progression of Established Caries. J Dent Res 2015; 94:955-60. [PMID: 25883108 DOI: 10.1177/0022034515582224] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Dental caries remains one of the most common infectious diseases of humankind, which develops slowly throughout life, affecting children, adolescents, and adults. A vaccine against caries is urgently needed. We previously developed recombinant flagellin as a mucosal adjuvant for anti-Streptococcus mutans vaccines by nasal immunization. Furthermore, we demonstrated a fusion protein strategy that combined flagellin and the target surface adhesion protein (PAc) in a single construct. This construct enhanced specific IgA responses in oral fluids and provided improved prophylactic protection against caries. In the present study, we observed prolonged progression of dental caries in rats after S. mutans Ingbritt challenge. In addition, we observed a therapeutic effect of the flagellin-PAc fusion protein (KF-rPAc) against dental caries as a mucosal vaccine with a new immunization protocol. The present study demonstrated that KF-rPAc by nasal immunization can promote PAc-specific systemic and mucosal antibody responses and inhibit dental caries progression efficiently after the implant of S. mutans into the oral cavity of the rats. The rats immunized with KF-rPAc exhibited 53.9% caries reduction compared with the sham-immunized rats. Our data support the concept of administration of KF-rPAc to humans after infection and even caries that has begun to alleviate caries progression. In conclusion, our study demonstrated that KF-rPAc could be used as an anticaries therapeutic mucosal vaccine.
Collapse
Affiliation(s)
- R Bao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan, Hubei, China
| | - J Y Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Y Sun
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - D H Zhou
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Y Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Y M Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Y Cao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Y Xiao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - W Li
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - J Yu
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - B L Zhao
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - M H Zhong
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - H M Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| |
Collapse
|
21
|
EV71 infection correlates with viral IgG preexisting at pharyngo-laryngeal mucosa in children. Virol Sin 2015; 30:146-52. [PMID: 25813631 DOI: 10.1007/s12250-014-3555-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/10/2015] [Indexed: 10/23/2022] Open
Abstract
Enterovirus 71 (EV71) infection causes severe central nervous system damage, particularly for children under the age of 5 years old, which remains a major public health burden worldwide. Clinical data released that children may be repeatedly infected by different members in enterovirus and get even worsen. Mucosa, especially epithelium of alimentary canal, was considered the primary site of EV71 infection. It has been elusive whether the preexsiting viral antibody in mucosa plays a role in EV71 infection. To answer this question, we respectively measured viral antibody response and EV71 RNA copy number of one hundred throat swab specimens from clinically confirmed EV71-infected children. The results released that low-level of mucosal IgG antibody against EV71 broadly existed in young population. More importantly, it further elucidated that the children with mucosal preexsiting EV71 IgG were prone to be infected, which suggested a former viral IgG mediated enhancement of viral infection in vivo.
Collapse
|
22
|
Immune Adjuvant Effect of Molecularly-defined Toll-Like Receptor Ligands. Vaccines (Basel) 2014; 2:323-53. [PMID: 26344622 PMCID: PMC4494261 DOI: 10.3390/vaccines2020323] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 01/07/2023] Open
Abstract
Vaccine efficacy is optimized by addition of immune adjuvants. However, although adjuvants have been used for over a century, to date, only few adjuvants are approved for human use, mostly aimed at improving vaccine efficacy and antigen-specific protective antibody production. The mechanism of action of immune adjuvants is diverse, depending on their chemical and molecular nature, ranging from non-specific effects (i.e., antigen depot at the immunization site) to specific activation of immune cells leading to improved host innate and adaptive responses. Although the detailed molecular mechanism of action of many adjuvants is still elusive, the discovery of Toll-like receptors (TLRs) has provided new critical information on immunostimulatory effect of numerous bacterial components that engage TLRs. These ligands have been shown to improve both the quality and the quantity of host adaptive immune responses when used in vaccine formulations targeted to infectious diseases and cancer that require both humoral and cell-mediated immunity. The potential of such TLR adjuvants in improving the design and the outcomes of several vaccines is continuously evolving, as new agonists are discovered and tested in experimental and clinical models of vaccination. In this review, a summary of the recent progress in development of TLR adjuvants is presented.
Collapse
|
23
|
Comparative genomics and immunoinformatics approach for the identification of vaccine candidates for enterohemorrhagic Escherichia coli O157:H7. Infect Immun 2014; 82:2016-26. [PMID: 24595137 DOI: 10.1128/iai.01437-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 strains are major human food-borne pathogens, responsible for bloody diarrhea and hemolytic-uremic syndrome worldwide. Thus far, there is no vaccine for humans against EHEC infections. In this study, a comparative genomics analysis was performed to identify EHEC-specific antigens useful as potential vaccines. The genes present in both EHEC EDL933 and Sakai strains but absent in nonpathogenic E. coli K-12 and HS strains were subjected to an in silico analysis to identify secreted or surface-expressed proteins. We obtained a total of 65 gene-encoding protein candidates, which were subjected to immunoinformatics analysis. Our criteria of selection aided in categorizing the candidates as high, medium, and low priority. Three members of each group were randomly selected and cloned into pVAX-1. Candidates were pooled accordingly to their priority group and tested for immunogenicity against EHEC O157:H7 using a murine model of gastrointestinal infection. The high-priority (HP) pool, containing genes encoding a Lom-like protein (pVAX-31), a putative pilin subunit (pVAX-12), and a fragment of the type III secretion structural protein EscC (pVAX-56.2), was able to induce the production of EHEC IgG and sIgA in sera and feces. HP candidate-immunized mice displayed elevated levels of Th2 cytokines and diminished cecum colonization after wild-type challenge. Individually tested HP vaccine candidates showed that pVAX-12 and pVAX-56.2 significantly induced Th2 cytokines and production of fecal EHEC sIgA, with pVAX-56.2 reducing EHEC cecum colonization. We describe here a bioinformatics approach able to identify novel vaccine candidates potentially useful for preventing EHEC O157:H7 infections.
Collapse
|
24
|
Kozlova D, Sokolova V, Zhong M, Zhang E, Yang J, Li W, Yang Y, Buer J, Westendorf AM, Epple M, Yan H. Calcium phosphate nanoparticles show an effective activation of the innate immune response in vitro and in vivo after functionalization with flagellin. Virol Sin 2013; 29:33-9. [PMID: 24374818 DOI: 10.1007/s12250-014-3379-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/19/2013] [Indexed: 12/20/2022] Open
Abstract
For subunit vaccines, adjuvants play a key role in shaping the magnitude, persistence and form of targeted antigen-specific immune response. Flagellin is a potent immune activator by bridging innate inflammatory responses and adaptive immunity and an adjuvant candidate for clinical application. Calcium phosphate nanoparticles are efficient carriers for different biomolecules like DNA, RNA, peptides and proteins. Flagellin-functionalized calcium phosphate nanoparticles were prepared and their immunostimulatory effect on the innate immune system, i.e. the cytokine production, was studied. They induced the production of the proinflammatory cytokines IL-8 (Caco-2 cells) and IL-1β (bone marrow-derived macrophages; BMDM) in vitro and IL-6 in vivo after intraperitoneal injection in mice. The immunostimulation was more pronounced than with free flagellin.
Collapse
Affiliation(s)
- Diana Kozlova
- Institute of Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Essen, 45117, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Tarahomjoo S. Utilizing bacterial flagellins against infectious diseases and cancers. Antonie van Leeuwenhoek 2013; 105:275-88. [PMID: 24276957 DOI: 10.1007/s10482-013-0075-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/12/2013] [Indexed: 12/31/2022]
Abstract
The flagellum is the organelle providing motility to bacterial cells and its activity is coupled to the cellular chemotaxis machinery. The flagellar filament is the largest portion of the flagellum, which consists of repeating subunits of the protein flagellin. Receptors of the innate immune system including Toll like receptor 5, ICE protease activating factor, and neuronal apoptosis inhibitory protein 5 signal in response to bacterial flagellins. In addition to inducing innate immune responses, bacterial flagellins mediate the development of adaptive immune responses to both flagellins and coadministered antigens. Therefore, these proteins have intensively been investigated for the vaccine development and the immunotherapy. This review describes the utilization of bacterial flagellins for the construction of vaccines against infectious diseases and cancer immunotherapy. Furthermore, the key factors affecting the performance of these systems are highlighted.
Collapse
Affiliation(s)
- Shirin Tarahomjoo
- Department of Biotechnology, Razi Vaccine and Serum Research Institute, 31975/148, Karaj, Iran,
| |
Collapse
|
26
|
Abstract
Snapshot of key developments in the patent literature accompanied by explanatory synopses
Collapse
|
27
|
Brandtzaeg P. Secretory IgA: Designed for Anti-Microbial Defense. Front Immunol 2013; 4:222. [PMID: 23964273 PMCID: PMC3734371 DOI: 10.3389/fimmu.2013.00222] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/16/2013] [Indexed: 01/30/2023] Open
Abstract
Prevention of infections by vaccination remains a compelling goal to improve public health. Mucosal vaccines would make immunization procedures easier, be better suited for mass administration, and most efficiently induce immune exclusion - a term coined for non-inflammatory antibody shielding of internal body surfaces, mediated principally by secretory immunoglobulin A (SIgA). The exported antibodies are polymeric, mainly IgA dimers (pIgA), produced by local plasma cells (PCs) stimulated by antigens that target the mucose. SIgA was early shown to be complexed with an epithelial glycoprotein - the secretory component (SC). A common SC-dependent transport mechanism for pIgA and pentameric IgM was then proposed, implying that membrane SC acts as a receptor, now usually called the polymeric Ig receptor (pIgR). From the basolateral surface, pIg-pIgR complexes are taken up by endocytosis and then extruded into the lumen after apical cleavage of the receptor - bound SC having stabilizing and innate functions in the secretory antibodies. Mice deficient for pIgR show that this is the only receptor responsible for epithelial export of IgA and IgM. These knockout mice show a variety of defects in their mucosal defense and changes in their intestinal microbiota. In the gut, induction of B-cells occurs in gut-associated lymphoid tissue, particularly the Peyer's patches and isolated lymphoid follicles, but also in mesenteric lymph nodes. PC differentiation is accomplished in the lamina propria to which the activated memory/effector B-cells home. The airways also receive such cells from nasopharynx-associated lymphoid tissue but by different homing receptors. This compartmentalization is a challenge for mucosal vaccination, as are the mechanisms used by the mucosal immune system to discriminate between commensal symbionts (mutualism), pathobionts, and overt pathogens (elimination).
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Centre for Immune Regulation (CIR), University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
28
|
Yang J, Zhang E, Liu F, Zhang Y, Zhong M, Li Y, Zhou D, Chen Y, Cao Y, Xiao Y, He B, Yang Y, Sun Y, Lu M, Yan H. Flagellins of Salmonella Typhi and nonpathogenic Escherichia coli are differentially recognized through the NLRC4 pathway in macrophages. J Innate Immun 2013; 6:47-57. [PMID: 23816851 DOI: 10.1159/000351476] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/19/2013] [Indexed: 01/14/2023] Open
Abstract
Flagellin is recognized by both Toll-like receptor (TLR)5 and NAIP5/NLRC4 inflammasome receptors. We hypothesized that the flagellins derived from different bacteria might differentially activate TLR5 and/or NAIP5/NLRC4 signal pathways. To test this, the immune recognition of recombinant flagellins derived from pathogenic Salmonella Typhi (SF) and the nonpathogenic Escherichia coli K12 strain MG1655 (KF) were examined by the activation of TLR5 and NLRC4 pathways in various cell types. While flagellins SF and KF were not distinguishable in activating the TLR5 pathway, KF induced significantly less interleukin-1β production and pyroptotic cell death in peritoneal macrophages than SF, and showed markedly lower efficiency in activating caspase-1 through the NLRC4 pathway than SF. Macrophages may differentially recognize flagellins by intracellular sensors and thereby initiate the immune response to invading pathogenic bacteria. Our findings suggest an active role of flagellin as an important determinant in host differential immune recognition and for the control of bacteria infection.
Collapse
Affiliation(s)
- Jingyi Yang
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yan H. Salivary IgA enhancement strategy for development of a nasal-spray anti-caries mucosal vaccine. SCIENCE CHINA-LIFE SCIENCES 2013; 56:406-13. [PMID: 23633072 DOI: 10.1007/s11427-013-4473-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 03/21/2013] [Indexed: 01/09/2023]
Abstract
Dental caries remains one of the most common global chronic diseases caused by Streptococcus mutans, which is prevalent all over the world. The caries prevalence of children aged between 5-6 years old in China is still in very high rate. A potent and effective anti-caries vaccine has long been expected for caries prevention but no vaccines have been brought to market till now mainly due to the low ability to induce and maintain protective antibody in oral fluids. This review will give a brief historical retrospect on study of dental caries and pathogenesis, effective targets for anti-caries vaccines, oral immune system and immunization against dental caries. Then, salivary IgA antibodies and the protective responses are discussed in the context of the ontogeny of mucosal immunity to indigenous oral streptococcal. The methods and advancement for induction of specific anti-caries salivary sIgA antibodies and enhancement of specific anti-caries salivary sIgA antibodies by intranasal immunization with a safe effective mucosal adjuvant are described. The progress in the enhancement of salivary sIgA antibodies and anti-caries protection by intranasal immunization with flagellin-PAc fusion protein will be highlighted. Finally, some of the main strategies that have been used for successful mucosal vaccination of caries vaccine are reviewed, followed by discussion of the mucosal adjuvant choice for achieving protective immunity at oral mucosal membranes for development of a nasal-spray or nasal-drop anti-caries vaccine for human.
Collapse
Affiliation(s)
- Huimin Yan
- Mucosal Immunity Research Group, State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
30
|
Brandtzaeg P. Secretory immunity with special reference to the oral cavity. J Oral Microbiol 2013; 5:20401. [PMID: 23487566 PMCID: PMC3595421 DOI: 10.3402/jom.v5i0.20401] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 01/29/2013] [Accepted: 02/15/2013] [Indexed: 11/14/2022] Open
Abstract
The two principal antibody classes present in saliva are secretory IgA (SIgA) and IgG; the former is produced as dimeric IgA by local plasma cells (PCs) in the stroma of salivary glands and is transported through secretory epithelia by the polymeric Ig receptor (pIgR), also named membrane secretory component (SC). Most IgG in saliva is derived from the blood circulation by passive leakage mainly via gingival crevicular epithelium, although some may be locally produced in the gingiva or salivary glands. Gut-associated lymphoid tissue (GALT) and nasopharynx-associated lymphoid tissue (NALT) do not contribute equally to the pool of memory/effector B cells differentiating to mucosal PCs throughout the body. Thus, enteric immunostimulation may not be the best way to activate the production of salivary IgA antibodies although the level of specific SIgA in saliva may still reflect an intestinal immune response after enteric immunization. It remains unknown whether the IgA response in submandibular/sublingual glands is better related to B-cell induction in GALT than the parotid response. Such disparity is suggested by the levels of IgA in submandibular secretions of AIDS patients, paralleling their highly upregulated intestinal IgA system, while the parotid IgA level is decreased. Parotid SIgA could more consistently be linked to immune induction in palatine tonsils/adenoids (human NALT) and cervical lymph nodes, as supported by the homing molecule profile observed after immune induction at these sites. Several other variables influence the levels of antibodies in salivary secretions. These include difficulties with reproducibility and standardization of immunoassays, the impact of flow rate, acute or chronic stress, protein loss during sample handling, and uncontrolled admixture of serum-derived IgG and monomeric IgA. Despite these problems, saliva is an easily accessible biological fluid with interesting scientific and clinical potentials.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Per Brandtzaeg, Department of Pathology, Oslo University Hospital, Rikshospitalet, PO Box 4950 Oslo, NO-0424 Norway. Tel: +47-23072743, Fax: 47-23071511.
| |
Collapse
|
31
|
Yang J, Zhong M, Zhang Y, Zhang E, Sun Y, Cao Y, Li Y, Zhou D, He B, Chen Y, Yang Y, Yu J, Yan H. Antigen replacement of domains D2 and D3 in flagellin promotes mucosal IgA production and attenuates flagellin-induced inflammatory response after intranasal immunization. Hum Vaccin Immunother 2013; 9:1084-92. [PMID: 23377752 DOI: 10.4161/hv.23809] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Targeting early infection in mucosal sites is one of the primary goals for mucosal vaccines so as to prevent pathogen mucosal transmission and infection. The TLR5 agonist flagellin was deemed to be a mucosal adjuvant candidate for clinical usage. However, the high antigenicity of flagellin and the possible inflammatory injury induced by flagellin might restrict its clinical usage. Here HIV-1 p24 protein was selected as an antigen model and we replaced the main antigenicity region domains D2 and D3 of non-pathogenic E.coli-derived flagellin (KF). The derived soluble protein KFD-p24 3D was then compared with KF-p24, which fused p24 directly to the C-terminal of KF. In vitro and ex vivo experiments showed that KFD-p24 3D has lower TLR5 agonist efficacy and less immunocyte-activating efficacy. Interestingly, the production of KF- specific antibody was highly reduced, and KFD-p24 3D induced IgA-biased antibody responses in mucosal sites. Moreover, KFD-p24 3D induced far fewer systemic inflammatory responses and abrogated detectable inflammatory side effects on mice, even at the high dose. The properties of enhanced IgA generation and attenuated inflammatory responses broaden the safe-dose range of KFD-p24 3D flagellin, creating a potentially promising mucosal adjuvant.
Collapse
Affiliation(s)
- Jingyi Yang
- Mucosal Immunity Research Group; State Key Laboratory of Virology; Wuhan Institute of Virology; Chinese Academy of Sciences; Wuhan, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Adair P, Burnside G, Pine C. Analysis of Health Behaviour Change Interventions for Preventing Dental Caries Delivered in Primary Schools. Caries Res 2013; 47 Suppl 1:2-12. [DOI: 10.1159/000351829] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/10/2013] [Indexed: 11/19/2022] Open
|