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
|
Zhang F, Ni L, Zhang Z, Luo X, Wang X, Zhou W, Chen J, Liu J, Qu Y, Liu K, Guo L. Recombinant L. lactis vaccine LL-plSAM-WAE targeting four virulence factors provides mucosal immunity against H. pylori infection. Microb Cell Fact 2024; 23:61. [PMID: 38402145 PMCID: PMC10893618 DOI: 10.1186/s12934-024-02321-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) causes chronic gastric disease. An efficient oral vaccine would be mucosa-targeted and offer defense against colonization of invasive infection in the digestive system. Proteolytic enzymes and acidic environment in the gastrointestinal tract (GT) can, however, reduce the effectiveness of oral vaccinations. For the creation of an edible vaccine, L. lactis has been proposed as a means of delivering vaccine antigens. RESULTS We developed a plSAM (pNZ8148-SAM) that expresses a multiepitope vaccine antigen SAM-WAE containing Urease, HpaA, HSP60, and NAP extracellularly (named LL-plSAM-WAE) to increase the efficacy of oral vaccinations. We then investigated the immunogenicity of LL-plSAM-WAE in Balb/c mice. Mice that received LL-plSAM-WAE or SAM-WAE with adjuvant showed increased levels of antibodies against H. pylori, including IgG and sIgA, and resulted in significant reductions in H. pylori colonization. Furthermore, we show that SAM-WAE and LL-plSAM-WAE improved the capacity to target the vaccine to M cells. CONCLUSIONS These findings suggest that recombinant L. lactis could be a promising oral mucosa vaccination for preventing H. pylori infection.
Collapse
Affiliation(s)
- Furui Zhang
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China
| | - Linhan Ni
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhen Zhang
- Department of Geriatrics and Special Needs Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xuequan Wang
- Key Laboratory of Radiation Oncology of Taizhou, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, 317000, China
| | - Wenmiao Zhou
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiale Chen
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Jing Liu
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China
| | - Yuliang Qu
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China.
| | - Kunmei Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, 750004, China.
| | - Le Guo
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China.
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
3
|
Danielewicz N, Rosato F, Dai W, Römer W, Turnbull WB, Mairhofer J. Microbial carbohydrate-binding toxins – From etiology to biotechnological application. Biotechnol Adv 2022; 59:107951. [DOI: 10.1016/j.biotechadv.2022.107951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/22/2022] [Accepted: 04/02/2022] [Indexed: 02/06/2023]
|
4
|
Mohammadzadeh R, Soleimanpour S, Pishdadian A, Farsiani H. Designing and development of epitope-based vaccines against Helicobacter pylori. Crit Rev Microbiol 2021; 48:489-512. [PMID: 34559599 DOI: 10.1080/1040841x.2021.1979934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori infection is the principal cause of serious diseases (e.g. gastric cancer and peptic ulcers). Antibiotic therapy is an inadequate strategy in H. pylori eradication because of which vaccination is an inevitable approach. Despite the presence of countless vaccine candidates, current vaccines in clinical trials have performed with poor efficacy which makes vaccination extremely challenging. Remarkable advancements in immunology and pathogenic biology have provided an appropriate opportunity to develop various epitope-based vaccines. The fusion of proper antigens involved in different aspects of H. pylori colonization and pathogenesis as well as peptide linkers and built-in adjuvants results in producing epitope-based vaccines with excellent therapeutic efficacy and negligible adverse effects. Difficulties of the in vitro culture of H. pylori, high genetic variation, and unfavourable immune responses against feeble epitopes in the complete antigen are major drawbacks of current vaccine strategies that epitope-based vaccines may overcome. Besides decreasing the biohazard risk, designing precise formulations, saving time and cost, and induction of maximum immunity with minimum adverse effects are the advantages of epitope-based vaccines. The present article is a comprehensive review of strategies for designing and developing epitope-based vaccines to provide insights into the innovative vaccination against H. pylori.
Collapse
Affiliation(s)
- Roghayeh Mohammadzadeh
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Reference Tuberculosis Laboratory, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Pishdadian
- Department of Immunology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Bioinformatic prediction and identification of immunogenic epitopes of the antigenic 14-3-3 protein of Echinococcus multilocularis. Acta Trop 2021; 220:105955. [PMID: 33979643 DOI: 10.1016/j.actatropica.2021.105955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Alveolar echinococcosis is a high-risk parasitic disease caused by the larval stage of Echinococcus multilocularis. The study aimed to predict and identify the dominant Th1/Th2 and B cell epitopes of the antigen protein 14-3-3 beta:alpha from Echinococcus multilocularis. METHODS A comparison of the four amino acid sequences of 14-3-3 beta:alpha was respectively derived from Echinococcus multilocularis, Rattus norvegicus, Canis lupus familiaris, and Homo sapiens was carried out by CLUSTALW to provide a basis for excluding similar epitopes. The amino acid sequence information was analyzed by SOPMA and the homology model was established by Swiss-Model. IEDB and SYFPEITHI were used to predict T cell epitopes. According to the Bcepred and ABCpred, the B cell epitopes were comprehensively predicted and analyzed. The dominant epitopes were validated by Lymphocyte Proliferation, ELISA, ELISpot, and Flow cytometry. RESULTS Eight potential epitopes of 14-3-3 from Echinococcus multilocularis were screened according to the results of prediction and analysis: 14-3-31-15, 14-3-36-21, 14-3-371-86, 14-3-3144-157, 14-3-3145-166, 14-3-3146-160, 14-3-3153-161, and 14-3-3164-177. The 3D structure model of the protein was constructed and the location distribution of potential epitope was ascertained. Respectively, the epitopes of the dominant antigen of B cells were validated as 14-3-3145-166 and 14-3-3164-177; the Th1 dominant antigen epitopes were 14-3-36-21, 14-3-3145-166; and the Th2 dominant epitopes was 14-3-3145-166. CONCLUSION In this study, two dominant antigen epitopes of B cells, two Th1 dominant antigen epitopes, and one Th2 dominant antigen epitope were validated. Our work provides a basis for the subsequent development of efficient and safe vaccines targeting epitopes of Echinococcus multilocularis.
Collapse
|
6
|
Liu M, Zhong Y, Chen J, Liu Y, Tang C, Wang X, Zhang Y, Wang P, Logan SM, Chen W, Wei B. Oral immunization of mice with a multivalent therapeutic subunit vaccine protects against Helicobacter pylori infection. Vaccine 2020; 38:3031-3041. [PMID: 32139315 DOI: 10.1016/j.vaccine.2020.02.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori is a human class I carcinogen and no effective prophylactic or therapeutic H. pylori vaccine has yet been marketed. H. pylori can escape the host immune response, but the precise immune protection mechanisms in humans remain unknown. In this study, we developed a multivalent, subunit H. pylori vaccine candidate by formulating three commonly used H. pylori antigens, neutrophil-activating protein (NAP), urease subunit A (UreA) and subunit B (UreB) with the mucosal adjuvant, a double-mutant heat-labile toxin (dmLT) from Escherichia coli, and evaluated its immunogenicity and therapeutic efficacy in a mouse model of H. pylori infection. We found that oral immunization of H. pylori-infected mice significantly reduced gastric bacterial colonization at both 2 and 8 weeks after immunization. The reduction in bacterial burdens was accompanied with significantly increased serum antigen-specific IgG responses and mucosal IgA responses. Moreover, oral immunization also induced Th1/Th17 immune responses, which may play a synergistic role with the specific antibodies in the elimination of H. pylori. Thus, our vaccine candidate appears able to overcome the immune evasion mechanism of H. pylori, restore the suppression of Th2 immune responses with the induction of a strong humoral immune response. These results lay the foundation for the development of an optimized oral therapeutic H. pylori vaccine with increased immunogenicity of UreA and UreB, as well as providing long-term immunity.
Collapse
Affiliation(s)
- Meiying Liu
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Youxiu Zhong
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Jing Chen
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Yu Liu
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Chongfa Tang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Xuewei Wang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Yanbin Zhang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Ping Wang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Susan M Logan
- Human Health Therapeutics (HHT) Research Center, National Research Council Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6, Canada
| | - Wangxue Chen
- Human Health Therapeutics (HHT) Research Center, National Research Council Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6, Canada.
| | - Bo Wei
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China.
| |
Collapse
|
7
|
Abbas G, Zafar I, Ahmad S, Azam SS. Immunoinformatics design of a novel multi-epitope peptide vaccine to combat multi-drug resistant infections caused by Vibrio vulnificus. Eur J Pharm Sci 2020; 142:105160. [PMID: 31751777 DOI: 10.1016/j.ejps.2019.105160] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/23/2019] [Accepted: 11/16/2019] [Indexed: 12/19/2022]
Abstract
Multi-drug resistant Vibrio vulnificus is a Gram-negative bacillus responsible for diseases, such as: sepsis, septicemia, gastroenteritis, and fatal necrotizing fasciitis in humans. The treatment and prevention of V. vulnificus infections are challenging because of resistance to antibiotics and the non-availability of a licensed vaccine. Considering this, an in-silico based approach comprising subtractive proteomics, immunoinformatics, molecular docking, and dynamics simulation studies is applied herein to identify potential epitope vaccine candidates for the mentioned pathogen. Two potential vaccine candidates: vibC and flgL are filtered based on essentiality, outer membrane localization, virulence, antigenic, pathway mapping, and cellular protein-protein network analysis. Using immunoinformatic tools, 9-mer B-cell derived T-cell antigenic epitopes are predicted for the said shortlisted two proteins that are demonstrating excellent affinity for predominant HLA allele (DRB1*0101) in human population. Screened peptides are used further in multi-epitope peptide designing and linked to an adjuvant to enhance the immunogenic properties of the designed construct. Furthermore, the construct was docked blindly to TLR4 immune receptor, and analyzed in conformational dynamics simulation to decipher the complex affinity and understand time dependent behavior, respectively. We expect this designed in silico construct to be useful for vaccinologists to evaluate its immune protective efficacy in in vivo animal models.
Collapse
Affiliation(s)
- Ghulam Abbas
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan
| | - Iqra Zafar
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan
| | - Sajjad Ahmad
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
8
|
Xu N, Ruan G, Liu W, Hu C, Huang A, Zeng Z, Luo S, Zhang Z, Fan M, Ye F, Xi T, Xing Y. Vaccine-induced gastric CD4 + tissue-resident memory T cells proliferate in situ to amplify immune response against Helicobacter pylori insult. Helicobacter 2019; 24:e12652. [PMID: 31414552 DOI: 10.1111/hel.12652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tissue-resident memory T cells accelerate the clearance of pathogens during recall response. However, whether CD4+ TRM cells themselves can provide gastric immunity is unclear. MATERIALS AND METHODS We established a parabiosis model between the enhanced green fluorescent protein and wild-type mice that the circulation system was shared, and the wild-type partner was vaccinated with H pylori vaccine composed of CCF and silk fibroin in gastric subserous layer to induce gastric EGFP+ CD4+ TRM cells. Antigen-specific EGFP+ CD4+ T cells and proliferous TRM cells were analyzed by flow cytometry. The colonization of H pylori was detected by quantitative real-time PCR. EGFP+ CD4+ TRM cells and the inflammation of the stomach were observed by histology. RESULTS A parabiosis animal model was employed to identify the cells that introduced by vaccination in GSL. Antigen-specific EGFP+ CD4+ T cells could be detected at day 7 post-vaccination. Thirty days later, EGFP+ CD4+ TRM cells were established with a phenotype of CD69+ CD103- . Of note, we found that when circulating lymphocytes were depleted by FTY720 administration, these TRM cells could proliferate in situ and differentiate into effector Th1 cells after H pylori challenge. A decrease in H pylori colonization was observed in the vaccinated mice but not unvaccinated mice. Further, we found that although FTY720 was administrated, mounted pro-inflammatory myeloid cells still emerged in the stomach of the vaccinated mice, which might contribute to the reduction of H pylori colonization. CONCLUSIONS Our study reveals that H pylori vaccine-induced CD4+ TRM cells can proliferate and differentiate in situ to enhance gastric local immunity during recall response.
Collapse
Affiliation(s)
- Ningyin Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Guojing Ruan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Chupeng Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - An Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhenxing Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Menghui Fan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Feng Ye
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
9
|
Luo S, Liu W, Zeng Z, Ye F, Hu C, Xu N, Huang A, Xi T, Xing Y. Toxic adjuvants alter the function and phenotype of dendritic cells to initiate adaptive immune responses induced by oral Helicobacter pylori vaccines. Helicobacter 2018; 23:e12536. [PMID: 30247802 DOI: 10.1111/hel.12536] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Toxic adjuvant is considered as an indispensable constituent for oral Helicobacter pylori (H. pylori) vaccines. However, the elaborate role of toxic adjuvant in the initiation of adaptive immune response is largely undescribed. MATERIALS AND METHODS We employed an acid-resistant HP55/PLGA nanoparticles (NPs) delivery system encapsulating three antigens (Hsp, Nap, and Lpp20) from H. pylori and accompanied with three adjuvants (LPS, CpG, and chimeric flagellum (CF)) to explore the underlying mechanism of the adjuvant constituent. H. pylori-specific antibody responses were detected by ELISA. Gastric inflammatory and Th1/Th17 responses were analyzed by flow cytometry. Expressions of inflammatory cytokines were measured by quantitative real-time PCR. RESULTS In bone marrow-derived dendritic cells' (BMDCs) model, the addition of toxic adjuvants is responsible for the proinflammatory function, but not the mature phenotype of BMDCs. In vivo, intestinal loop injection with NPs + LPS, rather than NPs alone, altered the dendritic cell (DC) phenotypes in mesenteric lymph nodes and drove a local proinflammatory microenvironment. In a prophylactic vaccination model, mice immunized with NPs + adjuvants significantly reduced the gastric colonization of H. pylori, induced antigen-specific antibody responses and Th1/Th17 cell responses. After H. pylori challenge, these mice showed potent recall responses involving both neutrophil and inflammatory monocyte infiltration. Additionally, TLR4 knockout mice were immunized with NPs + LPS and NPs + CF, respectively; only the recipients of NPs + CF orchestrated a protective response to control bacterial infection. CONCLUSIONS Our study indicated that toxic adjuvants within oral H.pylori vaccines altered the function and phenotype of dendritic cells and facilitated the establishment of proinflammatory microenvironment to initiate adaptive immune responses.
Collapse
Affiliation(s)
- Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Feng Ye
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chupeng Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Ningyin Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - An Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
10
|
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
|
11
|
Liu H, Liu W, Tan Z, Zeng Z, Yang H, Luo S, Wang L, Xi T, Xing Y. Promoting Immune Efficacy of the Oral Helicobacter pylori Vaccine by HP55/PBCA Nanoparticles against the Gastrointestinal Environment. Mol Pharm 2018; 15:3177-3186. [PMID: 30011213 DOI: 10.1021/acs.molpharmaceut.8b00251] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immunogenicity of oral subunit vaccines is poor partly as a result of the harsh milieu of the gastrointestinal (GI) tract. For some pathogens that restrictedly inhabit the GI tract, a vaccine that works in situ may provide more potent protection than vaccines that operate parenterally. Yet, no appropriate delivery system is available for oral subunit vaccines. In this study, we designed HP55/poly( n-butylcyanoacrylate) (PBCA) nanoparticles (NPs) to carry Helicobacter pylori ( H. pylori) subunit vaccine CCF for oral administration in a prophylactic mice model. These NPs, which are synthesized using an interfacial polymerization method, protected the CCF antigen not only from the acidic pH in simulated gastric fluid (SGF, pH 1.2) but also from the proteolysis in simulated intestinal fluid (SIF, pH 7.4). Oral vaccination of mice with HP55/PBCA-CCF NPs promoted the production of serum antigen-specific antibodies, mucosal secretory IgA, and proinflammatory cytokines. Moreover, a Th1/Th17 response and augmented lymphocytes were found in the gastric tissue of HP55/PBCA-CCF NP-immunized mice, which might eventually limit H. pylori colonization. Collectively, these results indicate that HP55/PBCA NPs are promising carriers against the severe situation of the GI tract and thereby may be further utilized for other orally administrated vaccines or drugs.
Collapse
Affiliation(s)
- Hai Liu
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Wei Liu
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Zhoulin Tan
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Zhiqin Zeng
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Huimin Yang
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Shuanghui Luo
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Linlin Wang
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Tao Xi
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Yingying Xing
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| |
Collapse
|
12
|
Pan X, Ke H, Niu X, Li S, Lv J, Pan L. Protection Against Helicobacter pylori Infection in BALB/c Mouse Model by Oral Administration of Multivalent Epitope-Based Vaccine of Cholera Toxin B Subunit-HUUC. Front Immunol 2018; 9:1003. [PMID: 29867978 PMCID: PMC5951970 DOI: 10.3389/fimmu.2018.01003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/23/2018] [Indexed: 12/20/2022] Open
Abstract
Vaccination is an increasingly important alternative approach to control Helicobacter pylori infection, since H. pylori resistance to previously efficacious antibiotic regimens is increased, and H. pylori eradication treatment for upper gastrointestinal diseases is becoming less successful. Fortunately, an efficient oral monovalent H. pylori vaccine has been developed. However, compared with monovalent vaccines, multivalent vaccines have the potential to induce more effective and comprehensive protection against H. pylori infection. In this study, we designed and produced a multivalent epitope-based vaccine cholera toxin B subunit (CTB)-HUUC with the intramucosal adjuvant CTB and tandem copies of B-cell epitopes (HpaA132-141, UreA183-203, and UreB321-339) and T-cell epitopes (HpaA88-100, UreA27-53, UreB229-251, UreB317-329, UreB373-385, UreB438-452, UreB546-561, CagA149-164, and CagA196-217) from H. pylori adhesion A subunit (HpaA), urease A subunit (UreA), urease B subunit (UreB), and cytotoxin-associated antigen (CagA). Serum IgG, stomach, and intestine mucosal sIgA from mice after CTB-HUUC vaccination neutralized H. pylori urease activity in vitro. CTB-HUUC vaccination promoted H. pylori-specific lymphocyte responses and a mixed CD4+ T cell immune response as indicated by IFN-γ, interleukin-4, and interleukin-17 production in mice. Both oral prophylactic and therapeutic CTB-HUUC vaccinations reduced gastric urease activity and H. pylori infection and protected stomachs in mice. Taken together, CTB-HUUC is a promising potent and safe multivalent vaccine in controlling H. pylori infection in BALB/c mouse model.
Collapse
Affiliation(s)
- Xing Pan
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Hong Ke
- Department of Hemotology, People's Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaojuan Niu
- Department of Pharmacology, Hubei University of Medicine, Shiyan, China
| | - Shan Li
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Jun Lv
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Longrui Pan
- Department of Pharmacology, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
13
|
Li R, Yang Q, Guo L, Feng L, Wang W, Liu K, Tang F, Ge RL. Immunological features and efficacy of the recombinant subunit vaccine LTB-EMY162 against Echinococcus multilocularis metacestode. Appl Microbiol Biotechnol 2018; 102:2143-2154. [PMID: 29354854 DOI: 10.1007/s00253-018-8771-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 12/15/2022]
Abstract
Alveolar echinococcosis is a zoonotic disease caused by the infection of the larval stage Echinococcus multilocularis with worldwide distribution especially in the northwest China. It is important to develop a well-tolerated immunoprophylaxis against E. multilocularis for alveolar echinococcosis control. In this study, a prokaryotic expression system for recombinant immunogen LTB-EMY162 was established, and the immunological features, sensitized lymphocyte, IL-4/IFN-γ secreted, prophylactic effect, and therapeutic effect were also evaluated. Arctic Express (DE3) system, Ni2+-charged and molecular sieve chromatography were used to obtain a high-purity 29 kDa protein. The ELISA and lymphocyte proliferation assay showed that LTB-EMY162 induced high-titer specific IgG against EMY162 and E. multilocularis protoscoleces protein in BALB/c mice and promoted sensitized T lymphocyte cell proliferation, and LTB-EMY162 stimulated Th cell to secrete IL-4 and IFN-γ and induced a Th1/Th2 mixed type immunological response. We also found that LTB-EMY162 significantly inhibited the cysts formation by challenging with 1000 E. multilocularis protoscoleces. The growth of protoscoleces and cysts were also significantly decreased by treating with LTB-EMY162 in 1000 protoscoleces intraperitoneal injection therapeutic mice model. In conclusion, we have constructed a subunit vaccine LTB-EMY162 which has prevention and therapeutic effect against E. multilocularis infection.
Collapse
MESH Headings
- Animals
- Antibodies, Helminth/immunology
- Antigens, Helminth/administration & dosage
- Antigens, Helminth/genetics
- Antigens, Helminth/immunology
- Echinococcus multilocularis/genetics
- Echinococcus multilocularis/growth & development
- Echinococcus multilocularis/immunology
- Female
- Humans
- Interferon-gamma/immunology
- Interleukin-4/immunology
- Life Cycle Stages
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- T-Lymphocytes/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Runle Li
- Research Center for High Altitude Medicine, Qinghai University, 16# Kunlun Road, Xining, 810001, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory for Echinococcosis, 16# Kunlun Road, Xining, 810001, China
| | - Quanyu Yang
- Research Center for High Altitude Medicine, Qinghai University, 16# Kunlun Road, Xining, 810001, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory for Echinococcosis, 16# Kunlun Road, Xining, 810001, China
| | - Le Guo
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Lin Feng
- Research Center for High Altitude Medicine, Qinghai University, 16# Kunlun Road, Xining, 810001, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory for Echinococcosis, 16# Kunlun Road, Xining, 810001, China
| | - Wei Wang
- Research Center for High Altitude Medicine, Qinghai University, 16# Kunlun Road, Xining, 810001, China
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China
- Qinghai Key Laboratory for Echinococcosis, 16# Kunlun Road, Xining, 810001, China
| | - Kunmei Liu
- Ningxia Key Laboratory of Cerebrocranial Diseases, School of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Feng Tang
- Research Center for High Altitude Medicine, Qinghai University, 16# Kunlun Road, Xining, 810001, China.
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China.
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China.
- Qinghai Key Laboratory for Echinococcosis, 16# Kunlun Road, Xining, 810001, China.
- Research Center for High Altitude Zoonosis, Qinghai University, Xining, 810001, China.
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, 16# Kunlun Road, Xining, 810001, China.
- Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China.
- Qinghai Key Laboratory of Science and Technology for High Altitude Medicine, 16# Kunlun Road, Xining, 810001, China.
- Qinghai Key Laboratory for Echinococcosis, 16# Kunlun Road, Xining, 810001, China.
- Research Center for High Altitude Zoonosis, Qinghai University, Xining, 810001, China.
| |
Collapse
|
14
|
Ji J, Sundquist J, Sundquist K. Cholera Vaccine Use Is Associated With a Reduced Risk of Death in Patients With Colorectal Cancer: A Population-Based Study. Gastroenterology 2018; 154:86-92.e1. [PMID: 28923497 DOI: 10.1053/j.gastro.2017.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/01/2017] [Accepted: 09/11/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Cholera toxin can act as a modulator of the immune response with anti-inflammatory effects; it reduces development of colon polyps in mouse models of colorectal cancer (CRC). We performed a population-based study to determine whether, in patients with a diagnosis of CRC, subsequent administration of the cholera vaccine (killed Vibrio cholerae O1 whole cells and recombinant cholera toxin B subunit) affects mortality. METHODS We identified patients from the Swedish Cancer Register who were diagnosed with CRC from July 2005 through December 2012. These patients were linked to the Swedish Prescribed Drug Register to retrieve cholera vaccine use. We used Cox regression analysis to calculate the hazard ratio (HR) of death from CRC and overall mortality in patients with post-diagnostic use of cholera vaccine compared with matched controls. RESULTS A total of 175 patients were diagnosed with CRC and given a prescription for the cholera vaccine after their cancer diagnosis. Compared with propensity score-matched controls and adjusted for confounding factors, patients with CRC who received the cholera vaccine had a decreased risk of death from CRC (HR, 0.53; 95% CI, 0.29-0.99) and a decreased risk of death overall (HR, 0.59; 95% CI, 0.37-0.94). The decrease in mortality with cholera vaccination was largely observed, irrespective of patient age or tumor stage at diagnosis or sex. CONCLUSIONS In a population-based study, we associated administration of the cholera vaccine after CRC diagnosis with decreased risk of death from CRC and overall mortality.
Collapse
Affiliation(s)
- Jianguang Ji
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden.
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University/Region Skåne, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
15
|
Liu W, Tan Z, Liu H, Zeng Z, Luo S, Yang H, Zheng L, Xi T, Xing Y. Nongenetically modified Lactococcus lactis-adjuvanted vaccination enhanced innate immunity against Helicobacter pylori. Helicobacter 2017; 22. [PMID: 28805287 DOI: 10.1111/hel.12426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Gram-positive enhancer matrix particles (GEM) produced by Lactococcus lactis can enhance vaccine-induced immune response. However, the mechanism under which this adjuvant mounts the efficacy of orally administered vaccines remains unexplored. MATERIALS AND METHODS We used a prophylactic mice model to investigate the mechanism of GEM-adjuvanted vaccination. Helicobacter pylori urease-specific antibody response was monitored and detected in murine serum by ELISA. Urease-specific splenic cytokine profile was examined. Gastric inflammatory responses were measured on day 43 or 71 by quantitative real-time PCR, flow cytometry and histology. RESULTS We found that GEM enhanced the efficiency of oral H. pylori vaccine by promoting innate immunity. The vaccine CUE-GEM composed of GEM particles and recombinant antigen CTB-UE provided protection of immunized mice against H. pylori insult. The protective response was associated with induction of postimmunization gastritis and local Th1/Th17 cell-medicated immune response. We showed that innate inflammatory responses including neutrophil chemokines CXCL1-2, neutrophils, and antimicrobial proteins S100A8 and MUC1 were significantly elevated. Within all infected mice, S100A8 and MUC1 levels were negatively correlated with H. pylori burden. Strikingly, mice receiving GEM also show reduction of colonization, possibly through natural host response pathways to recruit CD4+ T cells and promote S100A8 expression. CONCLUSIONS These findings suggest that GEM-based vaccine may impact Th1/Th17 immunity to orchestrate innate immune response against H. pylori infection.
Collapse
Affiliation(s)
- Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhoulin Tan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Hai Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Huimin Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
16
|
Tan Z, Liu W, Liu H, Li C, Zhang Y, Meng X, Tang T, Xi T, Xing Y. Oral Helicobacter pylori vaccine-encapsulated acid-resistant HP55/PLGA nanoparticles promote immune protection. Eur J Pharm Biopharm 2017; 111:33-43. [DOI: 10.1016/j.ejpb.2016.11.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 11/02/2016] [Indexed: 12/19/2022]
|
17
|
Li Y, Chen Z, Ye J, Ning L, Luo J, Zhang L, Jiang Y, Xi Y, Ning Y. Antibody Production and Th1-biased Response Induced by an Epitope Vaccine Composed of Cholera Toxin B Unit and Helicobacter pylori Lpp20 Epitopes. Helicobacter 2016; 21:234-48. [PMID: 26332255 DOI: 10.1111/hel.12268] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The epitope vaccine is an attractive potential for prophylactic and therapeutic vaccination against Helicobacter pylori (H. pylori) infection. Lpp20 is one of major protective antigens which trigger immune response after H. pylori invades host and has been considered as an excellent vaccine candidate for the control of H. pylori infection. In our previous study, one B-cell epitope and two CD4(+) T-cell epitopes of Lpp20 were identified. OBJECTIVE In this study, an epitope vaccine composed of mucosal adjuvant cholera toxin B subunit (CTB) and these three identified Lpp20 epitopes were constructed to investigate the efficacy of this epitope vaccine in mice. METHODS The epitope vaccine including CTB, one B-cell, and two CD4(+) T-cell epitopes of Lpp20 was constructed and named CTB-Lpp20, which was then expressed in Escherichia coli and used for intraperitoneal immunization in BALB/c mice. The immunogenicity, specificity, and ability to induce antibodies against Lpp20 and cytokine secretion were evaluated. After that, CTB-Lpp20 was intragastrically immunized to investigate the prophylactic and therapeutic efficacy in infected mice. RESULTS The results indicated that the epitope vaccine CTB-Lpp20 possessed good immunogenicity and immunoreactivity and could elicit specific high level of antibodies against Lpp20 and the cytokine of IFN-γ and IL-17. Additionally, CTB-Lpp20 significantly decreased H. pylori colonization in H. pylori challenging mice, and the protection was correlated with IgG, IgA, and sIgA antibody and Th1-type cytokines. CONCLUSION This study will be better for understanding the protective immunity of epitope vaccine, and CTB-Lpp20 may be an alternative strategy for combating H. pylori invasion.
Collapse
Affiliation(s)
- Yan Li
- School of Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Zhongbiao Chen
- School of Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Jianbin Ye
- School of Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Lijun Ning
- School of Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Jun Luo
- Department of microbiology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lili Zhang
- School of Nursing, Southern Medical University, Guangzhou, 510515, China
| | - Yin Jiang
- School of Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Yue Xi
- School of Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Yunshan Ning
- School of Biotechnology, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
18
|
Therapeutic efficacy of oral immunization with a non-genetically modified Lactococcus lactis-based vaccine CUE-GEM induces local immunity against Helicobacter pylori infection. Appl Microbiol Biotechnol 2016; 100:6219-6229. [DOI: 10.1007/s00253-016-7333-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/04/2016] [Accepted: 01/17/2016] [Indexed: 12/11/2022]
|
19
|
A novel chimeric flagellum fused with the multi-epitope vaccine CTB-UE prevents Helicobacter pylori-induced gastric cancer in a BALB/c mouse model. Appl Microbiol Biotechnol 2015; 99:9495-502. [DOI: 10.1007/s00253-015-6705-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/16/2022]
|
20
|
Baldauf KJ, Royal JM, Hamorsky KT, Matoba N. Cholera toxin B: one subunit with many pharmaceutical applications. Toxins (Basel) 2015; 7:974-96. [PMID: 25802972 PMCID: PMC4379537 DOI: 10.3390/toxins7030974] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/16/2015] [Indexed: 12/22/2022] Open
Abstract
Cholera, a waterborne acute diarrheal disease caused by Vibrio cholerae, remains prevalent in underdeveloped countries and is a serious health threat to those living in unsanitary conditions. The major virulence factor is cholera toxin (CT), which consists of two subunits: the A subunit (CTA) and the B subunit (CTB). CTB is a 55 kD homopentameric, non-toxic protein binding to the GM1 ganglioside on mammalian cells with high affinity. Currently, recombinantly produced CTB is used as a component of an internationally licensed oral cholera vaccine, as the protein induces potent humoral immunity that can neutralize CT in the gut. Additionally, recent studies have revealed that CTB administration leads to the induction of anti-inflammatory mechanisms in vivo. This review will cover the potential of CTB as an immunomodulatory and anti-inflammatory agent. We will also summarize various recombinant expression systems available for recombinant CTB bioproduction.
Collapse
Affiliation(s)
- Keegan J Baldauf
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Joshua M Royal
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY 42303, USA.
| | - Krystal Teasley Hamorsky
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY 42303, USA.
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| | - Nobuyuki Matoba
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
- Owensboro Cancer Research Program of James Graham Brown Cancer Center at University of Louisville School of Medicine, Owensboro, KY 42303, USA.
| |
Collapse
|
21
|
Therapeutic efficacy of the multi-epitope vaccine CTB-UE against Helicobacter pylori infection in a Mongolian gerbil model and its microRNA-155-associated immuno-protective mechanism. Vaccine 2014; 32:5343-52. [PMID: 25093281 DOI: 10.1016/j.vaccine.2014.07.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/11/2014] [Accepted: 07/09/2014] [Indexed: 12/24/2022]
Abstract
Vaccination is an effective means of preventing infectious diseases, including those caused by Helicobacter pylori. In this study, we constructed a novel multi-epitope vaccine, CTB-UE, composed of the cholera toxin B subunit and tandem copies of the B and Th cell epitopes from the H. pylori urease A and B subunits. We evaluated the therapeutic efficacy of the multi-epitope vaccine CTB-UE against H. pylori infection in a Mongolian gerbil model and studied its immuno-protective mechanisms. The experimental results indicated that urease activity, H. pylori colonisation density, the levels of IL-8 and TNF-α in the serum, and the levels of COX-2 and NAP in gastric tissue were significantly lower and the IgG level in the serum and the IFN-γ level in spleen lymphocytes were significantly higher in the vaccinated group compared with the model control group; additionally, gastric mucosal inflammation was notably alleviated following vaccination. The results showed that CTB-UE had a good therapeutic effect on H. pylori infection. The immuno-protective mechanism was closely related to the immune response mediated by microRNA-155, the expression of which was strongly up-regulated after CTB-UE administration. The expression levels of the microRNA-155 target proteins IFN-γRα, AID, and PU.1 were significantly down-regulated; these results indicated that CTB-UE induced an immune response biased towards Th1 cells by up-regulating microRNA-155 to inhibit IFN-γRα expression and induced a humoral immune response towards B cells by up-regulating microRNA-155 to inhibit PU.1 and AID expression. These results demonstrate that the multi-epitope vaccine CTB-UE may be a promising therapeutic vaccine against H. pylori infection and is a new therapeutic tool for human use.
Collapse
|
22
|
Li X, Xing Y, Guo L, Lv X, Song H, Xi T. Oral immunization with recombinant Lactococcus lactis delivering a multi-epitope antigen CTB-UE attenuates Helicobacter pylori infection in mice. Pathog Dis 2014; 72:78-86. [PMID: 24687988 DOI: 10.1111/2049-632x.12173] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 12/23/2022] Open
Abstract
Urease is an essential virulence factor and colonization factor for Helicobacter pylori (H. pylori) and is considered as an excellent vaccine candidate antigen. However, conventional technologies for preparing an injectable vaccine require purification of the antigenic protein and preparation of an adjuvant. Lactococcus lactis NZ9000 (L. lactis) could serve as an antigen-delivering vehicle for the development of edible vaccine. In previous study, we constructed a multi-epitope vaccine, designated CTB-UE, which is composed of the mucosal adjuvant cholera toxin B subunit (CTB), three Th cell epitopes and two B-cell epitopes from urease subunits. To develop a novel type of oral vaccine against H. pylori, genetically modified L. lactis strains were established to secrete this epitope vaccine extracellularly in this study. Oral prophylactic immunization with recombinant L. lactis significantly elicited humoral anti-urease antibody responses (P < 0.001) and reduced the gastric colonization of H. pylori from 7.14 ± 0.95 to 4.68 ± 0.98 log10 CFU g(-1) stomach. This L. lactis oral vaccine offers a promising vaccine candidate for the control of H. pylori infection.
Collapse
Affiliation(s)
- Xinyang Li
- Biotechnology Center, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | |
Collapse
|
23
|
Immunological features and efficacy of a multi-epitope vaccine CTB-UE against H. pylori in BALB/c mice model. Appl Microbiol Biotechnol 2013; 98:3495-507. [DOI: 10.1007/s00253-013-5408-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 11/11/2013] [Accepted: 11/11/2013] [Indexed: 12/31/2022]
|
24
|
Konieczna I, Zarnowiec P, Kwinkowski M, Kolesinska B, Fraczyk J, Kaminski Z, Kaca W. Bacterial urease and its role in long-lasting human diseases. Curr Protein Pept Sci 2013; 13:789-806. [PMID: 23305365 PMCID: PMC3816311 DOI: 10.2174/138920312804871094] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/15/2012] [Accepted: 09/03/2012] [Indexed: 02/07/2023]
Abstract
Urease is a virulence factor found in various pathogenic bacteria. It is essential in colonization of a host organism and in maintenance of bacterial cells in tissues. Due to its enzymatic activity, urease has a toxic effect on human cells. The presence of ureolytic activity is an important marker of a number of bacterial infections. Urease is also an immunogenic protein and is recognized by antibodies present in human sera. The presence of such antibodies is connected with progress of several long-lasting diseases, like rheumatoid arthritis, atherosclerosis or urinary tract infections. In bacterial ureases, motives with a sequence and/or structure similar to human proteins may occur. This phenomenon, known as molecular mimicry, leads to the appearance of autoantibodies, which take part in host molecules destruction. Detection of antibodies-binding motives (epitopes) in bacterial proteins is a complex process. However, organic chemistry tools, such as synthetic peptide libraries, are helpful in both, epitope mapping as well as in serologic investigations. In this review, we present a synthetic report on a molecular organization of bacterial ureases - genetic as well as structural. We characterize methods used in detecting urease and ureolytic activity, including techniques applied in disease diagnostic processes and in chemical synthesis of urease epitopes. The review also provides a summary of knowledge about a toxic effect of bacterial ureases on human body and about occurrence of anti-urease antibodies in long-lasting diseases.
Collapse
Affiliation(s)
- Iwona Konieczna
- Department of Microbiology, Institute of Biology, The Jan Kochanowski University, ul. Swietokrzyska 15, 25-406 Kielce, Poland.
| | | | | | | | | | | | | |
Collapse
|
25
|
Hinc K, Iwanicki A, Obuchowski M. New stable anchor protein and peptide linker suitable for successful spore surface display in B. subtilis. Microb Cell Fact 2013; 12:22. [PMID: 23448318 PMCID: PMC3599258 DOI: 10.1186/1475-2859-12-22] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 02/18/2013] [Indexed: 01/28/2023] Open
Abstract
Background In last decade spores have been successfully used as a surface display platform. Various peptides or proteins were displayed this way as functional enzymes or antigens. Nearly all attempts involved use of three coat proteins: CotB, CotC or CotG. Increasing knowledge of the structure of the spore coat allowed us to propose the use of other proteins whose localization in the spore envelope has been determined. We also propose the application of a new linker suitable for building fusion proteins. Results We show that a member of the outer coat, CotZ, is a good candidate as a new anchor protein useful in spore surface display. This protein allows use of relatively large passenger proteins and their efficient display on the spore surface. Analysis by Western- and dot-blotting, combined with immunofluorescence microscopy, allowed us to estimate the number of displayed fusion proteins molecules as 1.4 × 102 per spore. In addition, we present data indicating that the use of a peptide linker, which forms a stable α-helix, may greatly improve the display of anchored proteins on the spore surface. Conclusion CotZ can be used as an efficient anchor protein in the outer spore coat. Its localisation in the coat crust layer should guarantee surface display of passenger proteins. Moreover, a CotZ based fusion can tolerate relatively large passenger proteins for efficient spore surface display. In addition, to the properties of both the anchor and passenger proteins, an important issue is the nature of the linker. Here we present evidence that the linker, which forms a stable α-helix, may be crucial for successful display.
Collapse
Affiliation(s)
- Krzysztof Hinc
- Laboratory of Molecular Bacteriology, Intercollegiate Faculty of Biotechnology UG & MUG, Medical University of Gdańsk, Dębinki 1, Gdańsk 80-211, Poland
| | | | | |
Collapse
|
26
|
Liu S, Li Y, Xu Z, Wang Y. Subcutaneous or oral immunization of mice with Lactococcus lactis expressing F4 fimbrial adhesin FaeG. J Vet Med Sci 2013; 75:779-84. [PMID: 23386358 DOI: 10.1292/jvms.12-0267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the most common causes of diarrhea in neonatal and postweaning piglets. Fimbrial adhesion of ETEC has been considered an important colonization factor with antigenicity. To safely and effectively deliver the F4 (K88) fimbrial adhesin FaeG to the immune system, we have previously constructed the secretory expression vector pNZ8112-faeG, and FaeG was produced in cytoplasmic form in Lactococcus lactis. In this work, BALB/c mice were immunized with recombinant L. lactis to further determine the immunogenicity of recombinant FaeG (rFaeG) via the subcutaneous or oral route. Subcutaneous immunization in mice with recombinant L. lactis induced a significant increase in the F4-specific serum IgG titer and the number of antibody-secreting cells (ASCs) in the spleen. Oral immunization of mice with recombinant L. lactis induced mucosal and systemic F4-specific immune responses and increased the number of ASCs in the spleen, mesenteric lymph nodes and Peyer's patches. High-dose (2.8 × 10(11) CFU) recombinant strains and adjuvant cholera toxin B subunit enhanced specific mucosal immune responses. The results suggest the feasibility of delivering rFaeG expressed in L. lactis to the immune system in order to induce an F4-specific immune response.
Collapse
Affiliation(s)
- Shujie Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, Zhejiang, China
| | | | | | | |
Collapse
|
27
|
Guo L, Liu K, Zhao W, Li X, Li T, Tang F, Zhang R, Wu W, Xi T. Immunological features and efficacy of the reconstructed epitope vaccine CtUBE against Helicobacter pylori infection in BALB/c mice model. Appl Microbiol Biotechnol 2012; 97:2367-78. [PMID: 23064457 DOI: 10.1007/s00253-012-4486-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 09/25/2012] [Accepted: 09/29/2012] [Indexed: 01/04/2023]
Abstract
Urease is an essential virulence factor and colonization factor for Helicobacter pylori, of which the urease B subunit (UreB) is considered as an excellent vaccine candidate antigen. In previous study, an epitope vaccine with cholera toxin B subunit (CTB) and an epitope (UreB321-339) named CtUBE was constructed and the mice were protected significantly after intragastric vaccination with the CtUBE liposome vaccine. However, the fusion protein CtUBE was expressed as inclusion bodies and was difficultly purified. Besides, the immunogenicity and specificity of the CtUBE vaccine was not investigated in a fairly wide and detailed way. In this study, the fusion peptide CtUBE was reconstructed and expressed as a soluble protein with pectinase signal peptide at the N terminus and the 6-his tag at its C-terminal, and then the immunogenicity, specificity, prophylactic, and therapeutic efficacy of the reconstructed CtUBE (rCtUBE) vaccine were evaluated in BALB/c mice model after purification. The experimental results indicated that mice immunized with rCtUBE could produce comparatively high level of specific antibodies which could respond to natural H. pylori urease, UreB, or the minimal epitope UreB327-334 involved with the active site of urease, and showed effectively inhibitory effect on the enzymatic activity of urease. Besides, oral prophylactic or therapeutic immunization with rCtUBE significantly decreased H. pylori colonization compared with oral immunization with rCTB or PBS, and the protection was correlated with antigen-specific IgG, IgA, and mucosal sIgA antibody responses, and a Th2 cells response. This rCtUBE vaccine may be a promising vaccine candidate for the control of H. pylori infection.
Collapse
Affiliation(s)
- Le Guo
- School of Laboratory Medicine, Ningxia Medical University, Yinchuan 750004, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The immune response to Helicobacter pylori is a multifaceted group of mechanisms involving responses that are both protective and damaging to the host. The innate and the adaptive immune responses lead to damaging inflammatory responses, but these responses may fail, allowing for persistence of many infections. Thus, developing new therapeutics and effective vaccines against H. pylori has proven to be arduous. In this manuscript, we will examine the advances in knowledge made in the past year in understanding the host immune response to H. pylori and the progress toward developing a vaccine.
Collapse
Affiliation(s)
- Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Irina V. Pinchuk
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Ellen J. Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
29
|
Ameri Shah Reza M, Mousavi Gargari SL, Rasooli I, Jalali Nadoushan M, Ebrahimizadeh W. Inhibition of H. pylori colonization and prevention of gastritis in murine model. World J Microbiol Biotechnol 2012; 28:2513-9. [PMID: 22806157 DOI: 10.1007/s11274-012-1059-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/14/2012] [Indexed: 01/15/2023]
Abstract
Helicobacter pylori is a Gram-negative spiral bacterium that colonizes human gastric mucosa causing infection. In this study aiming at inhibition of H. pylori infection we made an attempt to evaluate immunogenicity of the total (UreC) and C-terminal (UreCc) fragments of H. pylori urease. Total UreC and its C-terminal fragment were expressed in E. coli. Recombinant proteins were analyzed by SDS-PAGE and western blot and then purified by Ni-NTA affinity chromatography. Female C57BL6/j mice were immunized with the purified proteins (UreC and UreCc). Antibody titers from isolated sera were measured by ELISA. Immunized mice were then challenged by oral gavage with live H. pylori Sydney strain SS1. Total of 109 CFU were inoculated into stomach of immunized and unimmunized healthy mice three times each at one day interval. Eight weeks after the last inoculation, the blood sample was collected and the serum antibody titer was estimated by ELISA. Stomach tissues from control and experimental animal groups were studied histopathologically. UreC and UreCc yielded recombinant proteins of 61 and 31 kDa respectively. ELIZA confirmed establishment of immunity and the antibodies produced thereby efficiently recognized H. pylori and inhibited its colonization in vivo. Pathological analysis did not reveal established infection in immunized mice challenged with H. pylori. The results support the idea that UreC and UreCc specific antibodies contribute to protection against H. pylori infections.
Collapse
|