1
|
Elbehiry A, Marzouk E, Abalkhail A, Sindi W, Alzahrani Y, Alhifani S, Alshehri T, Anajirih NA, ALMutairi T, Alsaedi A, Alzaben F, Alqrni A, Draz A, Almuzaini AM, Aljarallah SN, Almujaidel A, Abu-Okail A. Pivotal role of Helicobacter pylori virulence genes in pathogenicity and vaccine development. Front Med (Lausanne) 2025; 11:1523991. [PMID: 39850097 PMCID: PMC11756510 DOI: 10.3389/fmed.2024.1523991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/13/2024] [Indexed: 01/25/2025] Open
Abstract
One of the most prevalent human infections is Helicobacter pylori (H. pylori), which affects more than half of the global population. Although H. pylori infections are widespread, only a minority of individuals develop severe gastroduodenal disorders. The global resistance of H. pylori to antibiotics has reached concerning levels, significantly impacting the effectiveness of treatment. Consequently, the development of vaccines targeting virulence factors may present a viable alternative for the treatment and prevention of H. pylori infections. This review aims to provide a comprehensive overview of the current understanding of H. pylori infection, with a particular focus on its virulence factors, pathophysiology, and vaccination strategies. This review discusses various virulence factors associated with H. pylori, such as cytotoxin-associated gene A (cagA), vacuolating cytotoxin gene (vacA), outer membrane proteins (OMPs), neutrophil-activated protein (NAP), urease (ure), and catalase. The development of vaccines based on these virulence characteristics is essential for controlling infection and ensuring long-lasting protection. Various vaccination strategies and formulations have been tested in animal models; however, their effectiveness and reproducibility in humans remain uncertain. Different types of vaccines, including vector-based vaccines, inactivated whole cells, genetically modified protein-based subunits, and multiepitope nucleic acid (DNA) vaccines, have been explored. While some vaccines have demonstrated promising results in murine models, only a limited number have been successfully tested in humans. This article provides a thorough evaluation of recent research on H. pylori virulence genes and vaccination methods, offering valuable insights for future strategies to address this global health challenge.
Collapse
Affiliation(s)
- Ayman Elbehiry
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Eman Marzouk
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Wael Sindi
- Department of Population, Public and Environmental Health, General Administration of Health Services, Ministry of Defense, Riyadh, Saudi Arabia
| | - Yasir Alzahrani
- Department of Psychiatry, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Salem Alhifani
- Department of Psychiatry, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Turki Alshehri
- Department of Dental, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | - Nuha Abdulaziz Anajirih
- Department of Medical Emergency Services, Faculty of Health Sciences, Umm Al-Qura University, Al-Qunfudah, Saudi Arabia
| | - Turki ALMutairi
- Department of Education and Training, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Ahmad Alsaedi
- Department of Education and Training, Prince Sultan Military College of Health Sciences, Dammam, Saudi Arabia
| | - Feras Alzaben
- Department of Food Service, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Abdullah Alqrni
- Department of Preventive Medicine, King Fahad Armed Hospital, Jeddah, Saudi Arabia
| | - Abdelmaged Draz
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Abdulaziz M. Almuzaini
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Sahar N. Aljarallah
- Department of Pharmacy Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Abdulrahman Almujaidel
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Akram Abu-Okail
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
2
|
Cąkała-Jakimowicz M, Domaszewska-Szostek A, Puzianowska-Kuznicka M. Interruption of Lymph Flow Worsens the Skin Inflammation Caused by Saprophytic Staphylococcus epidermidis. Biomedicines 2023; 11:3234. [PMID: 38137455 PMCID: PMC10740757 DOI: 10.3390/biomedicines11123234] [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: 10/18/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Lymphedema is often complicated by chronic inflammation, leading to fibrosis, fat deposition, and inhibition of lymphangiogenesis. This study aimed to verify whether lymphedema itself or together with commensal bacterial flora infection contributes to the severity of local inflammation. Edema was induced by interruption of the lymph flow in the rat's hind limb. Immune cell infiltrates were examined by flow cytometry and immunohistochemistry. Nine-day edema alone did not affect immune cell content in the skin but resulted in a decrease in CD4+ T helper lymphocytes and monocytes in the draining popliteal lymph nodes. In turn, local saprophytic Staphylococcus epidermidis infection of the edematous limb resulted in dense infiltrates of CD68+ macrophages and monocytes, MHC class II antigen-presenting cells, CD90+ stem cells, thymocytes, and immature B cells in the skin, accompanied by a simultaneous reduction in density of CD4+ T helper lymphocytes and monocytes, OX62+ dendritic cells, CD68+ macrophages and monocytes, HiS48+ granulocytes, CD90+ stem cells, thymocytes, and immature B cells in the draining popliteal lymph nodes. These results indicate that the combination of edema and saprophytic bacteria infection induces severe inflammation in the peripheral tissues and results in a delay of antibacterial protection processes in neighboring lymphatic organs.
Collapse
Affiliation(s)
- Marta Cąkała-Jakimowicz
- Department of Human Epigenetics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Anna Domaszewska-Szostek
- Department of Human Epigenetics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Monika Puzianowska-Kuznicka
- Department of Human Epigenetics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
| |
Collapse
|
3
|
Katsande PM, Nguyen VD, Nguyen TLP, Nguyen TKC, Mills G, Bailey DMD, Christie G, Hong HA, Cutting SM. Prophylactic immunization to Helicobacter pylori infection using spore vectored vaccines. Helicobacter 2023; 28:e12997. [PMID: 37314018 PMCID: PMC10909515 DOI: 10.1111/hel.12997] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/08/2023] [Accepted: 05/23/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND Helicobacter pylori infection remains a major public health threat leading to gastrointestinal illness and increased risk of gastric cancer. Mostly affecting populations in developing countries no vaccines are yet available and the disease is controlled by antimicrobials which, in turn, are driving the emergence of AMR. MATERIALS AND METHODS We have engineered spores of Bacillus subtilis to display putative H. pylori protective antigens, urease subunit A (UreA) and subunit B (UreB) on the spore surface. Following oral dosing of mice with these spores, we evaluated immunity and colonization in animals challenged with H. pylori. RESULTS Oral immunization with spores expressing either UreA or UreB showed antigen-specific mucosal responses (fecal sIgA) including seroconversion and hyperimmunity. Following challenge, colonization by H. pylori was significantly reduced by up to 1-log. CONCLUSIONS This study demonstrates the utility of bacterial spores for mucosal vaccination to H. pylori infection. The heat stability and robustness of Bacillus spores coupled with their existing use as probiotics make them an attractive solution for either protection against H. pylori infection or potentially for therapy and control of active infection.
Collapse
Affiliation(s)
| | - Van Duy Nguyen
- Institute of Biotechnology and EnvironmentNha Trang UniversityNha TrangVietnam
| | | | - Thi Kim Cuc Nguyen
- Institute of Biotechnology and EnvironmentNha Trang UniversityNha TrangVietnam
| | - Gabrielle Mills
- Department of Chemical Engineering & BiotechnologyUniversity of CambridgeCambridgeUK
| | - David M. D. Bailey
- Department of Chemical Engineering & BiotechnologyUniversity of CambridgeCambridgeUK
| | - Graham Christie
- Department of Chemical Engineering & BiotechnologyUniversity of CambridgeCambridgeUK
| | - Huynh Anh Hong
- Department of Biological SciencesRoyal Holloway University of LondonEghamUK
| | - Simon M. Cutting
- Department of Biological SciencesRoyal Holloway University of LondonEghamUK
| |
Collapse
|
4
|
Khan MA, Amin A, Farid A, Ullah A, Waris A, Shinwari K, Hussain Y, Alsharif KF, Alzahrani KJ, Khan H. Recent Advances in Genomics-Based Approaches for the Development of Intracellular Bacterial Pathogen Vaccines. Pharmaceutics 2022; 15:pharmaceutics15010152. [PMID: 36678781 PMCID: PMC9863128 DOI: 10.3390/pharmaceutics15010152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
Infectious diseases continue to be a leading cause of morbidity and mortality worldwide. The majority of infectious diseases are caused by intracellular pathogenic bacteria (IPB). Historically, conventional vaccination drives have helped control the pathogenesis of intracellular bacteria and the emergence of antimicrobial resistance, saving millions of lives. However, in light of various limitations, many diseases that involve IPB still do not have adequate vaccines. In response to increasing demand for novel vaccine development strategies, a new area of vaccine research emerged following the advent of genomics technology, which changed the paradigm of vaccine development by utilizing the complete genomic data of microorganisms against them. It became possible to identify genes related to disease virulence, genetic patterns linked to disease virulence, as well as the genetic components that supported immunity and favorable vaccine responses. Complete genomic databases, and advancements in transcriptomics, metabolomics, structural genomics, proteomics, immunomics, pan-genomics, synthetic genomics, and population biology have allowed researchers to identify potential vaccine candidates and predict their effects in patients. New vaccines have been created against diseases for which previously there were no vaccines available, and existing vaccines have been improved. This review highlights the key issues and explores the evolution of vaccines. The increasing volume of IPB genomic data, and their application in novel genome-based techniques for vaccine development, were also examined, along with their characteristics, and the opportunities and obstacles involved. Critically, the application of genomics technology has helped researchers rapidly select and evaluate candidate antigens. Novel vaccines capable of addressing the limitations associated with conventional vaccines have been developed and pressing healthcare issues are being addressed.
Collapse
Affiliation(s)
- Muhammad Ajmal Khan
- Division of Life Science, Center for Cancer Research, and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
- Correspondence: (M.A.K.); or (H.K.)
| | - Aftab Amin
- Division of Life Science, Center for Cancer Research, and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Awais Farid
- Division of Environment and Sustainability, Hong Kong University of Science and Technology, Hong Kong, China
| | - Amin Ullah
- Molecular Virology Laboratory, Department of Microbiology and Biotechnology, Abasyn University, Peshawar 25000, Pakistan
| | - Abdul Waris
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Khyber Shinwari
- Institute of Chemical Engineering, Department Immuno-Chemistry, Ural Federal University, Yekaterinbiurg 620002, Russia
| | - Yaseen Hussain
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Khalid J. Alzahrani
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Haroon Khan
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Correspondence: (M.A.K.); or (H.K.)
| |
Collapse
|
5
|
Zhang Y, Li X, Shan B, Zhang H, Zhao L. Perspectives from recent advances of Helicobacter pylori vaccines research. Helicobacter 2022; 27:e12926. [PMID: 36134470 DOI: 10.1111/hel.12926] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/25/2022] [Accepted: 08/17/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection is the main factor leading to some gastric diseases. Currently, H. pylori infection is primarily treated with antibiotics. However, with the widespread application of antibiotics, H. pylori resistance to antibiotics has also gradually increased year by year. Vaccines may be an alternative solution to clear H. pylori. AIMS By reviewing the recent progress on H. pylori vaccines, we expected it to lead to more research efforts to accelerate breakthroughs in this field. MATERIALS & METHODS We searched the research on H. pylori vaccine in recent years through PubMed®, and then classified and summarized these studies. RESULTS The study of the pathogenic mechanism of H. pylori has led to the development of vaccines using some antigens, such as urease, catalase, and heat shock protein (Hsp). Based on these antigens, whole-cell, subunit, nucleic acid, vector, and H. pylori exosome vaccines have been tested. DISCUSSION At present, researchers have developed many types of vaccines, such as whole cell vaccines, subunit vaccines, vector vaccines, etc. However, although some of these vaccines induced protective immunity in mouse models, only a few were able to move into human trials. We propose that mRNA vaccine may play an important role in preventing or treating H. pylori infection. The current study shows that we have developed various types of vaccines based on the virulence factors of H. pylori. However, only a few vaccines have entered human clinical trials. In order to improve the efficacy of vaccines, it is necessary to enhance T-cell immunity. CONCLUSION We should fully understand the pathogenic mechanism of H. pylori and find its core antigen as a vaccine target.
Collapse
Affiliation(s)
- Ying Zhang
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoya Li
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baoen Shan
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongtao Zhang
- University of Pennsylvania School of Medicine Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lianmei Zhao
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
6
|
Ghasemi A, Wang S, Sahay B, Abbott JR, Curtiss R. Protective immunity enhanced Salmonella vaccine vectors delivering Helicobacter pylori antigens reduce H. pylori stomach colonization in mice. Front Immunol 2022; 13:1034683. [PMID: 36466847 PMCID: PMC9716130 DOI: 10.3389/fimmu.2022.1034683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2024] Open
Abstract
Helicobacter pylori is a major cause of gastric mucosal inflammation, peptic ulcers, and gastric cancer. Emerging antimicrobial-resistant H. pylori has hampered the effective eradication of frequent chronic infections. Moreover, a safe vaccine is highly demanded due to the absence of effective vaccines against H. pylori. In this study, we employed a new innovative Protective Immunity Enhanced Salmonella Vaccine (PIESV) vector strain to deliver and express multiple H. pylori antigen genes. Immunization of mice with our vaccine delivering the HpaA, Hp-NAP, UreA and UreB antigens, provided sterile protection against H. pylori SS1 infection in 7 out of 10 tested mice. In comparison to the control groups that had received PBS or a PIESV carrying an empty vector, immunized mice exhibited specific and significant cellular recall responses and antigen-specific serum IgG1, IgG2c, total IgG and gastric IgA antibody titers. In conclusion, an improved S. Typhimurium-based live vaccine delivering four antigens shows promise as a safe and effective vaccine against H. pylori infection.
Collapse
Affiliation(s)
- Amir Ghasemi
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Jeffrey R. Abbott
- Department of Comparative, Diagnostic and Population Medicine, University of Florida, Gainesville, FL, United States
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| |
Collapse
|
7
|
Osterloh A. Vaccination against Bacterial Infections: Challenges, Progress, and New Approaches with a Focus on Intracellular Bacteria. Vaccines (Basel) 2022; 10:751. [PMID: 35632507 PMCID: PMC9144739 DOI: 10.3390/vaccines10050751] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/08/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Many bacterial infections are major health problems worldwide, and treatment of many of these infectious diseases is becoming increasingly difficult due to the development of antibiotic resistance, which is a major threat. Prophylactic vaccines against these bacterial pathogens are urgently needed. This is also true for bacterial infections that are still neglected, even though they affect a large part of the world's population, especially under poor hygienic conditions. One example is typhus, a life-threatening disease also known as "war plague" caused by Rickettsia prowazekii, which could potentially come back in a war situation such as the one in Ukraine. However, vaccination against bacterial infections is a challenge. In general, bacteria are much more complex organisms than viruses and as such are more difficult targets. Unlike comparatively simple viruses, bacteria possess a variety of antigens whose immunogenic potential is often unknown, and it is unclear which antigen can elicit a protective and long-lasting immune response. Several vaccines against extracellular bacteria have been developed in the past and are still used successfully today, e.g., vaccines against tetanus, pertussis, and diphtheria. However, while induction of antibody production is usually sufficient for protection against extracellular bacteria, vaccination against intracellular bacteria is much more difficult because effective defense against these pathogens requires T cell-mediated responses, particularly the activation of cytotoxic CD8+ T cells. These responses are usually not efficiently elicited by immunization with non-living whole cell antigens or subunit vaccines, so that other antigen delivery strategies are required. This review provides an overview of existing antibacterial vaccines and novel approaches to vaccination with a focus on immunization against intracellular bacteria.
Collapse
Affiliation(s)
- Anke Osterloh
- Department of Infection Immunology, Research Center Borstel, Parkallee 22, 23845 Borstel, Germany
| |
Collapse
|
8
|
Vaillant L, Oster P, McMillan B, Orozco Fernandez E, Velin D. GM-CSF is key in the efficacy of vaccine-induced reduction of Helicobacter pylori infection. Helicobacter 2022; 27:e12875. [PMID: 35092634 PMCID: PMC9285700 DOI: 10.1111/hel.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/22/2021] [Accepted: 01/16/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) colonizes the human gastric mucosa with a high worldwide prevalence. Currently, H. pylori is eradicated by the use of antibiotics. However, elevated antibiotic resistance suggests new therapeutic strategies need to be envisioned: one approach being prophylactic vaccination. Pre-clinical and clinical data show that a urease-based vaccine is efficient in decreasing H. pylori infection through the mobilization of T helper (Th) cells, especially Th17 cells. Th17 cells produce interleukins such as IL-22 and IL-17, among others, and are key players in vaccine efficacy. Recently, granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing Th17 cells have been identified. AIM This study explores the possibility that GM-CSF plays a role in the reduction of H. pylori infection following vaccination. RESULTS We demonstrate that GM-CSF+ IL-17+ Th17 cells accumulate in the stomach mucosa of H. pylori infected mice during the vaccine-induced reduction of H. pylori infection. Secondly, we provide evidence that vaccinated GM-CSF deficient mice only modestly reduce H. pylori infection. Conversely, we observe that an increase in GM-CSF availability reduces H. pylori burden in chronically infected mice. Thirdly, we show that GM-CSF, by acting on gastric epithelial cells, promotes the production of βdefensin3, which exhibits H. pylori bactericidal activities. CONCLUSION Taken together, we demonstrate a key role of GM-CSF, most probably originating from Th17 cells, in the vaccine-induced reduction of H. pylori infection.
Collapse
Affiliation(s)
- Laurie Vaillant
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Paul Oster
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Brynn McMillan
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Eulalia Orozco Fernandez
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Dominique Velin
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
9
|
A Novel Design of Multi-epitope Vaccine Against Helicobacter pylori by Immunoinformatics Approach. Int J Pept Res Ther 2021; 27:1027-1042. [PMID: 33424523 PMCID: PMC7778422 DOI: 10.1007/s10989-020-10148-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 12/18/2022]
Abstract
Helicobacter pylori (H. pylori) is a gram-negative spiral bacterium that caused infections in half of the world’s population and had been identified as type I carcinogen by the World Health Organization. Compared with antibiotic treatment which could result in drug resistance, the vaccine therapy is becoming a promising immunotherapy option against H. pylori. Further, the multi-epitope vaccine could provoke a wider immune protection to control H. pylori infection. In this study, the in-silico immunogenicity calculations on 381 protein sequences of H. pylori were performed, and the immunogenicity of selected proteins with top-ranked score were tested. The B cell epitopes and T cell epitopes from three well performed proteins UreB, PLA1, and Omp6 were assembled into six constructs of multi-epitope vaccines with random orders. In order to select the optimal constructs, the stability of the vaccine structure and the exposure of B cell epitopes on the vaccine surface were evaluated based on structure prediction and solvent accessible surface area analysis. Finally Construct S1 was selected and molecular docking showed that it had the potential of binding TLR2, TLR4, and TLR9 to stimulate strong immune response. In particular, this study provides good suggestions for epitope assembly in the construction of multi-epitope vaccines and it may be helpful to control H. pylori infection in the future.
Collapse
|
10
|
Keikha M, Eslami M, Yousefi B, Ghasemian A, Karbalaei M. Potential antigen candidates for subunit vaccine development against
Helicobacter pylori
infection. J Cell Physiol 2019; 234:21460-21470. [PMID: 31188484 DOI: 10.1002/jcp.28870] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Masoud Keikha
- Antimicrobial Resistance Research Center, Bu‐Ali Research Institute Mashhad University of Medical Sciences Mashhad Iran
- Department of Microbiology and Virology, School of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Majid Eslami
- Cancer Research Center Semnan University of Medical Sciences Semnan Iran
| | - Bahman Yousefi
- Department of Immunology Semnan University of Medical Sciences Semnan Iran
| | - Abdolmajid Ghasemian
- Department of Biology, Tehran Central Branch Islamic Azad University Tehran Iran
| | - Mohsen Karbalaei
- Department of Microbiology and Virology, School of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Department of Microbiology and Virology, School of Medicine Jiroft University of Medical Sciences Jiroft Iran
| |
Collapse
|
11
|
Takeshita H, Watanabe E, Norose Y, Ito Y, Takahashi H. Neutralizing antibodies for Helicobacter pylori urease inhibit bacterial colonization in the murine stomach in vivo. Biomed Res 2019; 40:87-95. [PMID: 30982804 DOI: 10.2220/biomedres.40.87] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Helicobacter pylori (H. pylori) urease is a key protein for persistent infection of the bacteria in the stomach. Although H. pylori generally induce anti-H. pylori-specific antibodies (Abs), these Abs do not usually work for eradication or prevention of the H. pylori infection. In our previous study, we identified a linear epitope composed of 19-mer peptides termed UB-33, CHHLDKSIKEDVQFADSRI, within the large subunit of H. pylori urease. Anti-UB-33-specific Abs neutralized the enzymatic activity of H. pylori urease in vitro. In the present study, we evaluated the effect of immunization of BALB/c mice with H. pylori UB-33 peptide. After confirming the production of anti-UB-33-specific Abs, mice were challenged orally with H. pylori Sydney Strain-1 (SS-1). Mice producing anti-UB-33-specific Abs were not infected with SS-1, and the amount of SS-1 isolate in their stomach was significantly reduced. Also, the urease-negative mutant of H. pylori, HPP1801, did not colonize in the stomach, indicating that H. pylori urease was a critical element for infection of H. pylori in the gastric mucosa. Moreover, mice producing UB-33-specific Abs apparently suppressed H. pylori infection in the stomach where anti-UB-33 Abs were secreted in the gastric juice, indicating that H. pylori colonization was inhibited in the presence of anti-UB-33 Abs. In addition, the neutralization activity of sera from mice immunized with purified urease was less potent than that in the sera from mice immunized with UB-33. Furthermore, the recognition of epitope UB-33 was mediated through Toll-like receptor 2 (TLR2) on the B-1 cells using TLR2-knockout BALB/c mice in vivo. These results indicate that liner peptide UB-33 should be used for immunization to induce neutralizing Abs instead of purified H. pylori urease to prevent H. pylori infection and their colonization in the stomach.
Collapse
Affiliation(s)
- Hikaru Takeshita
- Department of Microbiology and Immunology, Nippon Medical School.,Department of Pediatrics, Nippon Medical School
| | - Eri Watanabe
- Department of Microbiology and Immunology, Nippon Medical School
| | - Yoshihiko Norose
- Department of Microbiology and Immunology, Nippon Medical School
| | | | - Hidemi Takahashi
- Department of Microbiology and Immunology, Nippon Medical School
| |
Collapse
|
12
|
Jackson DG. Leucocyte Trafficking via the Lymphatic Vasculature- Mechanisms and Consequences. Front Immunol 2019; 10:471. [PMID: 30923528 PMCID: PMC6426755 DOI: 10.3389/fimmu.2019.00471] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/21/2019] [Indexed: 01/15/2023] Open
Abstract
The lymphatics fulfill a vital physiological function as the conduits through which leucocytes traffic between the tissues and draining lymph nodes for the initiation and modulation of immune responses. However, until recently many of the molecular mechanisms controlling such migration have been unclear. As a result of careful research, it is now apparent that the process is regulated at multiple stages from initial leucocyte entry and intraluminal crawling in peripheral tissue lymphatics, through to leucocyte exit in draining lymph nodes where the migrating cells either participate in immune responses or return to the circulation via efferent lymph. Furthermore, it is increasingly evident that most if not all leucocyte populations migrate in lymph and that such migration is not only important for immune modulation, but also for the timely repair and resolution of tissue inflammation. In this article, I review the latest research findings in these areas, arising from new insights into the distinctive ultrastructure of lymphatic capillaries and lymph node sinuses. Accordingly, I highlight the emerging importance of the leucocyte glycocalyx and its novel interactions with the endothelial receptor LYVE-1, the intricacies of endothelial chemokine secretion and sequestration that direct leucocyte trafficking and the significance of the process for normal immune function and pathology.
Collapse
Affiliation(s)
- David G Jackson
- MRC Human Immunology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Mohit E, Nasr R, Ghazvini K, Bandegi AR, Akbari Eidgahi MR. Evaluation of the Effect of Promoter Type on the Immunogenicity of the Live Recombinant Salmonella Vaccines Expressing Escherichia Coli Heat-labile Enterotoxins (LTB). IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:98-110. [PMID: 31086557 PMCID: PMC6447867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Enterotoxigenic Escherichia coli (ETEC)-induced diarrhoea is the second most common cause of death in children in the developing countries. Heat labile toxin (LT) is responsible for ETEC-induced diarrhoea. In the present study, a novel live ETEC vaccine based on subunit B of LT (LTB) expression in attenuated PhoPc Salmonella strain was developed. Herein, we aimed to compare the in-vitro activity of promoters including constitutive tac, IPTG inducible trc, and in-vivo-inducible (nirB and nirB78-23) in PhoPc. Additionally, the ability of these recombinant PhoPc/pLTBs to induce LTB-specific antibody responses in BALB/c mice after nasal immunization was evaluated. In-vitro studies demonstrated that PhoPc has the ability to produce rLTB. Furthermore, nirB promoter directed significantly more LTB expression in PhoPc/pnirBLTB under anaerobic condition without induction compared to the amount of rLTB secreted by PhoPc/ptrcLTB in bacterial soup under uninduced condition (6.06 ± 0.05 vs. 1.4 ± 0.46 μg/109 cfu, p < 0.01). In addition, the constitutive rLTB expression from tac promoter was more than its expression from uninduced trc promoter in bacterial soup (4.2 ± 0.92 vs. 1.4 ± 0.46 (μg/109 cfu)) and pellet (27.4 ± 0.89 vs. 13.4 ± 1.42 (μg/109 cfu), p < 0.0001). However, the mice immunized with PhoPc/ptrcLTB elicited the superior anti-LTB responses among the PhoPc containing the examined prompters, which were significantly higher than those induced by PhoPc/pnirB78-23LTB and PhoPc/pnirB, 6 weeks after the first immunization. Totally, it could be concluded that in-vitro analysis of promoters for LTB expression in PhoPc may not necessarily predict the recombinant PhoPc immunogenicity.
Collapse
Affiliation(s)
- Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Nasr
- Biotechnology Research Center, Department of Biotechnolog, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Kiarash Ghazvini
- Department of Biochemistry, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Ahmad Reza Bandegi
- Biotechnology Research Center, Department of Biotechnolog, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran. ,Corresponding author: E-mail:
| | | |
Collapse
|
14
|
Development of an arCagA Antigen-Based Assay for the Detection of Helicobacter pylori in Stool Specimens. Jundishapur J Microbiol 2016. [DOI: 10.5812/jjm.39119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
15
|
von Bargen K, Gagnaire A, Arce-Gorvel V, de Bovis B, Baudimont F, Chasson L, Bosilkovski M, Papadopoulos A, Martirosyan A, Henri S, Mège JL, Malissen B, Gorvel JP. Cervical Lymph Nodes as a Selective Niche for Brucella during Oral Infections. PLoS One 2015; 10:e0121790. [PMID: 25919005 PMCID: PMC4412401 DOI: 10.1371/journal.pone.0121790] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/03/2015] [Indexed: 12/23/2022] Open
Abstract
Cervical lymph nodes (CLN) are the first lymph nodes encountered by material taking the oral route. To study their role in orally acquired infections, we analyzed 307 patients of up to 14 years treated in the university clinic of Skopje, Macedonia, for brucellosis, a zoonotic bacterial disease frequently acquired by ingestion of contaminated dairy products. From these children, 36% had lymphadenopathy. Among orally infected children, lymphadenopathy with CLN being the only lymph nodes affected was significantly more frequent as compared to those infected by contact with animals (83% vs. 63%), suggesting a possible involvement of CLN during orally acquired human brucellosis. Using a murine model where bacteria are delivered into the oral cavity, we show that Brucella quickly and selectively colonize the CLN where they proliferate and persist over long periods of time for up to 50 days post-infection. A similar efficient though less specific drainage to CLN was found for Brucella, Salmonella typhimurium and fluorescent microspheres delivered by gavage, a pathway likely representing a mixed infection mode of intragastric and oral infection, suggesting a central pathway of drained material. Microspheres as well as bacteria drained to CLN predominately reside in cells expressing CD68 and no or low levels of CD11c. Even though no systemic response could be detected, Brucella induced a locally restricted inflammatory reaction with increased expression levels of interferon γ, interleukin (IL)-6, IL-12, granzyme B and a delayed induction of Nos2. Inflammation led to pronounced lymphadenopathy, infiltration of macrophages/monocytes expressing high levels of major histocompatibility complex II and to formation of epitheloid granulomas. Together, these results highlight the role of CLN in oral infections as both, an initial and efficient trap for bacterial invaders and as possible reservoir for chronic pathogens. They likewise cast a new light on the significance of oral routes for means of vaccination.
Collapse
Affiliation(s)
- Kristine von Bargen
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Aurélie Gagnaire
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Vilma Arce-Gorvel
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Béatrice de Bovis
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Fannie Baudimont
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Lionel Chasson
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Mile Bosilkovski
- University Clinic for Infectious Diseases and Febrile Conditions, Skopje, Republic of Macedonia
| | - Alexia Papadopoulos
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Anna Martirosyan
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Jean-Louis Mège
- Unité des Rickettsies, Aix-Marseille University, Centre National de la Recherche Scientifique (CNRS), UMR6020, Faculté de Médecine, Marseille, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
- * E-mail:
| |
Collapse
|
16
|
|
17
|
|
18
|
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
|
19
|
Kaparakis-Liaskos M, Tate MD, Price JD, Pearse M, Wijburg OLC. Increased antigen specific T cell numbers in the absence of altered migration or division rates as a result of mucosal cholera toxin administration. PLoS One 2013; 8:e59934. [PMID: 23544110 PMCID: PMC3609821 DOI: 10.1371/journal.pone.0059934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 02/22/2013] [Indexed: 11/18/2022] Open
Abstract
Cholera toxin (CT) is a mucosal adjuvant capable of inducing strong immune responses to co-administered antigens following oral or intranasal immunization of mice. To date, the direct effect of CT on antigen-specific CD4(+) T cell migration and proliferation profiles in vivo is not well characterized. In this study, the effect of CT on the migration pattern and proliferative responses of adoptively transferred, CD4(+) TCR transgenic T cells in orally or intranasally vaccinated mice, was analyzed by flow cytometry. GFP-expressing or CFSE-labeled OT-II lymphocytes were adoptively transferred to naïve C57BL/6 mice, and mice were subsequently vaccinated with OVA with or without CT via the oral or intranasal route. CT did not alter the migration pattern of antigen-specific T cells, regardless of the route of immunization, but increased the number of transgenic CD4(+) T cells in draining lymphoid tissue. This increase in the number of transgenic CD4(+) T cells was not due to cells undergoing more rounds of cellular division in vivo, suggesting that CT may exert an indirect adjuvant effect on CD4(+) T cells. The findings reported here suggest that CT functions as a mucosal adjuvant by increasing the number of antigen specific CD4(+) T cells independent of their migration pattern or kinetics of cellular division.
Collapse
Affiliation(s)
- Maria Kaparakis-Liaskos
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Clayton, Victoria, Australia
| | - Michelle D. Tate
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Clayton, Victoria, Australia
| | - Jason D. Price
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Odilia L. C. Wijburg
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
20
|
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
|
21
|
Li Y, Jiang Y, Xi Y, Zhang L, Luo J, He D, Zeng S, Ning Y. Identification and characterization of H-2d restricted CD4+ T cell epitopes on Lpp20 of Helicobacter pylori. BMC Immunol 2012; 13:68. [PMID: 23234363 PMCID: PMC3534527 DOI: 10.1186/1471-2172-13-68] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 11/30/2012] [Indexed: 12/12/2022] Open
Abstract
Background Previous investigation has demonstrated that CD4+ T cells play a crucial role in effective immunity against Helicobacter pylori (H.pylori) infection. It has been well proved that Lpp20 is one of major protective antigens that induce immune responses after H.pylori invades host. Therefore it is valuable to identify CD4+ T cell epitopes on Lpp20, which is uncharacterized. Methods Putative epitopes of H-2d restricted CD4+ T cell on Lpp20 of H.pylori were predicted by the SYFPEITHI algorithm and then eight hypothetical epitope peptides were synthesized. After BALB/c mice were primed with recombinant Lpp20, splenic CD4+ T cells were isolated and stimulated with synthesized peptides to measure T cell proliferation and MHC restriction. Cytokine profile was determined by ELISA and real-time PCR. Two identified epitopes were used to immunize mice to investigate CD4+ T cell response by flow cytometry. Results Two of eight peptides were able to stimulate CD4+ T cell proliferation and were mapped to residues 83-97aa and 58-72aa on Lpp20 respectively. These two peptides additively stimulated Th1 cells to secrete IFN-γ. The percentage of CD4+ T cell from mice immunized with two identified epitopes respectively was higher than the control group. Conclusion The identification and characterization of two CD4+ T cell epitopes of Lpp20 helps understand the protective immunity of Lpp20 in H.pylori infection and design effective epitope vaccines against H.pylori.
Collapse
Affiliation(s)
- Yan Li
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, North1838 Guangzhou Road, Guangzhou 510515, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Garai P, Gnanadhas DP, Chakravortty D. Salmonella enterica serovars Typhimurium and Typhi as model organisms: revealing paradigm of host-pathogen interactions. Virulence 2012; 3:377-88. [PMID: 22722237 PMCID: PMC3478240 DOI: 10.4161/viru.21087] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The lifestyle of intracellular pathogens has always questioned the skill of a microbiologist in the context of finding the permanent cure to the diseases caused by them. The best tool utilized by these pathogens is their ability to reside inside the host cell, which enables them to easily bypass the humoral immunity of the host, such as the complement system. They further escape from the intracellular immunity, such as lysosome and inflammasome, mostly by forming a protective vacuole-bound niche derived from the host itself. Some of the most dreadful diseases are caused by these vacuolar pathogens, for example, tuberculosis by Mycobacterium or typhoid fever by Salmonella. To deal with such successful pathogens therapeutically, the knowledge of a host-pathogen interaction system becomes primarily essential, which further depends on the use of a model system. A well characterized pathogen, namely Salmonella, suits the role of a model for this purpose, which can infect a wide array of hosts causing a variety of diseases. This review focuses on various such aspects of research on Salmonella which are useful for studying the pathogenesis of other intracellular pathogens.
Collapse
Affiliation(s)
- Preeti Garai
- Department of Microbiology and Cell Biology, Centre for Infectious Disease Research and Biosafety Laboratories, Indian Institute of Science, Bangalore, India
| | | | | |
Collapse
|
23
|
Avasthi TS, Ahmed N. Helicobacter pylori and type 1 diabetes mellitus: possibility of modifying chronic disease susceptibility with vaccinomics at the anvil. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2011; 15:589-96. [PMID: 21688972 DOI: 10.1089/omi.2010.0138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The human gastric pathogen, Helicobacter pylori, colonizes more than 50% of the world population and is a well-known cause of peptic ulcer disease. H. pylori has been epidemiologically linked to various other diseases, among which its putative link with certain complex diseases such as type 1 diabetes mellitus (T1DM) is of interest. Although antibiotic resistance is a significant clinical problem in H. pylori infection control, the exact cause and much of the underlying mechanisms of T1DM are not clearly understood. In addition, commensal microflora, gut-adapted microbial communities, and plausible roles of some of the chronic human pathogens add an important dimension to the control of T1DM. Given this, the present review attempts to analyze and examine the confounding association of H. pylori and T1DM and the approaches to tackle them, and how the emerging field of vaccinomics might help in this pursuit.
Collapse
Affiliation(s)
- Tiruvayipati Suma Avasthi
- Pathogen Biology Laboratory, Department of Biotechnology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | |
Collapse
|
24
|
Food-Grade Expression of Helicobacter pylori UreB Subunit in Lactococcus lactis and its Immunoreactivity. Curr Microbiol 2011; 62:1726-31. [PMID: 21431835 DOI: 10.1007/s00284-011-9920-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 03/04/2011] [Indexed: 01/31/2023]
|
25
|
Hinc K, Isticato R, Dembek M, Karczewska J, Iwanicki A, Peszyńska-Sularz G, De Felice M, Obuchowski M, Ricca E. Expression and display of UreA of Helicobacter acinonychis on the surface of Bacillus subtilis spores. Microb Cell Fact 2010; 9:2. [PMID: 20082702 PMCID: PMC2841587 DOI: 10.1186/1475-2859-9-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 01/18/2010] [Indexed: 01/02/2023] Open
Abstract
Background The bacterial endospore (spore) has recently been proposed as a new surface display system. Antigens and enzymes have been successfully exposed on the surface layers of the Bacillus subtilis spore, but only in a few cases the efficiency of expression and the effective surface display and have been determined. We used this heterologous expression system to produce the A subunit of the urease of the animal pathogen Helicobater acinonychis. Ureases are multi-subunit enzymes with a central role in the virulence of various bacterial pathogens and necessary for colonization of the gastric mucosa by the human pathogen H. pylori. The urease subunit UreA has been recognized as a major antigen, able to induce high levels of protection against challenge infections. Results We expressed UreA from H. acinonychis on the B. subtilis spore coat by using three different spore coat proteins as carriers and compared the efficiency of surface expression and surface display obtained with the three carriers. A combination of western-, dot-blot and immunofluorescence microscopy allowed us to conclude that, when fused to CotB, UreA is displayed on the spore surface (ca. 1 × 103 recombinant molecules per spore), whereas when fused to CotC, although most efficiently expressed (7-15 × 103 recombinant molecules per spore) and located in the coat layer, it is not displayed on the surface. Experiments with CotG gave results similar to those with CotC, but the CotG-UreA recombinant protein appeared to be partially processed. Conclusion UreA was efficiently expressed on the spore coat of B. subtilis when fused to CotB, CotC or CotG. Of these three coat proteins CotC allows the highest efficiency of expression, whereas CotB is the most appropriate for the display of heterologous proteins on the spore surface.
Collapse
Affiliation(s)
- Krzysztof Hinc
- Department of Structural and Functional Biology, Federico II University of Naples, via Cinthia 4, Naples 80126, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gao L, Weck MN, Michel A, Pawlita M, Brenner H. Association between chronic atrophic gastritis and serum antibodies to 15 Helicobacter pylori proteins measured by multiplex serology. Cancer Res 2009; 69:2973-80. [PMID: 19318564 DOI: 10.1158/0008-5472.can-08-3477] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Infection with Helicobacter pylori is a major risk factor for chronic atrophic gastritis (CAG), a precursor lesion of intestinal gastric cancer. The pathogenicity of the bacterium is thought to play an important role in determining the extent and severity of clinical outcome. We aimed to assess the associations between CAG and the serostatus of antibodies to 15 H. pylori proteins. The analyses were based on 534 cases with serologically defined CAG and 1,068 age-matched and sex-matched controls participating in a population-based study conducted in Saarland, Germany among 9,953 men and women ages 50 to 74 years. A newly developed H. pylori multiplex serology method was used to detect antibodies specific to 15 H. pylori antigens. Significant associations were observed between seropositivity for all 15 specific antibodies and the presence of CAG. Exclusion of severe cases, who might have lost the infection in the course of CAG progression, substantially increased the observed associations. In H. pylori-seropositive subjects, cytotoxin-associated gene A (CagA), vacuolating toxin (VacA), helicobacter cysteine-rich protein C (HcpC), and the chaperonin GroEL were identified as independent virulence factors for CAG with adjusted odds ratios (95% confidence interval) of 3.52 (2.01-6.10), 3.19 (1.44-7.05), 4.03 (1.53-10.65), and 2.65 (1.06-6.62), respectively; the simultaneous presence of all four independent virulence factors was associated with an 18-fold risk of CAG. In conclusion, HcpC and GroEL were identified as new independent virulence factors, and in combination with the established virulence factors, CagA and VacA, were strongly associated with CAG.
Collapse
Affiliation(s)
- Lei Gao
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
27
|
Li HX, Mao XH, Shi Y, Ma Y, Wu YN, Zhang WJ, Luo P, Yu S, Zhou WY, Guo Y, Wu C, Guo G, Zou QM. Screening and identification of a novel B-cell neutralizing epitope from Helicobacter pylori UreB. Vaccine 2008; 27:5013-9. [PMID: 18948159 DOI: 10.1016/j.vaccine.2009.05.009] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 04/24/2009] [Accepted: 05/04/2009] [Indexed: 02/08/2023]
Abstract
Urease plays a crucial role in the survival and pathogenesis of Helicobacter pylori (H. pylori), and antibody neutralizing the urease activity may be implicated for the protection against H. pylori infection. Previously, a neutralizing monoclonal antibody (MAb) 6E6 against UreB of H. pylori was developed. In this work, we try to identify the B-cell epitope recognized by neutralizing MAb 6E6. Following screening a series of truncated proteins of UreB, an epitope was primarily localized in the aa 200-230 of UreB. Subsequently, we screened the overlapping synthetic peptides covering the aa 200-230 and identified a novel B-cell epitope (U(211-225), IEAGAIGFKIHEDWG) that was recognized by specific MAb 6E6. The newly identified epitope may help understanding of the protective immunity against H. pylori and be implicated for vaccine development.
Collapse
Affiliation(s)
- Hai-Xia Li
- Department of Clinical Microbiology and Clinical Immunology, The Third Military Medical University, Chongqing 400038, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wu C, Shi Y, Guo H, Zou WY, Guo G, Xie QH, Mao XH, Tong WD, Zou QM. Protection against Helicobacter pylori infection in mongolian gerbil by intragastric or intramuscular administration of H. pylori multicomponent vaccine. Helicobacter 2008; 13:191-9. [PMID: 18466394 DOI: 10.1111/j.1523-5378.2008.00609.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Development of Helicobacter pylori vaccine would be a new effective strategy for prevention and treatment of H. pylori infection. Recombinant H. pylori vaccine comprising a single subunit antigen can only induce immune response with limited protection efficiency. In this study, the protective effect of H. pylori multicomponent vaccines consisting of three recombinant subunit antigens was investigated using the Mongolian gerbil model. MATERIALS AND METHODS Mongolian gerbils were immunized with different formulations of three recombinant H. pylori antigens (UreB, HspA, and HpaA) with two different adjuvants (Al(OH)3, LT(R72DITH)) by intragastric (i.g.) or intramuscular (i.m.) routes. The protective effects of multicomponent vaccines were assessed after H. pylori challenge in different studies. The specific IgG antibodies in serum were monitored by ELISA, and the mRNA expressions of IL-4 and IFN-gamma in spleen tissue were detected by reverse transcribed polymerase chain reaction (RT-PCR). RESULTS The protective effect against H. pylori challenge in gerbils immunized with three recombinant antigens and LT(R72DITH) or Al(OH)3 was significantly higher than that in single- or double-antigen vaccine-immunized and control gerbils. Furthermore, the protective effect of the triple-antigen vaccine combined with the LT(R72DITH) adjuvant (average 86.3%) was significantly greater than that of vaccine combined with the Al(OH)3 adjuvant (average 53.4%). After the first immunization, the anti-UreB/HspA/HpaA serum IgG level in gerbils immunized with triple-antigen vaccine combined with Al(OH)3 was higher than that in gerbils immunized with the vaccine combined with LT(R72DITH). Splenic interferon (IFN)-gamma and interleukin (IL)-4 transcript levels were significantly increased in LT(R72DITH) vaccine-immunized gerbils as compared to the Al(OH)3 vaccine group. Moreover, splenic IL-4 mRNA levels were higher than IFN-gamma in gerbils immunized with triple-antigen vaccine with either LT(R72DITH) or Al(OH)3. CONCLUSIONS This study indicated that the recombinant multicomponent vaccine provided effective protection against H. pylori infection as compared to the single-antigen vaccine. This protective immunity would be closely associated with a predominant Th2-type response.
Collapse
Affiliation(s)
- Chao Wu
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Abstract
Helicobacter pylori infection is highly prevalent worldwide and is an important cause of gastritis, peptic ulcer disease, gastric mucosa-associated lymphoid tissue lymphoma (MALToma), and gastric adenocarcinoma. Infection is usually acquired during childhood and tends to persist unless treated. Because eradication requires treatment with multidrug regimens, prevention of initial infection by a suitable vaccine is attractive. Although immunization with H pylori protein subunits has been encouraging in animals, similar vaccine trials in humans have shown adjuvant-related adverse effects and only moderate effectiveness. Newer immunization approaches (use of DNA, live vectors, bacterial ghosts, and microspheres) are being developed. Several questions about when and whom to vaccinate will need to be appropriately answered, and a cost-effective vaccine production and delivery strategy will have to be useful for developing countries. For this review, we searched MEDLINE using the Medical Subject Heading (MeSH) terms Helicobacter pylori and vaccines for articles in English from 1990 to 2007.
Collapse
Affiliation(s)
- Kanishtha Agarwal
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
31
|
Autenrieth SE, Autenrieth IB. Yersinia enterocolitica: subversion of adaptive immunity and implications for vaccine development. Int J Med Microbiol 2007; 298:69-77. [PMID: 17702651 DOI: 10.1016/j.ijmm.2007.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Enteric Yersinia spp. invade Peyer's patches, disseminate to lymphoid tissues, and induce mucosal and systemic immune responses. Many virulence factors of Yersinia enterocolitica have been investigated in detail and were found to act on host cells involved in innate and adaptive immunity. Recent work explored as to whether attenuated Y. enterocolitica or recombinant components of Y. enterocolitica can be used as tools for vaccination. We and others have tested whether by means of the type three secretion system in attenuated Y. enterocolitica strains antigens might be delivered to antigen-presenting cells in order to induce CD8 and CD4 T cell responses. Alternatively, recombinant components of Y. enterocolitica such as invasin protein which binds to beta1 integrins of host cells have been tested for their ability to target antigen along with microparticles (fused to invasin) to antigen-presenting cells and to act as adjuvant. The work summarized in this article demonstrates that Y. enterocolitica and its components might be useful tools for novel vaccination strategies; in fact, invasin when fused to antigen and coated to microparticles might induce both CD4 and CD8 T cell responses. Likewise, attenuated Y. enterocolitica live carrier strains were reported to induce both CD8 and some CD4 T cell responses. However, we need to know more about how Y. enterocolitica subverts functions of antigen-presenting cells in order to design mutants with optimized antigen delivery features and deletion in those virulence factor that contribute to subversion of innate or adaptive immune responses.
Collapse
Affiliation(s)
- Stella E Autenrieth
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen, Elfriede-Aulhorn-Strasse 6, D-72076 Tübingen, Germany
| | | |
Collapse
|
32
|
Abstract
Helicobacter pylori, a Gram-negative flagellate bacterium that infects the stomach of more than half of the global population, is regarded as the leading cause of chronic gastritis, peptic ulcer disease, and even gastric adenocarcinoma in some individuals. Although the bacterium induces strong humoral and cellular immune responses, it can persist in the host for decades. It has several virulence factors, some of them having vaccine potential as judged by immunoproteomic analysis. A few vaccination studies involving a small number of infected or uninfected humans with various H. pylori formulations such as the recombinant urease, killed whole cells, and live Salmonella vectors presenting the subunit antigens have not provided satisfactory results. One trial that used the recombinant H. pylori urease coadministered with native Escherichia coli enterotoxin (LT) demonstrated a reduction of H. pylori load in infected participants. Although extensive studies in the mouse model have demonstrated the feasibility of both therapeutic and prophylactic immunizations, the mechanism of vaccine-induced protection is poorly understood as several factors such as immunoglobulin and various cytokines do not contribute to protection. Transcriptome analyses in mice have indicated the role of nonclassical immune factors in vaccine-induced protection. The role of regulatory T cells in the persistence of H. pylori infection has also been suggested. A recently developed experimental H. pylori infection model in humans may be used for testing several new adjuvants and vaccine delivery systems that have been currently obtained. The use of vaccines with appropriate immunogens, routes of immunization, and adjuvants along with a better understanding of the mechanism of immune protection may provide more favorable results.
Collapse
Affiliation(s)
- Shahjahan Kabir
- Academic Research and Information Management, Uppsala, Sweden.
| |
Collapse
|
33
|
Shi Y, Wu C, Zhou WY, Mao XH, Guo G, Zou QM. Identification of H-2d restricted Th epitopes in Urease B subunit of Helicobacter pylori. Vaccine 2007; 25:2583-90. [PMID: 17240487 DOI: 10.1016/j.vaccine.2006.12.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2006] [Revised: 12/03/2006] [Accepted: 12/12/2006] [Indexed: 12/28/2022]
Abstract
CD4+ T cells play important roles in protection against Helicobacter pylori (H. pylori) infection. In order to better understand the immune responses of H. pylori infection and improve immune interventions against this pathogen, we identified the Th epitopes in UreB of H. pylori, an excellent vaccine candidate antigen. By using the RANKPEP prediction algorithm, we have identified and characterized three Th epitopes within the UreB antigen, which can be recognized by CD4+ T cells from BALB/c (H-2d) mice. They were U(546-561), U(229-244), and U(237-251). These epitopes have important value for studying the immune response of H. pylori infection and for designing effective vaccine against H. pylori.
Collapse
Affiliation(s)
- Yun Shi
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, The Third Military Medical University, Chongqing 400038, People's Republic of China
| | | | | | | | | | | |
Collapse
|
34
|
Nyström J, Svennerholm AM. Oral immunization with HpaA affords therapeutic protective immunity against H. pylori that is reflected by specific mucosal immune responses. Vaccine 2006; 25:2591-8. [PMID: 17239498 DOI: 10.1016/j.vaccine.2006.12.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2006] [Revised: 11/28/2006] [Accepted: 12/12/2006] [Indexed: 12/23/2022]
Abstract
In the present study, we evaluated the capacity of Helicobacter pylori adhesin A (HpaA), a H. pylori specific colonization factor, to induce therapeutic protection against H. pylori infection in mice. We found that oral immunization of H. pylori infected mice with HpaA induced protection, i.e. significant reduction in bacterial load in the stomach. This was even more pronounced when a combination of HpaA and urease was used. The protection was strongly related to specific mucosal CD4+ T cell responses with a Th1 profile as well as to mucosal IgA responses locally in the stomach. These findings suggest that HpaA is a promising vaccine candidate antigen for use in a therapeutic vaccine against H. pylori.
Collapse
Affiliation(s)
- Johanna Nyström
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy of Göteborg University, Box 435, 405 30 Göteborg, Sweden.
| | | |
Collapse
|
35
|
Kiama SG, Dreher D, Cochand L, Kok M, Obregon C, Nicod L, Gehr P. Host cell responses of Salmonella typhimurium infected human dendritic cells. Immunol Cell Biol 2006; 84:475-81. [PMID: 16869939 DOI: 10.1111/j.1440-1711.2006.01461.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Live attenuated Salmonella are attractive vaccine candidates for mucosal application because they induce both mucosal immune responses and systematic immune responses. After breaking the epithelium barrier, Salmonella typhimurium is found within dendritic cells (DC) in the Peyer's patches. Although there are abundant data on the interaction of S. typhimurium with murine epithelial cells, macrophages and DC, little is known about its interaction with human DC. Live attenuated S. typhimurium have recently been shown to efficiently infect human DC in vitro and induce production of cytokines. In this study, we have analysed the morphological consequences of infection of human DC by the attenuated S. typhimurium mutant strains designated PhoPc, AroA and SipB and the wild-type strains of the American Type Culture Collection (Manassas, VA, USA), ATCC 14028 and ATCC C53, by electron microscopy at 30 min, 3 h and 24 h after exposure. Our results show that genetic background of the strains profoundly influence DC morphology following infection. The changes included (i) membrane ruffling; (ii) formation of tight or spacious phagosomes; (iii) apoptosis; and (iv) spherical, pedunculated membrane-bound microvesicles that project from the plasma membrane. Despite the fact that membrane ruffling was much more pronounced with the two virulent strains, all mutants were taken up by the DC. The microvesicles were induced by all the attenuated strains, including SipB, which did not induce apoptosis in the host cell. These results suggest that Salmonella is internalized by human DC, inducing morphological changes in the DC that could explain immunogenicity of the attenuated strains.
Collapse
|
36
|
Wiedig CA, Kramer U, Garbom S, Wolf-Watz H, Autenrieth IB. Induction of CD8+ T cell responses by Yersinia vaccine carrier strains. Vaccine 2005; 23:4984-98. [PMID: 15985316 DOI: 10.1016/j.vaccine.2005.05.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Revised: 04/21/2005] [Accepted: 05/27/2005] [Indexed: 10/25/2022]
Abstract
Yersinia enterocolitica employs a type III secretion system (TTSS) to target virulence factors (e.g. YopE) into the cytosol of the host cells. We utilized the TTSS to introduce a recombinant antigen directly into the cytosol of host cells and to investigate the potential of Y. enterocolitica and Y. pseudotuberculosis as live carrier for vaccines. The model antigen ovalbumin (Ova) was fused to defined secretion or translocation domains of the Yersinia effector protein YopE and introduced into attenuated mutant strains of Y. enterocolitica and Y. pseudotuberculosis. In vitro experiments showed secretion and translocation of YopE-Ova hybrid proteins into host cells. To investigate the resulting immune responses, mice expressing transgenic Ova-specific T cell receptors were used. Both Y. enterocolitica and Y. pseudotuberculosis mutants induced efficaciously Ova-specific CD8+ T cell responses. The translocation domain of YopE was required for induction of CD8+ T cell responses in vivo, but not for T cell responses induced in vitro. The in vivo frequency of Ova-specific splenic T cells was up to six-fold higher in mice immunized with YopE-Ova-translocating Y. enterocolitica/Y. pseudotuberculosis mutants than in control mice. The Ova-specific T cells were shown to produce high amounts of IFN-gamma. We did not observe significant Ova-specific CD4+ T cell or antibody responses upon vaccination with either of the strains. In conclusion, Yersinia live carrier vaccine strains are suitable to target antigens into the MHC class I pathway and stimulate CD8+ T cell responses and thus, might be useful in vaccine approaches against intracellular pathogens.
Collapse
Affiliation(s)
- Carolin A Wiedig
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen, Elfriede-Aulhorn-Street 6, D-72076 Tübingen, Germany
| | | | | | | | | |
Collapse
|
37
|
Hikichi T, Kobayashi H, Oyama H, Yamamoto G, Watanabe H, Irisawa A, Obara K, Sato Y. Effectiveness of intragastric immunization with protein and oligodeoxynucleotides containing a CpG motif for inducing a gastrointestinal mucosal immune response in mice. Fukushima J Med Sci 2005; 51:19-31. [PMID: 16167670 DOI: 10.5387/fms.51.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE To investigate a new modality of mucosal vaccines, we evaluated the effectiveness of intragastric immunization for inducing a mucosal immune response in the gastrointestinal tract. METHODS Mice were immunized with beta-galactosidase (beta-gal) and synthesized oligodeoxynucleotides containing a CpG motif (CpG-DNA) by intragastric injection, and the immune response was compared with those induced by 3 other immunization forms: intranasal, oral, and intradermal. RESULTS Intragastric immunization with beta-gal and CpG-DNA induced significant anti-beta-gal fecal IgA production at 2 weeks; however, at 4 weeks the response was lacking. In contrast, intranasal immunization with beta-gal and CpG-DNA induced the highest anti-beta-gal fecal IgA production at 4 weeks. CONCLUSION Although intragastric immunization with protein and CpG-DNA induces a mucosal immune response in the gastrointestinal tract, intranasal immunization is the most effective to induce both mucosal and systemic immune responses. This finding may increase the possibility for developing vaccines against mucosal pathogens, especially Helicobacter pylori.
Collapse
Affiliation(s)
- Takuto Hikichi
- Department of Internal Medicine II, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Smythies LE, Novak MJ, Waites KB, Lindsey JR, Morrow CD, Smith PD. Poliovirus replicons encoding the B subunit of Helicobacter pylori urease protect mice against H. pylori infection. Vaccine 2005; 23:901-9. [PMID: 15603891 DOI: 10.1016/j.vaccine.2004.07.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2004] [Revised: 07/23/2004] [Accepted: 07/27/2004] [Indexed: 11/25/2022]
Abstract
We developed a novel vaccine for Helicobacter pylori based on a poliovirus vector in which capsid genes were replaced with the gene for the B subunit of H. pylori urease (UreB). Mice were vaccinated with UreB or control (L1) replicon and challenged with H. pylori. Twenty percent of mice vaccinated prophylactically with UreB, but 80% vaccinated with L1, and then challenged with H. pylori became infected (P = 0.003). Seventy-three percent of mice with established H. pylori infection vaccinated therapeutically with UreB replicon cleared their infection compared to 33% vaccinated with L1 (P = 0.067). In therapeutically vaccinated mice with residual infection, UreB-vaccinated animals had fewer H. pylori than L1-vaccinated mice (P < 0.05). Anti-urease antibody titres in prophylactically, but not therapeutically, vaccinated mice were markedly higher in animals that received UreB versus L1 replicon (P = 0.01). Vaccination with poliovirus vector containing the gene for the B subunit of H. pylori urease provides significant prophylactic and strong therapeutic protection against H. pylori in mice.
Collapse
Affiliation(s)
- Lesley E Smythies
- Department of Medicine (Gastroenterology), University of Alabama at Birmingham, ZRB 633, 703 19th Street South, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
40
|
Antigen Delivery Systems II: Development of Live Recombinant Attenuated Bacterial Antigen and DNA Vaccine Delivery Vector Vaccines. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50060-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Czinn SJ, Nedrud JG. Peptic Ulcers and Gastritis. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50073-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
42
|
Graham DY, Opekun AR, Osato MS, El-Zimaity HMT, Lee CK, Yamaoka Y, Qureshi WA, Cadoz M, Monath TP. Challenge model for Helicobacter pylori infection in human volunteers. Gut 2004; 53:1235-43. [PMID: 15306577 PMCID: PMC1774191 DOI: 10.1136/gut.2003.037499] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.7] [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 A reliable challenge model is needed to evaluate Helicobacter pylori vaccine candidates. METHODS A cag pathogenicity island negative, OipA positive, multiple antibiotic susceptible strain of H pylori obtained from an individual with mild gastritis (Baylor strain 100) was used to challenge volunteers. Volunteers received 40 mg of famotidine at bedtime and 10(4)-10(10) cfu of H pylori in beef broth the next morning. Infection was confirmed by (13)C urea breath test ((13)C-UBT), culture, and histology. Eradication therapy was given four or 12 weeks post challenge and eradication was confirmed by at least two separate UBTs, as well as culture and histology. RESULTS Twenty subjects (nine women and 11 men; aged 23-33 years) received a H pylori challenge. Eighteen (90%) became infected. Mild to moderate dyspeptic symptoms occurred, peaked between days 9 and 12, and resolved. Vomitus from one subject contained >10(3) viable/ml H pylori. By two weeks post challenge gastric histology showed typical chronic H pylori gastritis with intense acute and chronic inflammation. The density of H pylori (as assessed by cfu/biopsy) was similarly independent of the challenge dose. A minimal infectious dose was not found. Gastric mucosal interleukin 8 levels increased more than 20-fold by two weeks after the challenge. CONCLUSION Challenge reliably resulted in H pylori infection. Infection was associated with typical H pylori gastritis with intense polymorphonuclear cell infiltration and interleukin 8 induction in gastric mucosa, despite absence of the cag pathogenicity island. Experimental H pylori infection is one of the viable approaches to evaluate vaccine candidates.
Collapse
Affiliation(s)
- D Y Graham
- Veterans Affairs Medical Center, Rm 3A-320 (111D), 2002 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Johansson EL, Bergquist C, Edebo A, Johansson C, Svennerholm AM. Comparison of different routes of vaccination for eliciting antibody responses in the human stomach. Vaccine 2004; 22:984-90. [PMID: 15161075 DOI: 10.1016/j.vaccine.2003.09.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2002] [Revised: 08/28/2003] [Accepted: 09/05/2003] [Indexed: 11/27/2022]
Abstract
Determination of optimal routes to induce mucosal immune responses locally in the stomach and duodenum are important steps in the development of vaccines against Helicobacter pylori infection. In this study, we immunized H. pylori-infected individuals either nasally or rectally with a model antigen, i.e. cholera toxin B subunit, and compared the immune responses after these routes with the responses after oral or intrajejunal vaccination. Specific antibody levels in serum as well as specific antibody levels and antibody-secreting cells in biopsies from antrum and duodenum were determined by ELISA and ELISPOT methods. In contrast to oral vaccination, nasal and rectal vaccination did not induce significant increases in specific antibody-secreting cells either in the antrum or duodenum. Furthermore, when analyzing the antibody levels in saponin extracted biopsies, intrajejunal vaccination was superior to both nasal and rectal vaccination in inducing antigen-specific IgA levels in the stomach. We conclude that oral vaccination is the optimal route for induction of antigen-specific IgA antibody responses in the stomach and duodenum of humans, while nasal or rectal vaccination is less suitable for this purpose.
Collapse
Affiliation(s)
- Eva-Liz Johansson
- Department of Medical Microbiology and Immunology, Göteborg University Vaccine Institute, Göteborg University, Box 435, SE-405 30 Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
44
|
Chen M, Chen J, Liao W, Zhu S, Yu J, Leung WK, Hu P, Sung JJY. Immunization with attenuated Salmonella typhimurium producing catalase in protection against gastric Helicobacter pylori infection in mice. Helicobacter 2003; 8:613-25. [PMID: 14632677 DOI: 10.1111/j.1523-5378.2003.00182.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
AIM To evaluate the protective effect of live attenuated Salmonella typhimurium expressing catalase against gastric Helicobacter pylori infection in mice, and to explore the underlying mechanisms of the protective immune reaction. MATERIALS AND METHODS The H. pylori catalase gene was introduced into attenuated S. typhimurium strain SL3261. C57BL/6 mice were orally immunized with the SL3261 vaccine strain expressing catalase or with SL3261 alone or phosphate-buffered saline (PBS). Mice were sacrificed 4 weeks after immunization and 5 weeks after H. pylori challenge, respectively. RESULTS All PBS control mice were infected. Eight of 13 (61.5%) mice immunized with the SL3261 vaccine strain and three of 14 (21%) mice immunized with SL3261 alone showed protection against H. pylori infection. Serum anti-H. pylori IgG2a levels of S. typhimurium-immunized mice were higher than those of PBS controls, both before and after H. pylori challenge, while there were no differences for IgG1 and IgA. Similarly, mRNA expression of interleukin (IL)-2, IL-12 and interferon-gamma in the gastric mucosa of S. typhimurium-immunized mice was significantly higher than that of PBS controls both before and after challenge. Moreover, S. typhimurium-immunized mice were characterized by marked infiltration of lymphocyte and mononuclear cells in the gastric mucosa after challenge. IL-4 and IL-10 were not detected in any of the three groups. IL-6 expression was increased in the PBS group compared with the S. typhimurium-immunized groups after challenge. CONCLUSIONS This study demonstrates that oral immunization of mice with catalase delivered by an attenuated S. typhimurium strain offers protection against H. pylori infection. This protective immunity was mediated through a predominantly Th1-type response and was associated with post-immunization gastritis.
Collapse
Affiliation(s)
- Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guang Zhou, China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Rizos K, Lattemann CT, Bumann D, Meyer TF, Aebischer T. Autodisplay: efficacious surface exposure of antigenic UreA fragments from Helicobacter pylori in Salmonella vaccine strains. Infect Immun 2003; 71:6320-8. [PMID: 14573651 PMCID: PMC219551 DOI: 10.1128/iai.71.11.6320-6328.2003] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Live attenuated Salmonella strains expressing antigens of pathogens are promising oral vaccine candidates. There is growing evidence that the topology of expression of the foreign antigens can have a dramatic impact on the immunogenicity. We examined the potential of the AIDA-I (Escherichia coli adhesin involved in diffuse adherence) autotransporter domain to display antigenic fragments of the urease A subunit of Helicobacter pylori for the induction of a protective immune response. In the murine H. pylori model, protection is mainly mediated by CD4(+) T cells, and we therefore used the AIDA-I expression system to successfully express both nearly full-length UreA and defined T-helper-cell epitopes on the surface of an attenuated Salmonella enterica serovar Typhimurium vaccine strain. Surface exposure of the large UreA fragment or of one UreA T-cell epitope mediated a significant reduction in the level of H. pylori in immunized mice after challenge infection, whereas conventional cytoplasmic expression of UreA in Salmonella had no effect. These results support the concept that surface display increases the immunogenicity of recombinant antigens expressed on oral live vaccine carriers and further demonstrate the feasibility of immunizing against H. pylori with Salmonella vaccine strains expressing CD4(+) T-cell epitopes.
Collapse
Affiliation(s)
- Konstantin Rizos
- Creatogen AG, D-86156 Augsburg. Max-Planck-Institut für Infektionsbiologie, Abteilung Molekulare Biologie, D-10117 Berlin, Germany
| | | | | | | | | |
Collapse
|
46
|
Vajdy M, Singh M, Ugozzoli M, Briones M, Soenawan E, Cuadra L, Kazzaz J, Ruggiero P, Peppoloni S, Norelli F, Del Giudice G, O'Hagan D. Enhanced mucosal and systemic immune responses to Helicobacter pylori antigens through mucosal priming followed by systemic boosting immunizations. Immunology 2003; 110:86-94. [PMID: 12941145 PMCID: PMC1783019 DOI: 10.1046/j.1365-2567.2003.01711.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
It is estimated that Helicobacter pylori infects the stomachs of over 50% of the world's population and if not treated may cause chronic gastritis, peptic ulcer disease, gastric adenocarcinoma and gastric B-cell lymphoma. The aim of this study was to enhance the mucosal and systemic immune responses against the H. pylori antigens cytotoxin-associated gene A (CagA) and neutrophil-activating protein (NAP), through combinations of mucosal and systemic immunizations in female BALB/c mice. We found that oral or intranasal (i.n.) followed by i.m. immunizations induced significantly higher serum titres against NAP and CagA compared to i.n. alone, oral alone, i.m. alone, i.m. followed by i.n. or i.m. followed by oral immunizations. However, only oral followed by i.m. immunizations induced anti-NAP antibody-secreting cells in the stomach. Moreover, mucosal immunizations alone or in combination with i.m., but not i.m. immunizations alone, induced mucosal immunoglobulin A (IgA) responses in faeces. Any single route or combination of immunization routes with NAP and CagA preferentially induced antigen-specific splenic interleukin-4-secreting cells and far fewer interferon-gamma-secreting cells in the spleen. Moreover, i.n. immunizations alone or in combination with i.m. immunizations induced predominantly serum IgG1 and far less serum IgG2a. Importantly, we found that while both i.n. and i.m. recall immunizations induced similar levels of serum antibody responses, mucosal IgA responses in faeces were only achieved through i.n. recall immunization. Collectively, our data show that mucosal followed by systemic immunization significantly enhanced local and systemic immune responses and that i.n. recall immunization is required to induce both mucosal and systemic memory type responses.
Collapse
Affiliation(s)
- Michael Vajdy
- Immunology and Infectious DiseasesEmeryville, CA, USA
| | | | | | | | | | - Lina Cuadra
- Immunology and Infectious DiseasesEmeryville, CA, USA
| | - Jina Kazzaz
- Immunology and Infectious DiseasesEmeryville, CA, USA
| | | | | | | | | | - Derek O'Hagan
- Immunology and Infectious DiseasesEmeryville, CA, USA
| |
Collapse
|
47
|
Corthésy-Theulaz I, Corthésy B, Bachmann D, Velin D, Kraehenbuhl JP. Passive immunity in Helicobacter-challenged neonatal mice conferred by immunized dams lasts until weaning. Infect Immun 2003; 71:2226-9. [PMID: 12654845 PMCID: PMC152048 DOI: 10.1128/iai.71.4.2226-2229.2003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The objective of this study was to examine the effect of breast-feeding by immunized dams on Helicobacter colonization in newborns. Urease-based immunization regimens failed to protect nursing pups against H. felis, whereas H. felis lysate-cholera toxin resulted in protection. This observation correlated with a high recognition of cell surface-expressed bacterial antigens by milk antibodies. Protection lasted until weaning, indicating that infection is maintained at undetectable levels by passive immunity but then resumes when breast-feeding stops.
Collapse
Affiliation(s)
- Irène Corthésy-Theulaz
- Division of Gastroenterology, Nestlé Research Center, CHUV Lausanne, CH-1000 Lausanne 26, Switzerland.
| | | | | | | | | |
Collapse
|
48
|
Di Genaro MS, Waidmann M, Kramer U, Hitziger N, Bohn E, Autenrieth IB. Attenuated Yersinia enterocolitica mutant strains exhibit differential virulence in cytokine-deficient mice: implications for the development of novel live carrier vaccines. Infect Immun 2003; 71:1804-12. [PMID: 12654794 PMCID: PMC152075 DOI: 10.1128/iai.71.4.1804-1812.2003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Yersinia enterocolitica mutant strains, including mutants deficient in the chaperone SycH resulting in a functional deficiency in tyrosine phosphatase (YopH), Mn-cofactored superoxide dismutase (SodA), iron-repressive protein 1 (IRP-1), and Yersinia adhesin A (YadA), were demonstrated to be highly attenuated in wild-type C57BL/6 mice. TNFRp55(-/-), IL-12p40(-/-), and IL-18(-/-) mutant mice, in which the Yersinia wild-type strain causes severe systemic infections, were used to investigate whether these Yersinia mutant strains would be attenuated in immunodeficient hosts. A plasmid-cured Yersinia mutant strain was unable to colonize any of the mutant mice tested. A SycH-deficient mutant strain colonized intestinal tissues of these mice but was attenuated for systemic infection in all of the mutant mice. Both YadA- and Irp-1-deficient Yersinia mutants were still attenuated in IL-12(-/-) and IL-18(-/-) mice but were pathogenic in TNFRp55(-/-) mice. By contrast, a Yersinia sodA mutant was highly pathogenic for TNFRp55(-/-) and IL-12p40(-/-) mice while interleukin-18 (IL-18) was dispensable. This finding demonstrates that certain virulence factors enable yersiniae to compete with distinct cytokine-dependent host defense mechanisms. Moreover, while gamma interferon mRNA expression did not reflect protective host responses in cytokine-deficient mice, IL-10 expression coincided with a heavy splenic bacterial load and was associated with progressive infection courses. We can thus segregate minor (SodA), intermediate (YadA and IRP-1), and major (YopH) virulence factors of Y. enterocolitica. Finally, we demonstrate that, even in immunocompromised hosts, Yersinia sycH and, with some restrictions, irp-1 mutants may be suitable for use as live carrier vaccines.
Collapse
Affiliation(s)
- María S Di Genaro
- Max von Pettenkofer Institute for Hygiene and Medical Microbiology, Ludwig Maximilians University Munich, 80336 Munich, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Pasetti MF, Levine MM, Sztein MB. Animal models paving the way for clinical trials of attenuated Salmonella enterica serovar Typhi live oral vaccines and live vectors. Vaccine 2003; 21:401-18. [PMID: 12531639 DOI: 10.1016/s0264-410x(02)00472-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Attenuated Salmonella enterica serovar Typhi (S. Typhi) strains can serve as safe and effective oral vaccines to prevent typhoid fever and as live vectors to deliver foreign antigens to the immune system, either by the bacteria expressing antigens through prokaryotic expression plasmids or by delivering foreign genes carried on eukaryotic expression systems (DNA vaccines). The practical utility of such live vector vaccines relies on achieving a proper balance between minimizing the vaccine's reactogenicity and maximizing its immunogenicity. To advance to clinical trials, vaccine candidates need to be pre-clinically evaluated in relevant animal models that attempt to predict what their safety and immunogenicity profile will be when administered to humans. Since S. Typhi is a human-restricted pathogen, a major obstacle that has impeded the progress of vaccine development has been the shortcomings of the animal models available to assess vaccine candidates. In this review, we summarize the usefulness of animal models in the assessment of the degree of attenuation and immunogenicity of novel attenuated S. Typhi strains as vaccine candidates for the prevention of typhoid fever and as live vectors in humans.
Collapse
Affiliation(s)
- Marcela F Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine, Room 480, 685 West Baltimore Street, Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
50
|
Panthel K, Jechlinger W, Matis A, Rohde M, Szostak M, Lubitz W, Haas R. Generation of Helicobacter pylori ghosts by PhiX protein E-mediated inactivation and their evaluation as vaccine candidates. Infect Immun 2003; 71:109-16. [PMID: 12496155 PMCID: PMC143412 DOI: 10.1128/iai.71.1.109-116.2003] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Bacterial ghosts are empty cell envelopes, which may be generated by the controlled expression of the PhiX174 lysis gene E in gram-negative bacteria to obtain vaccine candidates. We describe here the application of this technology to Helicobacter pylori. The lysis gene cassette was cloned into an Escherichia coli-Helicobacter pylori shuttle vector and introduced into an H. pylori recipient strain by bacterial conjugation. Temperature induction of the lysis gene cassette revealed a quantitative killing of the H. pylori culture without induction of lysis-resistant bacteria. Biochemical and transmission electron microscopic studies identified structurally intact H. pylori. Prophylactic oral vaccination experiments using these H. pylori ghosts in the BALB/c mouse model showed a significant reduction of the bacterial load in the ghost group, as measured by a quantitative bacterial reisolation procedure. Ten of 10 and 5 of 10 mice were protected, respectively, without the use of a mucosal adjuvant. Coadministration of ghosts with cholera toxin as mucosal adjuvant resulted in a complete protection of 10 of 10 and 8 of 8 mice against H. pylori challenge, with three animals showing a sterile immunity.
Collapse
Affiliation(s)
- Klaus Panthel
- Max von Pettenkofer Institute for Hygiene and Medical Microbiology, LMU Munich, Germany
| | | | | | | | | | | | | |
Collapse
|