1
|
Sarkar S, Alipour Talesh G, Menheniott TR, Sutton P. Targeting Host Sulphonyl Urea Receptor 2 Can Reduce Severity of Helicobacter pylori Associated Gastritis. GASTRO HEP ADVANCES 2023; 2:721-732. [PMID: 39129876 PMCID: PMC11307979 DOI: 10.1016/j.gastha.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 03/06/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims While most Helicobacter pylori-infected individuals remain asymptomatic throughout their lifetime, in a significant proportion, the resulting severe chronic gastritis drives the development of gastric cancer. In this study, we examine a new therapeutic target, a host potassium channel regulatory subunit, SUR2 (encoded by ABCC9), with potential to protect against H pylori-associated diseases. Methods SUR2 gene (ABCC9) expression in human gastric biopsies was analyzed by quantitative polymerase chain reactions. Helicobacter-infected mice were administered the SUR2-channel agonists, pinacidil and nicorandil, then gastric tissues analyzed by histology, immunohistochemistry and quantitative polymerase chain reaction, and splenic tissues by enzyme-linked immunosorbent assays. In vitro studies were performed on human and mouse macrophages, human gastric epithelial cells and mouse splenocytes. Results ABCC9 expression in human and mouse stomachs is downregulated with H pylori infection. Treatment of Helicobacter-infected mice with SUR2 channel modulators, pinacidil or nicorandil, significantly reduced gastritis severity. In gastric epithelial cells, nicorandil-induced opening of the SUR2 channel increased intracellular K+ and prevented H pylori-mediated Ca2+ influx and downstream pro-inflammatory signaling. Conclusion SUR2 is a novel host factor that regulates Helicobacter pathogenesis. Pharmacological targeting of SUR2 provides a potential approach for reducing the severity of H pylori-associated gastritis, without eradicating infection.
Collapse
Affiliation(s)
- Sohinee Sarkar
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Ghazal Alipour Talesh
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Trevelyan R. Menheniott
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Philip Sutton
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
2
|
Taillieu E, Chiers K, Amorim I, Gärtner F, Maes D, Van Steenkiste C, Haesebrouck F. Gastric Helicobacter species associated with dogs, cats and pigs: significance for public and animal health. Vet Res 2022; 53:42. [PMID: 35692057 PMCID: PMC9190127 DOI: 10.1186/s13567-022-01059-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/10/2022] [Indexed: 12/14/2022] Open
Abstract
This article focuses on the pathogenic significance of Helicobacter species naturally colonizing the stomach of dogs, cats and pigs. These gastric "non-Helicobacter (H.) pylori Helicobacter species" (NHPH) are less well-known than the human adapted H. pylori. Helicobacter suis has been associated with gastritis and decreased daily weight gain in pigs. Several studies also attribute a role to this pathogen in the development of hyperkeratosis and ulceration of the non-glandular stratified squamous epithelium of the pars oesophagea of the porcine stomach. The stomach of dogs and cats can be colonized by several Helicobacter species but their pathogenic significance for these animals is probably low. Helicobacter suis as well as several canine and feline gastric Helicobacter species may also infect humans, resulting in gastritis, peptic and duodenal ulcers, and low-grade mucosa-associated lymphoid tissue lymphoma. These agents may be transmitted to humans most likely through direct or indirect contact with dogs, cats and pigs. Additional possible transmission routes include consumption of water and, for H. suis, also consumption of contaminated pork. It has been described that standard H. pylori eradication therapy is usually also effective to eradicate the NHPH in human patients, although acquired antimicrobial resistance may occasionally occur and porcine H. suis strains are intrinsically less susceptible to aminopenicillins than non-human primate H. suis strains and other gastric Helicobacter species. Virulence factors of H. suis and the canine and feline gastric Helicobacter species include urease activity, motility, chemotaxis, adhesins and gamma-glutamyl transpeptidase. These NHPH, however, lack orthologs of cytotoxin-associated gene pathogenicity island and vacuolating cytotoxin A, which are major virulence factors in H. pylori. It can be concluded that besides H. pylori, gastric Helicobacter species associated with dogs, cats and pigs are also clinically relevant in humans. Although recent research has provided better insights regarding pathogenic mechanisms and treatment strategies, a lot remains to be investigated, including true prevalence rates, exact modes of transmission and molecular pathways underlying disease development and progression.
Collapse
Affiliation(s)
- Emily Taillieu
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Koen Chiers
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Irina Amorim
- Instituto de Investigação E Inovação Em Saúde (i3S), Universidade Do Porto, Porto, Portugal.,Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), Porto, Portugal.,School of Medicine and Biomedical Sciences, Porto University, Porto, Portugal
| | - Fátima Gärtner
- Instituto de Investigação E Inovação Em Saúde (i3S), Universidade Do Porto, Porto, Portugal.,Institute of Pathology and Molecular Immunology, University of Porto (IPATIMUP), Porto, Portugal
| | - Dominiek Maes
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Christophe Van Steenkiste
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Antwerp University, Edegem, Belgium.,Department of Gastroenterology and Hepatology, General Hospital Maria Middelares, Ghent, Belgium
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
3
|
Cai Q, Shi P, Yuan Y, Peng J, Ou X, Zhou W, Li J, Su T, Lin L, Cai S, He Y, Xu J. Inflammation-Associated Senescence Promotes Helicobacter pylori-Induced Atrophic Gastritis. Cell Mol Gastroenterol Hepatol 2020; 11:857-880. [PMID: 33161156 PMCID: PMC7859172 DOI: 10.1016/j.jcmgh.2020.10.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS The association between cellular senescence and Helicobacter pylori-induced atrophic gastritis is not clear. Here, we explore the role of cellular senescence in H pylori-induced atrophic gastritis and the underlying mechanism. METHODS C57BL/6J mice were infected with H pylori for biological and mechanistic studies in vivo. Gastric precancerous lesions from patients and mouse models were collected and analyzed using senescence-associated beta-galactosidase, Sudan Black B, and immunohistochemical staining to analyze senescent cells, signaling pathways, and H pylori infection. Chromatin immunoprecipitation, luciferase reporter assays, and other techniques were used to explore the underlying mechanism in vitro. RESULTS Gastric mucosa atrophy was highly associated with cellular senescence. H pylori promoted gastric epithelial cell senescence in vitro and in vivo in a manner that depended on C-X-C motif chemokine receptor 2 (CXCR2) signaling. Interestingly, H pylori infection not only up-regulated the expression of CXCR2 ligands, C-X-C motif chemokine ligands 1 and 8, but also transcriptionally up-regulated the expression of CXCR2 via the nuclear factor-κB subunit 1 directly. In addition, CXCR2 formed a positive feedback loop with p53 to continually enhance senescence. Pharmaceutical inhibition of CXCR2 in an H pylori-infected mouse model attenuated mucosal senescence and atrophy, and delayed further precancerous lesion progression. CONCLUSIONS Our study showed a new mechanism of H pylori-induced atrophic gastritis through CXCR2-mediated cellular senescence. Inhibition of CXCR2 signaling is suggested as a potential preventive therapy for targeting H pylori-induced atrophic gastritis. GEO data set accession numbers: GSE47797 and GSE3556.
Collapse
Affiliation(s)
- Qinbo Cai
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China; Laboratory of General Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Peng Shi
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China; Laboratory of General Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yujie Yuan
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China
| | - Jianjun Peng
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China
| | - Xinde Ou
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China; Laboratory of General Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wen Zhou
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China; Laboratory of General Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jin Li
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China; Center for Digestive Disease, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Laboratory of General Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Taiqiang Su
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China; Center for Digestive Disease, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Laboratory of General Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liangliang Lin
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China
| | - Shirong Cai
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China
| | - Yulong He
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China; Center for Digestive Disease, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| | - Jianbo Xu
- Center of Gastrointestinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Diagnosis and Treatment of Gastric Cancer, Sun Yat-sen University, Guangdong, China.
| |
Collapse
|
4
|
Sayed IM, Sahan AZ, Venkova T, Chakraborty A, Mukhopadhyay D, Bimczok D, Beswick EJ, Reyes VE, Pinchuk I, Sahoo D, Ghosh P, Hazra TK, Das S. Helicobacter pylori infection downregulates the DNA glycosylase NEIL2, resulting in increased genome damage and inflammation in gastric epithelial cells. J Biol Chem 2020; 295:11082-11098. [PMID: 32518160 DOI: 10.1074/jbc.ra119.009981] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 05/30/2020] [Indexed: 01/08/2023] Open
Abstract
Infection with the Gram-negative, microaerophilic bacterium Helicobacter pylori induces an inflammatory response and oxidative DNA damage in gastric epithelial cells that can lead to gastric cancer (GC). However, the underlying pathogenic mechanism is largely unclear. Here, we report that the suppression of Nei-like DNA glycosylase 2 (NEIL2), a mammalian DNA glycosylase that specifically removes oxidized bases, is one mechanism through which H. pylori infection may fuel the accumulation of DNA damage leading to GC. Using cultured cell lines, gastric biopsy specimens, primary cells, and human enteroid-derived monolayers from healthy human stomach, we show that H. pylori infection greatly reduces NEIL2 expression. The H. pylori infection-induced downregulation of NEIL2 was specific, as Campylobacter jejuni had no such effect. Using gastric organoids isolated from the murine stomach in coculture experiments with live bacteria mimicking the infected stomach lining, we found that H. pylori infection is associated with the production of various inflammatory cytokines. This response was more pronounced in Neil2 knockout (KO) mouse cells than in WT cells, suggesting that NEIL2 suppresses inflammation under physiological conditions. Notably, the H. pylori-infected Neil2-KO murine stomach exhibited more DNA damage than the WT. Furthermore, H. pylori-infected Neil2-KO mice had greater inflammation and more epithelial cell damage. Computational analysis of gene expression profiles of DNA glycosylases in gastric specimens linked the reduced Neil2 level to GC progression. Our results suggest that NEIL2 downregulation is a plausible mechanism by which H. pylori infection impairs DNA damage repair, amplifies the inflammatory response, and initiates GC.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Ayse Z Sahan
- Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Tatiana Venkova
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Anirban Chakraborty
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Diane Bimczok
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, USA
| | - Ellen J Beswick
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Victor E Reyes
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Irina Pinchuk
- College of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, San Diego, California, USA.,Department of Computer Science and Engineering, Jacob's School of Engineering, San Diego, California, USA
| | - Pradipta Ghosh
- Department of Medicine and Cellular and Molecular Medicine, John and Rebecca Moore Cancer Center, University of California San Diego, San Diego, California, USA
| | - Tapas K Hazra
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Soumita Das
- Department of Pathology, University of California San Diego, San Diego, California, USA
| |
Collapse
|
5
|
Arshad U, Sarkar S, Alipour Talesh G, Sutton P. A lack of role for antibodies in regulating Helicobacter pylori colonization and associated gastritis. Helicobacter 2020; 25:e12681. [PMID: 32088936 DOI: 10.1111/hel.12681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Helicobacter pylori occupy a unique niche, located within the mucus layer lining the stomach, and attached to the apical surface of the gastric epithelium. As such, antibodies would be expected to play a major role in regulating infection and/or pathogenesis. However, experiments using antibody-deficient mice to study gastric helicobacter infection have yielded inconsistent results, although some pointed toward antibodies increasing colonization levels and decreasing gastritis severity. The variability in these studies is possibly due to their use of nonmatched wild-type controls. This current study presents the first evaluation of the role of antibodies in H pylori infection by comparing antibody-deficient mice with matched wild-type siblings. METHODS Matched wild-type and antibody-deficient μMT mice were generated by heterozygous crossings. In two separate experiments, appropriately genotyped sibling littermates were infected with H pylori for 4 months and then sera and stomachs were collected. RESULTS There was no difference in H pylori colonization levels between infected μMT mice and sibling wild-type controls. Similarly, there was no significant difference in the severity of gastritis between these groups of mice, although there was a trend toward less severe gastritis in μMT mice which was supported by a significantly lower IFNγ (Th1) gastric cytokine response. CONCLUSIONS Comparing matched antibody-deficient and antibody-competent mice indicates that an antibody response does not influence H pylori colonization levels. Contrary to previous studies, these results suggest antibodies might have a minor pro-inflammatory effect by promoting gastric Th1 cytokines, although this did not translate to a significant effect on gastritis severity.
Collapse
Affiliation(s)
- Umar Arshad
- Mucosal Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Vic., Australia
| | - Sohinee Sarkar
- Mucosal Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Vic., Australia
| | - Ghazal Alipour Talesh
- Mucosal Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Vic., Australia.,Department of Paediatrics, University of Melbourne, Parkville, Vic., Australia
| | - Philip Sutton
- Mucosal Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Vic., Australia.,Department of Paediatrics, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
6
|
Meyer TF, Morey P. A Future for a Vaccine Against the Cancer-Inducing Bacterium Helicobacter pylori? MUCOSAL VACCINES 2020:579-596. [DOI: 10.1016/b978-0-12-811924-2.00033-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Walduck AK, Raghavan S. Immunity and Vaccine Development Against Helicobacter pylori. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1149:257-275. [PMID: 31016627 DOI: 10.1007/5584_2019_370] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori is a highly-adapted gastrointestinal pathogen of humans and the immunology of this chronic infection is extremely complex. Despite the availability of antibiotic therapy, the global incidence of H. pylori infection remains high, particularly in low to middle-income nations. Failure of therapy and the spread of antibiotic resistance among the bacteria are significant problems and provide impetus for the development of new therapies and vaccines to treat or prevent gastric ulcer, and gastric carcinoma. The expansion of knowledge on gastric conventional and regulatory T cell responses, and the role of TH17 in chronic gastritis from studies in mouse models and patients have provided valuable insights into how gastritis is initiated and maintained. The development of human challenge models for testing candidate vaccines has meant a unique opportunity to study acute infection, but the field of vaccine development has not progressed as rapidly as anticipated. One clear lesson learned from previous studies is that we need a better understanding of the immune suppressive mechanisms in vivo to be able to design vaccine strategies. There is still an urgent need to identify practical surrogate markers of protection that could be deployed in future field vaccine trials. Important developments in our understanding of the chronic inflammatory response, progress and problems arising from human studies, and an outlook for the future of clinical vaccine trials will be discussed.
Collapse
Affiliation(s)
- Anna K Walduck
- School of Science, RMIT University, Melbourne, VIC, Australia.
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Lin L, Wei H, Yi J, Xie B, Chen J, Zhou C, Wang L, Yang Y. Chronic CagA-positive Helicobacter pylori infection with MNNG stimulation synergistically induces mesenchymal and cancer stem cell-like properties in gastric mucosal epithelial cells. J Cell Biochem 2019; 120:17635-17649. [PMID: 31209915 DOI: 10.1002/jcb.29031] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022]
Abstract
A CagA-positive Helicobacter pylori (H. pylori) infection can cause malignant transformation of human gastric mucosal epithelial cells, and N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) is a chemical carcinogen that induces gastric carcinogenesis. Whether this environmental chemocarcinogen may synergistically enhance the risk of H. pylori-infected gastric cancer remains unclear. In this study, we adopted a chronic CagA-positive H. pylori infection with or without MNNG coinduction to establish a cellular model in GES-1 cells and an animal model in C57BL/6J mice. The proliferation, cell phenotype, apoptosis, epithelial-mesenchymal transition (EMT), stemness and tumorigenicity of gastric mucosal epithelial cells were analyzed in vitro and in vivo. The results showed that chronic H. pylori-infected GES-1 cells displayed inhibited apoptosis, abnormal proliferation, enhanced invasion, and migration, increased EMT/mesenchymal phenotype, colony formation and stem cell-like properties, and enhanced tumorsphere-formatting efficiency as well as CD44 expression, a known gastric cancer stem cell (CSC) marker. MNNG synergistically promoted the above actions of chronic H. pylori infection. Further studies in chronic H. pylori-infected C57BL/6J mice models showed that an increased incidence of premalignant lesions in the gastric mucosa tissue of the H. pylori-infected mice had occurred, the mouse gastric mucosa cells exhibited similar mesenchymal and CSC-like properties in the above GES-1 cells, and precancerous lesions and EMT/CSC-like phenotypes were reinforced by the synergistic action of MNNG stimulation. H. pylori infection and/or MNNG induction were capable of causing enhanced expression and activation of Wnt2 and β-catenin, indicating that the Wnt/β-catenin pathway is involved in the actions of H. pylori and MNNG. Taken together, these findings suggest that chronic CagA-positive H. pylori infection with MNNG stimulation synergistically induces mesenchymal and CSC-like properties of gastric mucosal epithelial cells.
Collapse
Affiliation(s)
- Li Lin
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China.,Hematology Department, Gansu Provincial Cancer Hospital, Lanzhou, Gansu, China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Juan Yi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Bei Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Cunmin Zhou
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Li Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Yue Yang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
9
|
Liu W, Zeng Z, Luo S, Hu C, Xu N, Huang A, Zheng L, Sundberg EJ, Xi T, Xing Y. Gastric Subserous Vaccination With Helicobacter pylori Vaccine: An Attempt to Establish Tissue-Resident CD4+ Memory T Cells and Induce Prolonged Protection. Front Immunol 2019; 10:1115. [PMID: 31156652 PMCID: PMC6533896 DOI: 10.3389/fimmu.2019.01115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/01/2019] [Indexed: 12/22/2022] Open
Abstract
Tissue-resident memory T (Trm) cells are enriched at the sites of previous infection and required for enhanced protective immunity. However, the emergence of Trm cells and their roles in providing protection are unclear in the field of Helicobacter pylori (H. pylori) vaccinology. Here, our results suggest that conventional vaccine strategies are unable to establish a measurable antigen (Ag)-specific memory cell pool in stomach; in comparison, gastric subserous injection of mice with micro-dose of Alum-based H. pylori vaccine can induce a pool of local CD4+ Trm cells. Regional recruitment of Ag-specific CD4+ T cells depends on the engagement of Ag and adjuvant-induced inflammation. Prior subcutaneous vaccination enhanced this recruitment. A stable pool of Ag-specific CD4+ T cells can be detected for 240 days. Two weeks of FTY720 administration in immune mice suggests that these cells do not experience the recirculation. Immunohistochemistry results show that close to the vaccination site, abundant CD4+T cells locate on epithelial niches, independent of lymphocyte cluster. Paradigmatically, Ag-specific CD4+ T cells with a phenotype of CD69+CD103- are preferential on lymphocytes isolated from epithelium. Upon Helicobacter infection, CD4+ Trm cells orchestrate a swift recall response with the recruitment of circulating antigen-specific Th1/Th17 cells to trigger a tissue-wide pathogen clearance. This study investigates the vaccine-induced gastric CD4+ Trm cells in a mice model, and highlights the need for designing a vaccine strategy against H. pylori by establishing the protective CD4+ Trm cells.
Collapse
Affiliation(s)
- Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Chupeng Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Ningyin Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - An Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Eric J. Sundberg
- Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
10
|
Liu H, Liu W, Tan Z, Zeng Z, Yang H, Luo S, Wang L, Xi T, Xing Y. Promoting Immune Efficacy of the Oral Helicobacter pylori Vaccine by HP55/PBCA Nanoparticles against the Gastrointestinal Environment. Mol Pharm 2018; 15:3177-3186. [PMID: 30011213 DOI: 10.1021/acs.molpharmaceut.8b00251] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immunogenicity of oral subunit vaccines is poor partly as a result of the harsh milieu of the gastrointestinal (GI) tract. For some pathogens that restrictedly inhabit the GI tract, a vaccine that works in situ may provide more potent protection than vaccines that operate parenterally. Yet, no appropriate delivery system is available for oral subunit vaccines. In this study, we designed HP55/poly( n-butylcyanoacrylate) (PBCA) nanoparticles (NPs) to carry Helicobacter pylori ( H. pylori) subunit vaccine CCF for oral administration in a prophylactic mice model. These NPs, which are synthesized using an interfacial polymerization method, protected the CCF antigen not only from the acidic pH in simulated gastric fluid (SGF, pH 1.2) but also from the proteolysis in simulated intestinal fluid (SIF, pH 7.4). Oral vaccination of mice with HP55/PBCA-CCF NPs promoted the production of serum antigen-specific antibodies, mucosal secretory IgA, and proinflammatory cytokines. Moreover, a Th1/Th17 response and augmented lymphocytes were found in the gastric tissue of HP55/PBCA-CCF NP-immunized mice, which might eventually limit H. pylori colonization. Collectively, these results indicate that HP55/PBCA NPs are promising carriers against the severe situation of the GI tract and thereby may be further utilized for other orally administrated vaccines or drugs.
Collapse
Affiliation(s)
- Hai Liu
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Wei Liu
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Zhoulin Tan
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Zhiqin Zeng
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Huimin Yang
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Shuanghui Luo
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Linlin Wang
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Tao Xi
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| | - Yingying Xing
- School of Life Science and Technology and Jiangsu Key Laboratory of Carcinogenesis and Intervention , China Pharmaceutical University , No.24 Tongjia xiang , Nanjing 210009 , PR China
| |
Collapse
|
11
|
Sun H, Yuan H, Tan R, Li B, Guo G, Zhang J, Jing H, Qin Y, Zhao Z, Zou Q, Wu C. Immunodominant antigens that induce Th1 and Th17 responses protect mice against Helicobacter pylori infection. Oncotarget 2018; 9:12050-12063. [PMID: 29552292 PMCID: PMC5844728 DOI: 10.18632/oncotarget.23927] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/30/2017] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori has infected more than half of the world's population, causing gastritis, gastric ulcers, gastric mucosa-associated lymphoid tissue lymphoma and gastric cancer. The oral recombinant Helicobacter pylori vaccine currently used has made great progress in addressing this problem, however, its efficacy and longevity still need to be improved. Th1 and Th17 cells play essential roles in local protection against Helicobacter pylori in the stomach mucosa. Additionally, protective immunodominant antigens are the preferred for a vaccine. In this work, Helicobacter pylori whole cell lysate was separated into 30 groups based on molecular weight by molecular sieve chromatography. The group best promoting CD4 T cells proliferation was selected and evaluated by immunization. The detail proteins were then analyzed by LC-MS/MS and expressed in Escherichia coli. Eleven proteins were selected and the dominant ones were demonstrated. As a result, three protective immunodominant antigens, inosine 5'-monophosphate dehydrogenase, type II citrate synthase, and urease subunit beta, were selected from Helicobacter pylori whole cell. Two of them (inosine 5'-monophosphate dehydrogenase and type II citrate synthase) were newly identified, and one (urease subunit beta) was confirmed as previously reported. The mixture of the three antigens showed satisfactory protective efficiency, with significant lower H. pylori colonization level (P < 0.001) and stronger Th1 (P < 0.001) and Th17 (P < 0.001) responses than PBS control group. Thus, inosine 5'-monophosphate dehydrogenase, type II citrate synthase, and urease subunit beta are three protective antigens inducing dominant Th1 and Th17 responses to defend against Helicobacter pylori infection.
Collapse
Affiliation(s)
- Heqiang Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Hanmei Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Ranjing Tan
- Department of Dermatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Bin Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Gang Guo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Yi Qin
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Chao Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
12
|
Liu W, Tan Z, Liu H, Zeng Z, Luo S, Yang H, Zheng L, Xi T, Xing Y. Nongenetically modified Lactococcus lactis-adjuvanted vaccination enhanced innate immunity against Helicobacter pylori. Helicobacter 2017; 22. [PMID: 28805287 DOI: 10.1111/hel.12426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Gram-positive enhancer matrix particles (GEM) produced by Lactococcus lactis can enhance vaccine-induced immune response. However, the mechanism under which this adjuvant mounts the efficacy of orally administered vaccines remains unexplored. MATERIALS AND METHODS We used a prophylactic mice model to investigate the mechanism of GEM-adjuvanted vaccination. Helicobacter pylori urease-specific antibody response was monitored and detected in murine serum by ELISA. Urease-specific splenic cytokine profile was examined. Gastric inflammatory responses were measured on day 43 or 71 by quantitative real-time PCR, flow cytometry and histology. RESULTS We found that GEM enhanced the efficiency of oral H. pylori vaccine by promoting innate immunity. The vaccine CUE-GEM composed of GEM particles and recombinant antigen CTB-UE provided protection of immunized mice against H. pylori insult. The protective response was associated with induction of postimmunization gastritis and local Th1/Th17 cell-medicated immune response. We showed that innate inflammatory responses including neutrophil chemokines CXCL1-2, neutrophils, and antimicrobial proteins S100A8 and MUC1 were significantly elevated. Within all infected mice, S100A8 and MUC1 levels were negatively correlated with H. pylori burden. Strikingly, mice receiving GEM also show reduction of colonization, possibly through natural host response pathways to recruit CD4+ T cells and promote S100A8 expression. CONCLUSIONS These findings suggest that GEM-based vaccine may impact Th1/Th17 immunity to orchestrate innate immune response against H. pylori infection.
Collapse
Affiliation(s)
- Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhoulin Tan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Hai Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Huimin Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
The Human Stomach in Health and Disease: Infection Strategies by Helicobacter pylori. Curr Top Microbiol Immunol 2017; 400:1-26. [PMID: 28124147 DOI: 10.1007/978-3-319-50520-6_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori is a bacterial pathogen which commonly colonizes the human gastric mucosa from early childhood and persists throughout life. In the vast majority of cases, the infection is asymptomatic. H. pylori is the leading cause of peptic ulcer disease and gastric cancer, however, and these outcomes occur in 10-15% of those infected. Gastric adenocarcinoma is the third most common cause of cancer-associated death, and peptic ulcer disease is a significant cause of morbidity. Disease risk is related to the interplay of numerous bacterial host and environmental factors, many of which influence chronic inflammation and damage to the gastric mucosa. This chapter summarizes what is known about health and disease in H. pylori infection, and highlights the need for additional research in this area.
Collapse
|
14
|
Ng GZ, Ke BX, Laskowski A, Thorburn DR, Sutton P. No evidence of a role for mitochondrial complex I in Helicobacter pylori pathogenesis. Helicobacter 2017; 22. [PMID: 28181350 DOI: 10.1111/hel.12378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Complex I is the first enzyme complex in the mitochondrial respiratory chain, responsible for generating a large fraction of energy during oxidative phosphorylation. Recently, it has been identified that complex I deficiency can result in increased inflammation due to the generation of reactive oxygen species by innate immune cells. As a reduction in complex I activity has been demonstrated in human stomachs with atrophic gastritis, we investigated whether complex I deficiency could influence Helicobacter pylori pathogenesis. MATERIALS AND METHODS Ndufs6gt/gt mice have a partial complex I deficiency. Complex I activity was quantified in the stomachs and immune cells of Ndufs6gt/gt mice by spectrophotometric assays. Ndufs6gt/gt mice were infected with H. pylori and bacterial colonization assessed by colony-forming assay, gastritis assessed histologically, and H. pylori -specific humoral response quantified by ELISA. RESULTS The immune cells and stomachs of Ndufs6gt/gt mice were found to have significantly decreased complex I activity, validating the model for assessing the effects of complex I deficiency in H. pylori infection. However, there was no observable effect of complex I deficiency on either H. pylori colonization, the resulting gastritis, or the humoral response. CONCLUSIONS Although complex I activity is described to suppress innate immune responses and is decreased during atrophic gastritis in humans, our data suggest it does not affect H. pylori pathogenesis.
Collapse
Affiliation(s)
- Garrett Z Ng
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia.,Centre for Animal Biotechnology, School of Veterinary and Agricultural Science, University of Melbourne, Parkville, VIC 3010, Australia
| | - Bi-Xia Ke
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - Adrienne Laskowski
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - David R Thorburn
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia.,Victorian Clinical Genetics Services, The Royal Children's Hospital, Parkville, VIC 3052, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Philip Sutton
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC 3052, Australia.,Centre for Animal Biotechnology, School of Veterinary and Agricultural Science, University of Melbourne, Parkville, VIC 3010, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
15
|
Ng GZ, Menheniott TR, Every AL, Stent A, Judd LM, Chionh YT, Dhar P, Komen JC, Giraud AS, Wang TC, McGuckin MA, Sutton P. The MUC1 mucin protects against Helicobacter pylori pathogenesis in mice by regulation of the NLRP3 inflammasome. Gut 2016; 65:1087-99. [PMID: 26079943 DOI: 10.1136/gutjnl-2014-307175] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 03/11/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The mucin MUC1, best known for providing an epithelial barrier, is an important protective host factor in both humans and mice during Helicobacter pylori pathogenesis. This study aimed to identify the long-term consequences of MUC1 deficiency on H. pylori pathogenesis and the mechanism by which MUC1 protects against H. pylori gastritis. DESIGN Wildtype and Muc1(-/-) mice were infected for up to 9 months, and the gastric pathology, immunological response and epigenetic changes assessed. The effects of MUC1 on the inflammasome, a potent inflammatory pathway, were examined in macrophages and H. pylori-infected mice deficient in both MUC1 and inflammasome components. RESULTS Muc1(-/-) mice began to die 6 months after challenge, indicating Muc1 deficiency made H. pylori a lethal infection. Surprisingly, chimaeric mouse infections revealed MUC1 expression by haematopoietic-derived immune cells limits H. pylori-induced gastritis. Gastritis in infected Muc1(-/-) mice was associated with elevated interleukin (IL)-1β and epigenetic changes in their gastric mucosa similar to those in transgenic mice overexpressing gastric IL-1β, implicating MUC1 regulation of an inflammasome. In support of this, infected Muc1(-/-)Casp1(-/-) mice did not develop severe gastritis. Further, MUC1 regulated Nlrp3 expression via an nuclear factor (NF)-κB-dependent pathway and reduced NF-κB pathway activation via inhibition of IRAK4 phosphorylation. The importance of this regulation was proven using Muc1(-/-)Nlrp3(-/-) mice, which did not develop severe gastritis. CONCLUSIONS MUC1 is an important, previously unidentified negative regulator of the NLRP3 inflammasome. H. pylori activation of the NLRP3 inflammasome is normally tightly regulated by MUC1, and loss of this critical regulation results in the development of severe pathology.
Collapse
Affiliation(s)
- Garrett Z Ng
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Trevelyan R Menheniott
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Alison L Every
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew Stent
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Louise M Judd
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Yok Teng Chionh
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Poshmaal Dhar
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Jasper C Komen
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Andrew S Giraud
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, USA
| | - Michael A McGuckin
- Mucosal Diseases Program, Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Philip Sutton
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
16
|
Sjökvist Ottsjö L, Flach CF, Nilsson S, de Waal Malefyt R, Walduck AK, Raghavan S. Defining the Roles of IFN-γ and IL-17A in Inflammation and Protection against Helicobacter pylori Infection. PLoS One 2015; 10:e0131444. [PMID: 26168305 PMCID: PMC4500503 DOI: 10.1371/journal.pone.0131444] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 06/02/2015] [Indexed: 01/13/2023] Open
Abstract
CD4+ T cells have been shown to be essential for vaccine-induced protection against Helicobacter pylori infection. However, the effector mechanisms leading to reductions in the gastric bacterial loads of vaccinated mice remain unclear. We have investigated the function of IFN-γ and IL-17A for vaccine-induced protection and inflammation (gastritis) using IFN-γ-gene-knockout (IFN-γ-/-) mice, after sublingual or intragastric immunization with H. pylori lysate antigens and cholera toxin. Bacteria were enumerated in the stomachs of mice and related to the gastritis score and cellular immune responses. We report that sublingually and intragastrically immunized IFN-γ-/- mice had significantly reduced bacterial loads similar to immunized wild-type mice compared to respective unimmunized infection controls. The reduction in bacterial loads in sublingually and intragastrically immunized IFN-γ-/- mice was associated with significantly higher levels of IL-17A in stomach extracts and lower gastritis scores compared with immunized wild-type mice. To study the role of IL-17A for vaccine-induced protection in sublingually immunized IFN-γ-/- mice, IL-17A was neutralized in vivo at the time of infection. Remarkably, the neutralization of IL-17A in sublingually immunized IFN-γ-/- mice completely abolished protection against H. pylori infection and the mild gastritis. In summary, our results suggest that IFN-γ responses in the stomach of sublingually immunized mice promote vaccine-induced gastritis, after infection with H. pylori but that IL-17A primarily functions to reduce the bacterial load.
Collapse
Affiliation(s)
| | - Carl-Fredrik Flach
- Department of Microbiology & Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Rene de Waal Malefyt
- Department of Immunology, Merck Research Laboratories, Palo Alto, California, United States of America
| | - Anna K. Walduck
- School of Applied Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Sukanya Raghavan
- Department of Microbiology & Immunology, University of Gothenburg, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
17
|
Harbour SN, Mitchell HM, Sutton P. Host Nonresponsiveness Does not Interfere With Vaccine-Mediated Protection Against Gastric Helicobacter Infection. Helicobacter 2015; 20:217-22. [PMID: 25683672 DOI: 10.1111/hel.12197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Helicobacter pylori pathogenesis results from the inflammation induced by chronic infection. CBA mice are nonresponsive to gastric Helicobacter infection, providing a useful model for examining host regulation of Helicobacter-induced gastritis. We examined whether gastric Helicobacter nonresponsiveness impacts upon vaccine efficacy and whether immune-mediated protection could occur in the absence of inflammation. METHODS Mice were vaccinated prior to challenge with Helicobacter felis or H. pylori. Gastritis and H. felis colonization was evaluated histologically. H. pylori colonization was quantified by colony-forming assay. RESULTS Immunizations protected CBA mice against challenge with either H. felis or H. pylori. Protection against H. felis was marked by a loss of nonresponsiveness and development of an atrophic gastritis with mucus metaplasia. However, vaccine-induced protection against H. pylori was only associated with cell infiltration into the gastric mucosa. CONCLUSIONS Nonresponsiveness to gastric Helicobacter infection did not interfere with vaccination-induced protection. Vaccine-induced protective immunity against H. pylori was linked with the induction of cellular infiltration, but importantly not atrophic gastritis.
Collapse
Affiliation(s)
- Stacey N Harbour
- Centre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Parkville, Vic., 3010, Australia
| | - Hazel M Mitchell
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Philip Sutton
- Centre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Parkville, Vic., 3010, Australia.,Mucosal Immunology, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC., 3052, Australia
| |
Collapse
|
18
|
|
19
|
Protease-activated receptor 1 suppresses Helicobacter pylori gastritis via the inhibition of macrophage cytokine secretion and interferon regulatory factor 5. Mucosal Immunol 2015; 8:68-79. [PMID: 24866378 DOI: 10.1038/mi.2014.43] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 04/28/2014] [Indexed: 02/08/2023]
Abstract
Chronic gastritis from Helicobacter pylori infection is a major factor in the development of gastric adenocarcinoma. Factors that regulate gastritis severity are important in determining which individuals are susceptible to H. pylori-associated disease. Although protease-activated receptor 1 (PAR1) has been identified as one such host factor, its mechanism of action is unknown. Using chimeric mice, we demonstrated that PAR1-mediated protection against H. pylori gastritis requires bone marrow-derived cells. Analyses of the gastric mucosa revealed that PAR1 suppresses cellular infiltration and both T helper type 1 (Th1) and T helper type 17 (Th17) responses to infection. Moreover, PAR1 expression was associated with reduced vaccine-mediated protection against H. pylori. Analyses of H. pylori-stimulated macrophages revealed that PAR1 activation suppressed secretion of interleukin (IL)-12 and IL-23, key drivers of Th1 and Th17 immunity, respectively. Furthermore, PAR1 suppressed interferon regulatory factor 5 (IRF5), an important transcription factor for IL-12 and IL-23, both in the infected mucosa and following bacterial stimulation. PAR1 suppression of IRF5 and IL-12/23 secretion by macrophages provides a novel mechanism by which the host suppresses the mucosal Th1 and Th17 response to H. pylori infection. Dysregulation of this process is likely an important factor in the susceptibility of some individuals to H. pylori-associated disease.
Collapse
|
20
|
Baker L, Chitas AML, Hartley CA, Coppo MJC, Vaz PK, Stent A, Gilkerson JR, Devlin JM, Every AL. Recombinant herpesvirus glycoprotein G improves the protective immune response to Helicobacter pylori vaccination in a mouse model of disease. PLoS One 2014; 9:e96563. [PMID: 24794215 PMCID: PMC4008605 DOI: 10.1371/journal.pone.0096563] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/08/2014] [Indexed: 12/12/2022] Open
Abstract
Alphaherpesviruses, which have co-evolved with their hosts for more than 200 million years, evade and subvert host immune responses, in part, by expression of immuno-modulatory molecules. Alphaherpesviruses express a single, broadly conserved chemokine decoy receptor, glycoprotein G (gG), which can bind multiple chemokine classes from multiple species, including human and mouse. Previously, we demonstrated that infection of chickens with an infectious laryngotracheitis virus (ILTV) mutant deficient in gG resulted in altered host immune responses compared to infection with wild-type virus. The ability of gG to disrupt the chemokine network has the potential to be used therapeutically. Here we investigated whether gG from ILTV or equine herpesvirus 1 (EHV-1) could modulate the protective immune response induced by the Helicobacter pylori vaccine antigen, catalase (KatA). Subcutaneous immunisation of mice with KatA together with EHV-1 gG, but not ILTV gG, induced significantly higher anti-KatA IgG than KatA alone. Importantly, subcutaneous or intranasal immunisation with KatA and EHV-1 gG both resulted in significantly lower colonization levels of H. pylori colonization following challenge, compared to mice vaccinated with KatA alone. Indeed, the lowest colonization levels were observed in mice vaccinated with KatA and EHV-1 gG, subcutaneously. In contrast, formulations containing ILTV gG did not affect H. pylori colonisation levels. The difference in efficacy between EHV-1 gG and ILTV gG may reflect the different spectrum of chemokines bound by the two proteins. Together, these data indicate that the immuno-modulatory properties of viral gGs could be harnessed for improving immune responses to vaccine antigens. Future studies should focus on the mechanism of action and whether gG may have other therapeutic applications.
Collapse
Affiliation(s)
- Louise Baker
- Centre for Animal Biotechnology, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Andre M. L. Chitas
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Carol A. Hartley
- Centre for Equine Infectious Disease, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Mauricio J. C. Coppo
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Paola K. Vaz
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew Stent
- Centre for Animal Biotechnology, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - James R. Gilkerson
- Centre for Equine Infectious Disease, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Joanne M. Devlin
- Asia-Pacific Centre for Animal Health, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
| | - Alison L. Every
- Centre for Animal Biotechnology, Faculty of Veterinary Science, The University of Melbourne, Parkville, VIC, Australia
- * E-mail:
| |
Collapse
|
21
|
Chionh YT, Arulmuruganar A, Venditti E, Ng GZ, Han JX, Entwisle C, Ang CS, Colaco CA, McNulty S, Sutton P. Heat shock protein complex vaccination induces protection against Helicobacter pylori without exogenous adjuvant. Vaccine 2014; 32:2350-8. [PMID: 24625340 DOI: 10.1016/j.vaccine.2014.02.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/24/2014] [Accepted: 02/12/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND The development of a vaccine against the human gastric pathogen Helicobacter pylori, the main causative agent of gastric adenocarcinoma, has been hampered by a number of issues, including the lack of a mucosal adjuvant for use in humans. Heat shock proteins (Hsp), highly conserved molecules expressed by both bacteria and mammalian species, possess a range of functions, including acting as chaperones for cellular proteins and the ability to activate innate immune receptors. Hsp complex (HspC) vaccines, containing Hsp derived from pathogenic bacteria, are immunostimulatory without addition of an exogenous adjuvant and can induce immunity against their chaperoned proteins. In this study we explored in mice the potential utility of a H. pylori HspC vaccine. RESULTS Vaccination with H. pylori HspC, by either the subcutaneous or respiratory mucosal route, induced a strong antibody response, elevated gastric cytokine levels and significant protection against subsequent live challenge with this pathogen. The level of protection induced by non-adjuvanted HspC vaccine was equivalent to that which resulted from vaccination with adjuvanted vaccines. While protection induced by immunisation with adjuvanted vaccines was associated with the development of a moderate to severe atrophic gastritis, that induced by H. pylori HspC only resulted in a mild inflammatory response, despite an increase in pro-inflammatory gastric cytokines. This reduced gastritis correlated with an increase in IL-10 and IL-13 levels in the gastric tissues of HspC vaccinated, H. pylori challenged mice. CONCLUSIONS H. pylori HspC vaccines have the potential to overcome some of the issues preventing the development of a human vaccine against this pathogen: HspC induced protective immunity against H. pylori without addition of an adjuvant and without the induction of a severe inflammatory response. However, complete protection was not obtained so further optimisation of this technology is needed if a human vaccine is to become a reality.
Collapse
Affiliation(s)
- Yok Teng Chionh
- Mucosal Immunology, Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Parkville, VIC 3052, Australia; Centre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Parkville, VIC 3010, Australia
| | - Arthi Arulmuruganar
- Mucosal Immunology, Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Parkville, VIC 3052, Australia
| | - Elena Venditti
- ImmunoBiology Ltd., Babraham Research Campus, Babraham, Cambridge, UK
| | - Garrett Z Ng
- Mucosal Immunology, Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Parkville, VIC 3052, Australia
| | - Jia-Xi Han
- Mucosal Immunology, Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Parkville, VIC 3052, Australia
| | - Claire Entwisle
- ImmunoBiology Ltd., Babraham Research Campus, Babraham, Cambridge, UK
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | - Camilo A Colaco
- ImmunoBiology Ltd., Babraham Research Campus, Babraham, Cambridge, UK
| | - Shaun McNulty
- ImmunoBiology Ltd., Babraham Research Campus, Babraham, Cambridge, UK
| | - Philip Sutton
- Mucosal Immunology, Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Parkville, VIC 3052, Australia; Centre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
22
|
Sutton P, Chionh YT. Why can't we make an effective vaccine against Helicobacter pylori? Expert Rev Vaccines 2013; 12:433-41. [PMID: 23560923 DOI: 10.1586/erv.13.20] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori is a major human pathogen that colonizes the stomach and is the lead etiological agent for several pathologies. An effective vaccine against these bacteria would be invaluable for protecting against gastric adenocarcinoma. However, the development of such a vaccine has stalled and the field has progressed little in the last decade. In this review, the authors provide an opinion on key problems that are preventing the development of a H. pylori vaccine. Primarily, this involves the inability to produce a completely protective immune response. The knock-on effects of this include a loss of industry investment. Overcoming these problems will likely involve defeating the immune-evasion defenses of H. pylori, in particular the mechanism(s) by which it evades antibody-mediated attack.
Collapse
Affiliation(s)
- Philip Sutton
- Centre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Parkville, VIC 3010, Australia.
| | | |
Collapse
|
23
|
Vermoote M, Flahou B, Pasmans F, Ducatelle R, Haesebrouck F. Protective efficacy of vaccines based on the Helicobacter suis urease subunit B and γ-glutamyl transpeptidase. Vaccine 2013; 31:3250-6. [PMID: 23707444 DOI: 10.1016/j.vaccine.2013.05.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/18/2013] [Accepted: 05/10/2013] [Indexed: 01/01/2023]
Abstract
Helicobacter suis causes gastric lesions in pigs and humans. This study aimed to evaluate the protective efficacy of immunization with combinations of the H. suis urease subunit B (UreB) and γ-glutamyl transpeptidase (GGT), both recombinantly expressed in Escherichia coli (rUreB and rGGT, respectively). Mice were intranasally immunized with rUreB, rGGT or a combination of both proteins, administered simultaneously or sequentially. Control groups consisted of non-immunized and non-challenged mice (negative controls), sham-immunized and H. suis-challenged mice (sham-immunized controls), and finally, H. suis whole-cell lysate-immunized and H. suis challenged mice. Cholera toxin was used as mucosal adjuvant. All immunizations induced a significant reduction of gastric H. suis colonization, which was least pronounced in the groups immunized with rGGT and rUreB only. Consecutive immunization with rGGT followed by rUreB and immunization with the bivalent vaccine improved the protective efficacy compared to immunization with single proteins, with a complete clearance of infection observed in 50% of the animals. Immunization with whole-cell lysate induced a similar reduction of gastric bacterial colonization compared to rGGT and rUreB in combinations. Gastric lesions, however, were less pronounced in mice immunized with combinations of rUreB and rGGT compared to mice immunized with whole-cell lysate. In conclusion, vaccination with a combination of rGGT and rUreB protected mice against a subsequent H. suis infection and was not associated with severe post-vaccination gastric inflammation, indicating that it may be a promising method for control of H. suis infections.
Collapse
Affiliation(s)
- Miet Vermoote
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | | | | | | | | |
Collapse
|
24
|
Walduck AK, Becher D. Leptin, CD4(+) T(reg) and the prospects for vaccination against H. pylori infection. Front Immunol 2012; 3:316. [PMID: 23087691 PMCID: PMC3470998 DOI: 10.3389/fimmu.2012.00316] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/24/2012] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori infection induces chronic inflammation which is characterized not only by infiltrations of inflammatory cells such as neutrophils and CD4+ T cells, but also significant populations of regulatory T cells (Treg). These cells are important for disease pathogenesis because they are believed to contribute to the persistence of the infection. Despite encouraging results in animal models, the prospects for an effective H. pylori vaccine are currently poor because of generally disappointing results in preclinical and phase 1 trials. As a result, a current major focus of basic research on vaccination is to better understand the mechanisms regulating the inflammatory response with the view it can inform future vaccine design. Our studies in this area have focused on gastric CD4+ Treg in vaccinated mice, and raised the hypothesis that adipokines in particular leptin are involved the establishment of a protective gastric immune response. Here we discuss the hypothesis that vaccination deregulates Treg responses in the gastric mucosa, and that this process is mediated by leptin. We propose that reduced suppression permits a protective sub population of H. pylori-specific CD4+ T cells to exert protective effects, presumably via the gastric epithelium. Evidence from the literature and experimental approaches will be discussed.
Collapse
Affiliation(s)
- Anna K Walduck
- Department of Microbiology and Immunology, University of Melbourne Parkville, VIC, Australia
| | | |
Collapse
|
25
|
Ameri Shah Reza M, Mousavi Gargari SL, Rasooli I, Jalali Nadoushan M, Ebrahimizadeh W. Inhibition of H. pylori colonization and prevention of gastritis in murine model. World J Microbiol Biotechnol 2012; 28:2513-9. [PMID: 22806157 DOI: 10.1007/s11274-012-1059-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/14/2012] [Indexed: 01/15/2023]
Abstract
Helicobacter pylori is a Gram-negative spiral bacterium that colonizes human gastric mucosa causing infection. In this study aiming at inhibition of H. pylori infection we made an attempt to evaluate immunogenicity of the total (UreC) and C-terminal (UreCc) fragments of H. pylori urease. Total UreC and its C-terminal fragment were expressed in E. coli. Recombinant proteins were analyzed by SDS-PAGE and western blot and then purified by Ni-NTA affinity chromatography. Female C57BL6/j mice were immunized with the purified proteins (UreC and UreCc). Antibody titers from isolated sera were measured by ELISA. Immunized mice were then challenged by oral gavage with live H. pylori Sydney strain SS1. Total of 109 CFU were inoculated into stomach of immunized and unimmunized healthy mice three times each at one day interval. Eight weeks after the last inoculation, the blood sample was collected and the serum antibody titer was estimated by ELISA. Stomach tissues from control and experimental animal groups were studied histopathologically. UreC and UreCc yielded recombinant proteins of 61 and 31 kDa respectively. ELIZA confirmed establishment of immunity and the antibodies produced thereby efficiently recognized H. pylori and inhibited its colonization in vivo. Pathological analysis did not reveal established infection in immunized mice challenged with H. pylori. The results support the idea that UreC and UreCc specific antibodies contribute to protection against H. pylori infections.
Collapse
|
26
|
Alkyl hydroperoxide reductase: a candidate Helicobacter pylori vaccine. Vaccine 2012; 30:3876-84. [PMID: 22512976 DOI: 10.1016/j.vaccine.2012.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Revised: 03/20/2012] [Accepted: 04/01/2012] [Indexed: 02/06/2023]
Abstract
Helicobacter pylori (H. pylori) is the most important etiological agent of chronic active gastritis, peptic ulcer disease and gastric cancer. The aim of this study was to evaluate the efficacy of alkyl hydroperoxide reductase (AhpC) and mannosylated AhpC (mAhpC) as candidate vaccines in the C57BL/6J mouse model of H. pylori infection. Recombinant AhpC was cloned, over-expressed and purified in an unmodified form and was also engineered to incorporate N and C-terminal mannose residues when expressed in the yeast Pichia pastoris. Mice were immunized systemically and mucosally with AhpC and systemically with mAhpC prior to challenge with H. pylori. Serum IgG responses to AhpC were determined and quantitative culture was used to determine the efficacy of vaccination strategies. Systemic prophylactic immunization with AhpC/alum and mAhpC/alum conferred protection against infection in 55% and 77.3% of mice, respectively. Mucosal immunization with AhpC/cholera toxin did not protect against infection and elicited low levels of serum IgG in comparison with systemic immunization. These data support the use of AhpC as a potential vaccine candidate against H. pylori infection.
Collapse
|
27
|
Arnold IC, Hitzler I, Engler D, Oertli M, Agger EM, Müller A. The C-terminally encoded, MHC class II-restricted T cell antigenicity of the Helicobacter pylori virulence factor CagA promotes gastric preneoplasia. THE JOURNAL OF IMMUNOLOGY 2011; 186:6165-72. [PMID: 21518972 DOI: 10.4049/jimmunol.1003472] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Chronic infection with the human bacterial pathogen Helicobacter pylori causes gastritis and predisposes carriers to an increased gastric cancer risk. Consequently, H. pylori-specific vaccination is widely viewed as a promising strategy of gastric cancer prevention. H. pylori strains harboring the Cag pathogenicity island (PAI) are associated with particularly unfavorable disease outcomes in humans and experimental rodent models. We show in this study using a C57BL/6 mouse model of Cag-PAI(+) H. pylori infection that the only known protein substrate of the Cag-PAI-encoded type IV secretion system, the cytotoxin-associated gene A (CagA) protein, harbors MHC class II-restricted T cell epitopes. Several distinct nonoverlapping epitopes in CagA's central and C-terminal regions were predicted in silico and could be confirmed experimentally. CagA(+) infection elicits CD4(+) T cell responses in mice, which are strongly enhanced by prior mucosal or parenteral vaccination with recombinant CagA. The adoptive transfer of CagA-specific T cells to T cell-deficient, H. pylori-infected recipients is sufficient to induce the full range of preneoplastic immunopathology. Similarly, immunization with a cholera toxin-adjuvanted, CagA(+) whole-cell sonicate vaccine sensitizes mice to, rather than protects them from, H. pylori-associated gastric cancer precursor lesions. In contrast, H. pylori-specific tolerization by neonatal administration of H. pylori sonicate in conjunction with a CD40L-neutralizing Ab prevents H. pylori-specific, pathogenic T cell responses and gastric immunopathology. We conclude that active tolerization may be superior to vaccination strategies in gastric cancer prevention.
Collapse
Affiliation(s)
- Isabelle C Arnold
- Institute of Molecular Cancer Research, University of Zurich, 8057 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
28
|
Becher D, Deutscher ME, Simpfendorfer KR, Wijburg OL, Pederson JS, Lew AM, Strugnell RA, Walduck AK. Local recall responses in the stomach involving reduced regulation and expanded help mediate vaccine-induced protection against Helicobacter pylori in mice. Eur J Immunol 2010; 40:2778-90. [PMID: 21038469 DOI: 10.1002/eji.200940219] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori is recognised as the chief cause of chronic gastritis, ulcers and gastric cancer in humans. With increased incidence of treatment failure and antibiotic resistance, development of prophylactic or therapeutic vaccination is a desirable alternative. Although the results of vaccination studies in animal models have been promising, studies in human volunteers have revealed problems such as 'post-immunisation gastritis' and comparatively poor responses to vaccine antigens. The focus of this study was to compare the gastric and systemic cellular immune responses induced by recombinant attenuated Salmonella Typhimurium-based vaccination in the C57BL/6 model of H. pylori infection. Analysis of lymphocyte populations in the gastric mucosa, blood, spleen, paragastric LN and MLN revealed that the effects of vaccination were largely confined to the parenchymal stomach rather than lymphoid organs. Vaccine-induced protection was correlated with an augmented local recall response in the gastric mucosa, with increased proportions of CD4(+) T cells, neutrophils and reduced proportions of CD4(+) Treg. CD4(+) T cells isolated from the stomachs of vaccinated mice proliferated ex vivo in response to H. pylori antigen, and secreted Th1 cytokines, particularly IFN-γ. This detailed analysis of local gastric immune responses provides insight into the mechanism of vaccine-induced protection.
Collapse
Affiliation(s)
- Dorit Becher
- Department of Microbiology and Immunology, University of Melbourne, VIC, Australia
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Sublingual immunization protects against Helicobacter pylori infection and induces T and B cell responses in the stomach. Infect Immun 2010; 78:4251-60. [PMID: 20696831 DOI: 10.1128/iai.00536-10] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sublingual (SL) immunization has been described as an effective novel way to induce mucosal immune responses in the respiratory and genital tracts. We examined the potential of SL immunization against Helicobacter pylori to stimulate immune responses in the gastrointestinal mucosa and protect against H. pylori infection. Mice received two SL immunizations with H. pylori lysate antigens and cholera toxin as an adjuvant, and after challenge with live H. pylori bacteria, their immune responses and protection were evaluated, as were immune responses prior to challenge. SL immunization induced enhanced proliferative responses to H. pylori antigens in cervicomandibular lymph nodes and provided at least the same level of immune responses and protection as corresponding intragastric immunization. Protection in SL-immunized mice was associated with strong H. pylori-specific serum IgG and IgA antibody responses in the stomach and intestine, with strong proliferation and gamma interferon (IFN-γ) and interleukin-17 (IL-17) production by spleen and mesenteric lymph node T cells stimulated with H. pylori antigens in vitro, and with increased IFN-γ and IL-17 gene expression in the stomach compared to levels in infected unimmunized mice. Immunohistochemical studies showed enhanced infiltration of CD4(+) T cells and CD19(+) B cells into the H. pylori-infected stomach mucosa of SL-immunized but not unimmunized H. pylori-infected mice, which coincided with increased expression of the mucosal addressin cell adhesion molecule (MAdCAM-1) and T and B cell-attracting chemokines CXCL10 and CCL28. We conclude that, in mice, SL immunization can effectively induce protection against H. pylori infection in association with strong T and B cell infiltration into the stomach.
Collapse
|
30
|
Chambers MA. Transcutaneous immunization with lipid offers a new route of vaccination against Helicobacter pylori and a new candidate delivery vehicle. Expert Rev Vaccines 2010; 9:249-53. [PMID: 20218852 DOI: 10.1586/erv.10.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Needle-free methods of vaccination may allow rapid, simple and safe vaccination of large populations. Oral vaccination is the best established method but faces the hurdle of oral tolerance to the vaccine antigen. Skin-based transcutaneous immunization (TCI) offers an alternative needle-free route of vaccination that is able to induce protective immunity without the problem of oral tolerance. Helicobacter pylori is an important human pathogen associated with a number of gastrointestinal disorders, including gastritis, peptic ulcers and gastric tumors. Conventional treatments involving the use of antibiotics have a number of limitations and the development of an effective vaccine is the best long-term treatment option. A variety of experimental vaccines to Helicobacter have been reported. The paper reviewed here combines the approach of TCI with the use of a novel lipid antigen delivery system, hitherto only used for oral vaccination, to evaluate the potential for TCI for a simple vaccination strategy against Helicobacter and potentially other disease-causing organisms.
Collapse
Affiliation(s)
- Mark Andrew Chambers
- TB Research Group, Veterinary Laboratories Agency Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK.
| |
Collapse
|
31
|
Cui Y, Wang CL, Liu XW, Wang XH, Chen LL, Zhao X, Fu N, Lu FG. Two stomach-originated lactobacillus strains improve Helicobacter pylori infected murine gastritis. World J Gastroenterol 2010; 16:445-52. [PMID: 20101769 PMCID: PMC2811796 DOI: 10.3748/wjg.v16.i4.445] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the potential anti-Helicobacter pylori (H. pylori) and anti-inflammation in vivo effects of two lactobacillus strains from human stomach.
METHODS: Forty H. pylori infected Balb/c mice were randomly divided into 4 groups: proton pump inhibitor and antibiotics triple treated group, Lactobacillus fermenti (L. fermenti) treated group, Lactobacillus acidophilus treated group and normal saline control group. Ten uninfected mice were also included as blank control group. The infection of H. pylori was detected by rapid urease tests, Giemsa staining and bacterial culture. The colonization of H. pylori was assessed in bacterial density score and gastric inflammation was assessed in histological score. The colonization of L. fermenti was performed by fluorescent probe.
RESULTS: Histopathologic evaluation showed significant release of mucosal inflammation in gastric antrum and gastric body in lactobacillus treated groups and triple treated group. H. pylori eradication rate in both lactobacillus treated groups and triple treated group were higher than normal saline control group. Lactobacillus treated groups and triple treated group showed significant decrease of H. pylori bacterial density.
CONCLUSION: Both lactobacillus strains have a significant anti-H. pylori activity; L. fermenti displays more efficient antagonistic activity in vivo against H. pylori infection.
Collapse
|
32
|
Transcutaneous immunization with novel lipid-based adjuvants induces protection against gastric Helicobacter pylori infection. Vaccine 2009; 27:6983-90. [DOI: 10.1016/j.vaccine.2009.09.078] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 09/07/2009] [Accepted: 09/20/2009] [Indexed: 01/08/2023]
|
33
|
Inoue K, Shiota S, Yamada K, Gotoh K, Suganuma M, Fujioka T, Ahmed K, Iha H, Nishizono A. Evaluation of a new tumor necrosis factor-alpha-inducing membrane protein of Helicobacter pylori as a prophylactic vaccine antigen. Helicobacter 2009; 14:135-43. [PMID: 19751439 DOI: 10.1111/j.1523-5378.2009.00713.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Tumor necrosis factor (TNF)-alpha-inducing protein (Tip alpha) is a newly identified carcinogenic factor present in Helicobacter pylori. Tip alpha has the unique function of inducing TNF-alpha production by gastric cells in vitro and is assumed to be related with the development of gastritis and gastric cancer. We investigated the effects of vaccination with Tip alpha against H. pylori infection and analyzed the immune responses. METHODS C57BL/6 mice were immunized via the intranasal route with CpG, recombinant Tip alpha + CpG, and recombinant del-Tip alpha (a mutant of Tip alpha) + CpG. Eight weeks after the mice were infected with H. pylori (5 x 10(7) CFU), the number of colonizing bacteria in the stomach was calculated, and the histological severity of gastritis was evaluated. Levels of Tip alpha-specific IgG and IgA antibodies in mouse serum were measured by an enzyme-linked immunosorbent assay (ELISA). Local production of cytokines including Interleukin (IL)-10, TNF-alpha and Interferon (IFN)-gamma in gastric mucosa was also measured by real time-PCR. RESULTS Levels of Tip alpha-specific antibodies were significantly higher in Tip alpha-immunized and del-Tip alpha-immunized mice than in the infection control group. The numbers of colonizing bacteria were significantly reduced in Tip alpha-immunized mice (4.29 x 10(5) CFU/g) and del-Tip alpha immunized mice (2.5 x 10(5 )CFU/g) compared with infection control mice (5.7 x 10(6) CFU/g). The levels of IFN-gamma and IL-10 were significantly higher in del-Tip alpha-immunized mice than the infection control group. CONCLUSION Vaccinations with Tip alpha and del-Tip alpha were effective against H. pylori infection. The inhibition of H. pylori colonization is associated mainly with Th1 cell-mediated immunity.
Collapse
Affiliation(s)
- Kunimitsu Inoue
- Department of Microbiology, Faculty of Medicine, Oita University, Oita, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sayi A, Kohler E, Hitzler I, Arnold I, Schwendener R, Rehrauer H, Müller A. The CD4+ T cell-mediated IFN-gamma response to Helicobacter infection is essential for clearance and determines gastric cancer risk. THE JOURNAL OF IMMUNOLOGY 2009; 182:7085-101. [PMID: 19454706 DOI: 10.4049/jimmunol.0803293] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Chronic infection with the bacterial pathogen Helicobacter pylori is a risk factor for the development of gastric cancer, yet remains asymptomatic in the majority of individuals. We report here that the C57BL/6 mouse model of experimental infection with the closely related Helicobacter felis recapitulates this wide range in host susceptibility. Although the majority of infected animals develop premalignant lesions such as gastric atrophy, compensatory epithelial hyperplasia, and intestinal metaplasia, a subset of mice is completely protected from preneoplasia. Protection is associated with a failure to mount an IFN-gamma response to the infection and with a concomitant high Helicobacter burden. Using a vaccine model as well as primary infection and adoptive transfer models, we demonstrate that IFN-gamma, secreted predominantly by CD4(+)CD25(-) effector T(H) cells, is essential for Helicobacter clearance, but at the same time mediates the formation of preneoplastic lesions. We further provide evidence that IFN-gamma triggers a common transcriptional program in murine gastric epithelial cells in vitro and in vivo and induces their preferential transformation to the hyperplastic phenotype. In summary, our data suggest a dual role for IFN-gamma in Helicobacter pathogenesis that could be the basis for the differential susceptibility to H. pylori-induced gastric pathology in the human population.
Collapse
Affiliation(s)
- Ayca Sayi
- Institute of Molecular Cancer Research and
| | | | | | | | | | | | | |
Collapse
|
35
|
M-cell targeting of whole killed bacteria induces protective immunity against gastrointestinal pathogens. Infect Immun 2009; 77:2962-70. [PMID: 19380476 DOI: 10.1128/iai.01522-08] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
As the majority of human pathogens infect via a mucosal surface, delivery of killed vaccines by mucosal routes could potentially improve protection against many such organisms. Our ability to develop effective killed mucosal vaccines is inhibited by a lack of adjuvants that are safe and effective in humans. The Ulex europaeus agglutinin I (UEA-I) lectin specifically binds M cells lining the murine gastrointestinal tract. We explored the potential for M-cell-targeted vaccination of whole, killed Helicobacter pylori, the main causative agent of peptic ulcer disease and gastric cancer, and Campylobacter jejuni, the most common cause of diarrhea. Oral delivery of UEA-I-agglutinated H. pylori or C. jejuni induced a significant increase in both serum and intestinal antibody levels. This elevated response (i) required the use of whole bacteria, as it did not occur with lysate; (ii) was not mediated by formation of particulate clumps, as agglutination with a lectin with a different glycan specificity had no effect; and (iii) was not due to lectin-mediated, nonspecific immunostimulatory activity, as UEA-I codelivery with nonagglutinated bacteria did not enhance the response. Vaccination with UEA-I-agglutinated, killed whole H. pylori induced a protective response against subsequent live challenge that was as effective as that induced by cholera toxin adjuvant. Moreover, vaccination against C. jejuni by this approach resulted in complete protection against challenge in almost all animals. We believe that this is the first demonstration that targeting of whole killed bacteria to mucosal M cells can induce protective immunity without the addition of an immunostimulatory adjuvant.
Collapse
|
36
|
Flahou B, Hellemans A, Meyns T, Duchateau L, Chiers K, Baele M, Pasmans F, Haesebrouck F, Ducatelle R. Protective immunization with homologous and heterologous antigens against Helicobacter suis challenge in a mouse model. Vaccine 2009; 27:1416-21. [PMID: 19136039 DOI: 10.1016/j.vaccine.2008.12.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 12/10/2008] [Accepted: 12/20/2008] [Indexed: 12/31/2022]
Abstract
Helicobacter (H.) suis colonizes the stomach of more than 60% of slaughter pigs and is also of zoonotic importance. Recently, this bacterium was isolated in vitro, enabling the use of pure cultures for research purposes. In this study, mice were immunized intranasally or subcutaneously with whole bacterial cell lysate of H. suis or the closely related species H. bizzozeronii and H. cynogastricus, and subsequently challenged with H. suis. Control groups consisted of non-immunized and non-challenged mice (negative control group), as well as of sham-immunized mice that were inoculated with H. suis (positive control group). Urease tests on stomach tissue samples at 7 weeks after challenge infection were negative in all negative control mice, all intranasally immunized mice except one, and in all and 3 out of 5 animals of the H. cynogastricus and H. suis subcutaneously immunized groups, respectively. H. suis DNA was detected by PCR in the stomach of all positive control animals and all subcutaneously immunized/challenged animals. All negative control animals and some intranasally immunized/challenged mice were PCR-negative. In conclusion, immunization using antigens derived from the same or closely related bacterial species suppressed gastric colonization with H. suis, but complete protection was only achieved in a minority of animals following intranasal immunization.
Collapse
Affiliation(s)
- Bram Flahou
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
O'Rourke JL. Gene expression profiling in Helicobacter-induced MALT lymphoma with reference to antigen drive and protective immunization. J Gastroenterol Hepatol 2008; 23 Suppl 2:S151-6. [PMID: 19120889 DOI: 10.1111/j.1440-1746.2008.05553.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We have previously shown that long-term infection of BALB/c mice with gastric Helicobacter species results in the development of histopathological lesions that resemble those seen in patients diagnosed with gastric mucosa associated lymphoid tissue (MALT) lymphoma. This paper describes analysis of this disease at the molecular level through the use of microarray technology and immunohistochemical staining. We were able to monitor the genetic changes in the gastric mucosa characterized by distinct transcriptional signatures and correlate these with histological changes as the infection progressed from a chronic inflammatory infiltrate through to MALT lymphoma. This model system also enabled us to further dissect the role of antigen presentation and prophylactic immunization in the disease process. Antimicrobial therapy to eradicate the antigen correlated with significant reduction in pathology and major changes in the gene expression profile. Subsequent reintroduction of the antigen resulted in rapid tumor development which correlated with an increase in aggressively proliferating cells and changes in the cellular composition of the tumor. The response in vaccinated animals showed that the protected animals exhibited a strikingly different transcriptional profile compared to those of non-protected or control mice, indicating that the vaccination targeted the appropriate site leaving a long-lasting signature. The genes which were most significantly up-regulated included a number of adipocyte-specific factors, such as fat-cell specific cytokines and adipocyte surface markers. This study allowed for us to highlight the significance of antigen presentation in this disease and to hypothesis mechanisms associated with protective immunity.
Collapse
Affiliation(s)
- Jani L O'Rourke
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
38
|
Harbour SN, Every AL, Edwards S, Sutton P. Systemic immunization with unadjuvanted whole Helicobacter pylori protects mice against heterologous challenge. Helicobacter 2008; 13:494-9. [PMID: 19166414 DOI: 10.1111/j.1523-5378.2008.00640.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Adjuvant-free vaccines have many benefits, including decreased cost and toxicity. We examined the protective effect of systemic vaccination with adjuvant-free formalin-fixed Helicobacter pylori or bacterial lysate and the ability of this vaccine to induce protection against heterologous challenge. MATERIALS AND METHODS Mice were vaccinated subcutaneously with H. pylori 11637 lysate or formalin-fixed bacteria, with or without ISCOMATRIX adjuvant, then orally challenged with H. pylori SS1. Serum was taken prior to challenge to examine specific antibody levels induced by the vaccinations, and protection was assessed by colony-forming assay. RESULTS Vaccination with H. pylori 11637 lysate or formalin-fixed bacteria delivered systemically induced significantly higher levels of Helicobacter-specific serum IgG than the control, unvaccinated group and orally vaccinated group. After heterologous challenge with H. pylori SS1, all vaccinated groups had significantly lower levels of colonization compared with unvaccinated, control mice, regardless of the addition of adjuvant or route of delivery. Protection induced by systemic vaccination with whole bacterial preparations, without the addition of adjuvants, was only associated with a mild cellular infiltration into the gastric mucosa, with no evidence of atrophy. CONCLUSIONS Subcutaneous vaccination using unadjuvanted formalin-fixed H. pylori has the potential to be a simple, cost-effective approach to the development of a Helicobacter vaccine. Importantly, this vaccine was able to induce protection against heterologous challenge, a factor that would be crucial in any human Helicobacter vaccine. Further studies are required to determine mechanisms of protection and to improve protective ability.
Collapse
Affiliation(s)
- Stacey N Harbour
- Centre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
39
|
Skene CD, Doidge C, Sutton P. Evaluation of ISCOMATRIX™ and ISCOM™ vaccines for immunisation against Helicobacter pylori. Vaccine 2008; 26:3880-4. [DOI: 10.1016/j.vaccine.2008.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 04/18/2008] [Accepted: 05/07/2008] [Indexed: 12/23/2022]
|
40
|
Nyström-Asklin J, Adamsson J, Harandi AM. The adjuvant effect of CpG oligodeoxynucleotide linked to the non-toxic B subunit of cholera toxin for induction of immunity against H. pylori in mice. Scand J Immunol 2008; 67:431-40. [PMID: 18298617 DOI: 10.1111/j.1365-3083.2008.02085.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The present study was carried out to test the immunostimulatory and adjuvant effects of the non-toxic B subunit of cholera toxin (CTB), CpG oligodeoxynucleotide (ODN) and CpG ODN linked to CTB (CTB-CpG) for generation of immunity against H. pylori in mice. Herein, we showed that CTB-CpG induces more potent proinflammatory cytokine and chemokine responses in the cervical and the mesenteric lymph nodes (CLN and MLN, respectively) cells in vitro compared with those of CTB and CpG ODN. The adjuvant effects of these agents were examined following intranasal immunization of C57Bl/6 mice with H. pylori lysate in combination with CpG ODN, CTB or CTB-CpG. All three immunization regimes resulted in high H. pylori-specific IgG antibody responses; however, only the CTB-CpG and, to some extent, the CpG ODN immunized mice mounted a sustainable IgG2c antibody response. Importantly, mice immunized with H. pylori antigen and CTB-CpG or CpG ODN, but not CTB, developed strong H. pylori-specific proliferative and IFN-gamma responses in their MLN CD4+ T cells upon recall antigen stimulation in vitro. These mice also had significantly lower bacterial load compared with the control-infected mice. Furthermore, the CTB-CpG and the CpG ODN immunized mice developed increased specific IgA antibody responses in their gastrointestinal tracts following H. pylori challenge. These results imply that CTB-CpG and CpG ODN, but not CTB, could serve as nasal adjuvants for induction of a H. pylori-specific Th1 type immunity in MLN and also a specific mucosal IgA antibody response in the gastrointestinal tract upon H. pylori challenge.
Collapse
Affiliation(s)
- J Nyström-Asklin
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | | | | |
Collapse
|
41
|
CD8+ T cells are associated with severe gastritis in Helicobacter pylori-infected mice in the absence of CD4+ T cells. Infect Immun 2007; 76:1289-97. [PMID: 18025102 DOI: 10.1128/iai.00779-07] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Helicobacter pylori infection results in the development of chronic gastritis, and CD4+ T cells are a major component of the gastric cellular infiltrate. To examine whether CD4+ T cells are important in initiating and maintaining H. pylori-induced gastritis, mice deficient in CD4+ T cells (B6.BM1.GK 1.5 mice [GK 1.5 mice]) were infected with H. pylori. We found that as in normal mice, H. pylori-specific antibodies, mostly of the immunoglobulin M isotype, developed in GK 1.5 mice but were unable to cure H. pylori infection. Further, while the stomachs of H. pylori-infected GK 1.5 mice were more heavily infiltrated with CD8+ T cells and B cells, mice deficient in both CD4+ and CD8+ T cells developed mild inflammation comparable to the level observed for C57BL/6 mice. These observations suggest that CD4+ T cells may play an important role in regulating or suppressing gastric CD8+ T cells which, in the absence of CD4+ T cells, may mediate more-severe disease. These studies have revealed a potentially important role for CD8+ T cells in the gastric disease resulting from H. pylori infection.
Collapse
|
42
|
McGuckin MA, Every AL, Skene CD, Linden SK, Chionh YT, Swierczak A, McAuley J, Harbour S, Kaparakis M, Ferrero R, Sutton P. Muc1 mucin limits both Helicobacter pylori colonization of the murine gastric mucosa and associated gastritis. Gastroenterology 2007; 133:1210-8. [PMID: 17919495 DOI: 10.1053/j.gastro.2007.07.003] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Accepted: 06/28/2007] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS The MUC1 mucin is expressed on the cell surface of epithelial cells lining the gastric mucosa. Epidemiologic studies suggest that functional allelic variations in the MUC1 gene may play a role in human susceptibility to Helicobacter pylori-associated pathologies, including gastric adenocarcinoma. We have evaluated the impact of Muc1 expression on the colonization and pathogenesis of gastric Helicobacter infections. METHODS Wild-type and Muc1-deficient mice were infected with H pylori and colonization and gastritis levels determined. Primary gastric cells were used to examine the impact of Muc1 expression on bacterial adherence. RESULTS Mice lacking Muc1 were colonized by 5-fold more H pylori within 1 day of infection, and this difference was maintained for at least 2 months postinfection. Mice heterozygous for the null Muc1 allele developed intermediate bacterial colonization. Although wild-type mice developed only a mild gastritis when infected for 2 months with H pylori, Muc1(-/-) mice developed an atrophic gastritis marked by loss of parietal cells. We demonstrate H pylori adhesion to purified MUC1 and significantly increased adhesion to cultured murine Muc1 null gastric epithelial cells, suggesting that Muc1 acts as a decoy limiting binding to the cell surface. CONCLUSIONS Muc1 provides a protective barrier, which limits both acute and chronic colonization by H pylori, as well as playing a major role in limiting the inflammation induced by Helicobacter infection. We propose that Muc1 restricts access of H pylori to the epithelial surface, hence reducing exposure of the host to proinflammatory bacterial products.
Collapse
Affiliation(s)
- Michael A McGuckin
- Mucosal Diseases Program, Mater Medical Research Institute and University of Queensland, Mater Misericordiae Hospitals, Brisbane, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sutton P, Doidge C, Pinczower G, Wilson J, Harbour S, Swierczak A, Lee A. Effectiveness of vaccinationwith recombinant HpaA fromHelicobacter pyloriis influenced by host genetic background. ACTA ACUST UNITED AC 2007; 50:213-9. [PMID: 17567282 DOI: 10.1111/j.1574-695x.2006.00206.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Several studies have explored the production and immunogenicity of HpaA as a potential protective antigen against Helicobacter pylori but little is known regarding its protective capabilities. We therefore evaluated the protective efficacy of recombinant HpaA (rHpaA) as a candidate vaccine antigen against H. pylori. To explore the impact of genetic diversity, inbred and outbred mice were prophylactically and therapeutically immunized with rHpaA adjuvanted with cholera toxin (CT). Prophylactic immunization induced a reduction in bacterial colonization in BALB/c and QS mice, but was ineffective in C57BL/6 mice, despite induction of antigen-specific antibodies. By contrast, therapeutic immunization was effective in all three strains of mice. Prophylactic immunization with CT-adjuvanted rHpaA was more effective when delivered via the nasal route than following intragastric delivery in BALB/c mice. However, HpaA-mediated protection was inferior to that induced by bacterial lysate. Hence, protective efficacy is inducible with vaccines containing HpaA, most relevantly shown in an outbred population of mice. The effectiveness of protection induced by HpaA antigen was influenced by host genetics and was less effective than lysate. HpaA therefore has potential for the development of effective immunization against H. pylori but this would probably entail the antigen to be one component of a multiantigenic vaccine.
Collapse
MESH Headings
- Adhesins, Bacterial/immunology
- Adjuvants, Immunologic
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/blood
- Bacterial Vaccines/immunology
- Cholera Toxin/immunology
- Colony Count, Microbial
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay
- Female
- Helicobacter Infections/genetics
- Helicobacter Infections/prevention & control
- Helicobacter Infections/therapy
- Helicobacter pylori/growth & development
- Helicobacter pylori/immunology
- Immunoglobulin G/blood
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Stomach/microbiology
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Philip Sutton
- Centre for Animal Biotechnology, University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
44
|
Sutton P. Considering increased mouse stomach mass when calculating prophylactic vaccine efficacy against Helicobacter pylori. Helicobacter 2007; 12:210-2. [PMID: 17493000 DOI: 10.1111/j.1523-5378.2007.00494.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND Calculating the level of protection follow vaccination against Helicobacter pylori in mice is conventionally performed based on stomach mass. However, prophylactic vaccination is associated with a post-immunisation gastritis which results in considerable thickening of the gastric mucosa. RESULTS AND CONCLUSIONS Here, an analysis of nine experiments demonstrates that this post-immunisation gastritis can actually induce a significant increase in stomach mass. To determine whether this mass increase impacts upon the perceived level of protection, calculations were performed comparing colony-forming units (CFU) either per gram of tissue or per whole stomach. A trend was apparent towards increased stomach mass influencing the degree of protection if CFU were calculated per gram. However, as this effect was not significant, it appears valid to express CFU either per gram or per stomach (if age and gender matched mice are used).
Collapse
Affiliation(s)
- Philip Sutton
- Centre for Animal Biotechnology, Faculty of Veterinary Science, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
45
|
Taylor JM, Ziman ME, Fong J, Solnick JV, Vajdy M. Possible correlates of long-term protection against Helicobacter pylori following systemic or combinations of mucosal and systemic immunizations. Infect Immun 2007; 75:3462-9. [PMID: 17502399 PMCID: PMC1932914 DOI: 10.1128/iai.01470-06] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The ability to induce long-term immunity to Helicobacter pylori is necessary for an effective vaccine. This study was designed to establish the most efficient route(s) (systemic, mucosal, or a combination) of immunization for induction of long-term immunity and to define correlates of protection. Mice were immunized orally alone (oral group), intramuscularly (i.m.) alone (i.m. group), orally followed by i.m. (oral/i.m. group), or i.m. followed by orally (i.m./oral group). Long-term protective immunity to oral H. pylori challenge was observed 3 months after immunization through the i.m. or oral/i.m. route. Protection correlated with an increase in H. pylori-specific interleukin-12 and both immunoglobulin G1 (IgG1) and IgG2a serum titers following challenge. Mice that were not protected (oral or i.m./oral) had increased levels of IgA in both sera and Peyer's patches. This study demonstrates the ability to induce long-term immunity against H. pylori, provides correlates of protection, and illustrates the crucial role of the immunization route(s).
Collapse
Affiliation(s)
- Jennifer M Taylor
- Departments of Internal Medicine and Medical Microbiology and Immunology, Center for Comparative Medicine, University of California-Davis, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
46
|
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
|
47
|
Tan MP, Kaparakis M, Galic M, Pedersen J, Pearse M, Wijburg OLC, Janssen PH, Strugnell RA. Chronic Helicobacter pylori infection does not significantly alter the microbiota of the murine stomach. Appl Environ Microbiol 2007; 73:1010-3. [PMID: 17142378 PMCID: PMC1800740 DOI: 10.1128/aem.01675-06] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Accepted: 11/20/2006] [Indexed: 12/12/2022] Open
Abstract
We examined the impact of Helicobacter pylori infection on the murine gastric microbiota by culture and terminal-restriction fragment length polymorphism and found that neither acute nor chronic H. pylori infection substantially affected the gastric microbial composition. Interestingly, the total H. pylori burden detected by real-time PCR was significantly higher than that revealed by viable counts, suggesting that the antigenic load sustaining H. pylori-induced gastritis could be considerably higher than previously believed.
Collapse
Affiliation(s)
- Mai Ping Tan
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Robinson K, Argent RH, Atherton JC. The inflammatory and immune response to Helicobacter pylori infection. Best Pract Res Clin Gastroenterol 2007; 21:237-59. [PMID: 17382275 DOI: 10.1016/j.bpg.2007.01.001] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lifelong Helicobacter pylori infection and its associated gastric inflammation underlie peptic ulceration and gastric carcinogenesis. The immune and inflammatory responses to H. pylori are doubly responsible: gastric inflammation is the main mediator of pathology, and the immune and inflammatory response is ineffective, allowing lifelong bacterial persistence. However, despite inducing gastric inflammation, most infections do not cause disease, and bacterial, host and environmental factors determine individual disease risk. Although H. pylori avoids many innate immune receptors, specific virulence factors (including those encoded on the cag pathogenicity island) stimulate innate immunity to increase gastric inflammation and increase disease risk. An acquired T helper 1 response upregulates local immune effectors. The extent to which environmental factors (including parasite infection), host factors and H. pylori itself influence T-helper differentiation and regulatory T-cell responses remains controversial. Finally, effective vaccines have still not been developed: a better understanding of the immune response to H. pylori may help.
Collapse
Affiliation(s)
- Karen Robinson
- Wolfson Digestive Diseases Centre, University of Nottingham, C Floor, South Block, Queen's Medical Centre Campus, Nottingham University Hospital NHS Trust, Nottingham NG7 2UH, UK.
| | | | | |
Collapse
|
49
|
Avitzur Y, Galindo-Mata E, Jones NL. Oral vaccination against Helicobacter pylori infection is not effective in mice with Fas ligand deficiency. Dig Dis Sci 2005; 50:2300-6. [PMID: 16416178 DOI: 10.1007/s10620-005-3051-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Accepted: 03/08/2005] [Indexed: 01/05/2023]
Abstract
The aim of this study was to delineate the role of the Fas pathway in vaccination against Helicobacter pylori. C57BL/6 and Fas ligand-deficient (gld) mice were divided into 3 groups: control, H. pylori infected, and orally vaccinated (H. pylori whole cell sonicate and cholera toxin adjuvant). Oral vaccination prevented H. pylori colonization in 78% of C57BL/6 mice compared to only 18% of gld mice. Vaccination did not alter the degree of apoptosis in either strain of mice. Vaccination led to significant increase in interleukin (IL)-5 and IL-10 in C57BL/6 but not gld mice. H. pylori infection increased interferon (IFN)-gamma levels in C57BL/6 but not in gld mice while vaccination had no effect on IFN-gamma levels in either strain. Oral vaccination is not effective in Fas ligand-deficient mice likely owing to lack of effective cytokine responses. This indicates that the Fas pathway plays a critical role in promoting an appropriate effector response following H. pylori vaccination.
Collapse
Affiliation(s)
- Yaron Avitzur
- Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
50
|
Jeremy AHT, Du Y, Dixon MF, Robinson PA, Crabtree JE. Protection against Helicobacter pylori infection in the Mongolian gerbil after prophylactic vaccination. Microbes Infect 2005; 8:340-6. [PMID: 16213184 DOI: 10.1016/j.micinf.2005.06.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 06/21/2005] [Accepted: 06/28/2005] [Indexed: 12/23/2022]
Abstract
Vaccines against Helicobacter pylori could circumvent the problem of increasing antibiotic resistance. They would be particularly useful in developing countries, where re-infection rates are high following standard eradication regimes. The Mongolian gerbil is a good model for H. pylori infection, as the gastric pathology induced by infection is similar to that in humans. The H. pylori-induced inflammatory response in gerbils is considerably greater than in murine models. The aim of this study was to determine if gerbils could be vaccinated against H. pylori. Mongolian gerbils were vaccinated orally with an H. pylori whole cell sonicate preparation and cholera toxin adjuvant. Vaccinated gerbils and controls were challenged with the autologous H. pylori strain 42GX. All infection, and cholera toxin, control gerbils were H. pylori positive 6 weeks post-challenge. By contrast, a significant degree of protection was demonstrated in vaccinated gerbils. Only two of 10 of gerbils were H. pylori positive (P<0.001). Protection was associated with increased serum H. pylori IgG antibodies. Protected gerbils had histologically normal gastric mucosa and, in contrast to mice, no post-immunisation gastritis was evident. In the control groups, the degree of inflammation was variable, with some of the animals having corpus gastritis and corpus mucous metaplasia. The levels of gastric IL-12p40 and IFNgamma transcripts were significantly decreased in vaccinated animals compared to infection and cholera toxin controls (P<0.01). Gastric IL-10 and TGFbeta transcripts were found only at relatively low levels. These results demonstrate that Mongolian gerbils can be successfully vaccinated against H. pylori and protected from H. pylori-induced pathology.
Collapse
Affiliation(s)
- A H T Jeremy
- Molecular Medicine Unit, Level 7 Clinical Sciences Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | | | | | | | | |
Collapse
|