1
|
Francis JE, Skakic I, Majumdar D, Taki AC, Shukla R, Walduck A, Smooker PM. Solid Lipid Nanoparticles Delivering a DNA Vaccine Encoding Helicobacter pylori Urease A Subunit: Immune Analyses before and after a Mouse Model of Infection. Int J Mol Sci 2024; 25:1076. [PMID: 38256149 PMCID: PMC10816323 DOI: 10.3390/ijms25021076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
In this study, novel solid lipid particles containing the adjuvant lipid monophosphoryl lipid A (termed 'SLN-A') were synthesised. The SLN-A particles were able to efficiently bind and form complexes with a DNA vaccine encoding the urease alpha subunit of Helicobacter pylori. The resultant nanoparticles were termed lipoplex-A. In a mouse model of H. pylori infection, the lipoplex-A nanoparticles were used to immunise mice, and the resultant immune responses were analysed. It was found that the lipoplex-A vaccine was able to induce high levels of antigen-specific antibodies and an influx of gastric CD4+ T cells in vaccinated mice. In particular, a prime with lipoplex-A and a boost with soluble UreA protein induced significantly high levels of the IgG1 antibody, whereas two doses of lipoplex-A induced high levels of the IgG2c antibody. In this study, lipoplex-A vaccination did not lead to a significant reduction in H. pylori colonisation in a challenge model; however, these results point to the utility of the system for delivering DNA vaccine-encoded antigens to induce immune responses and suggest the ability to tailor those responses.
Collapse
Affiliation(s)
- Jasmine E. Francis
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (J.E.F.); (I.S.); (D.M.); (R.S.); (A.W.)
| | - Ivana Skakic
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (J.E.F.); (I.S.); (D.M.); (R.S.); (A.W.)
| | - Debolina Majumdar
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (J.E.F.); (I.S.); (D.M.); (R.S.); (A.W.)
| | - Aya C. Taki
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Ravi Shukla
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (J.E.F.); (I.S.); (D.M.); (R.S.); (A.W.)
| | - Anna Walduck
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (J.E.F.); (I.S.); (D.M.); (R.S.); (A.W.)
| | - Peter M. Smooker
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (J.E.F.); (I.S.); (D.M.); (R.S.); (A.W.)
| |
Collapse
|
2
|
Mohamed HRH, Elbasiouni SH, Farouk AH, Nasif KA, Nasraldin K, Safwat G. Alleviation of calcium hydroxide nanoparticles induced genotoxicity and gastritis by coadministration of calcium titanate and yttrium oxide nanoparticles in mice. Sci Rep 2023; 13:22011. [PMID: 38086889 PMCID: PMC10716372 DOI: 10.1038/s41598-023-49303-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Diverse applications of nanoparticles due to their unique properties has rapidly increased human exposure to numerous nanoparticles such as calcium hydroxide (Ca(OH)2), calcium titanate (CaTiO3), and yttrium oxide (Y2O3) nanoparticles almost in all aspect of daily life. However, very limited data are available on the effect of these nanoparticles on genomic DNA integrity and inflammation induction in the gastric tissues. Hence, this study estimated the effect of Ca(OH)2, CaTiO3, or/and Y2O3 nanoparticles multiple oral administration on the genomic DNA damage and inflammation induction in the mice gastric tissues. A suspension containing 50 mg/kg b.w of Ca(OH)2, CaTiO3, or Y2O3 nanoparticles were given orally to male mice separately or together simultaneously three times a week for two consecutive weeks. Multiple oral administration of Ca(OH)2 nanoparticles led to significant elevations in DNA damage induction and ROS generation, in contrast to the non-significant changes observed in the level of induced DNA damage and generated ROS after administration of CaTiO3 or Y2O3 nanoparticles separately or in combination with Ca(OH)2 nanoparticles. Oral administration of Ca(OH)2 nanoparticles alone also highly upregulated INOS and COX-2 genes expression and extremely decreased eNOS gene expression. However, high elevations in eNOS gene expression were detected after multiple administration of CaTiO3 and Y2O3 nanoparticles separately or together simultaneously with Ca(OH)2 nanoparticles. Meanwhile, non-remarkable changes were noticed in the expression level of INOS and COX-2 genes after administration of CaTiO3 and Y2O3 nanoparticles separately or simultaneously together with Ca(OH)2 nanoparticles. In conclusion: genomic DNA damage and inflammation induced by administration of Ca(OH)2 nanoparticles alone at a dose of 50 mg/kg were mitigated by about 100% when CaTiO3 and Y2O3 nanoparticles were coadministered with Ca(OH)2 nanoparticles until they reached the negative control level through altering the expression level of eNOS, INOS and COX-2 genes and scavenging gastric ROS. Therefore, further studies are recommended to investigate the toxicological properties of Ca(OH)2, CaTiO3 and Y2O3 nanoparticles and possibility of using CaTiO3 and Y2O3 nanoparticles to mitigate genotoxicity and inflammation induction by Ca(OH)2 nanoparticles.
Collapse
Affiliation(s)
- Hanan R H Mohamed
- Zoology Department Faculty of Science, Cairo University, Giza, Egypt.
| | - Salma H Elbasiouni
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th of October City, Egypt
| | - Ahmed H Farouk
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th of October City, Egypt
| | - Kirolls A Nasif
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th of October City, Egypt
| | - Karima Nasraldin
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th of October City, Egypt
| | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th of October City, Egypt
| |
Collapse
|
3
|
Mohamed HRH. Alleviation of Cadmium Chloride-Induced Acute Genotoxicity, Mitochondrial DNA Disruption, and ROS Generation by Chocolate Coadministration in Mice Liver and Kidney Tissues. Biol Trace Elem Res 2022; 200:3750-3761. [PMID: 34674108 DOI: 10.1007/s12011-021-02981-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/17/2021] [Indexed: 12/30/2022]
Abstract
Increased human exposure to cadmium compounds through ingesting contaminated food, water, and medications causes negative long-term health effects, which has led to the focus of recent researches on finding natural antioxidants to mitigate cadmium-induced toxicity. Therefore, the current study was undertaken to estimate the possible ameliorative effect of chocolate coadministration on acute cadmium chloride (CdCl2)-induced genomic instability and mitochondrial DNA damage in mice liver and kidney tissues. Concurrent administration of chocolate with CdCl2 dramatically decreased the DNA damage level and the number of apoptotic and necrotic cells compared to mice given CdCl2 alone. Extra-production of reactive oxygen species and increased expression of inducible nitric oxide synthase and heat shock proteins genes caused by CdCl2 administration were also highly decreased after chocolate coadministration. Conversely, chocolate coadministration restored the integrity of the mitochondrial membrane potential disrupted by CdCl2 administration, as well as the mitochondrial DNA copy number and expression level of heme oxygenase-1 gene were significantly upregulated after chocolate coadministration with CdCl2. Thus, it was concluded that the coadministration of chocolate alleviated CdCl2-induced genomic instability and mitochondrial DNA damage through its antioxidative and free radical scavenging capabilities, making chocolate a promising ameliorative product and recommended for inclusion in the daily human diet.
Collapse
|
4
|
Vaillant L, Oster P, McMillan B, Orozco Fernandez E, Velin D. GM-CSF is key in the efficacy of vaccine-induced reduction of Helicobacter pylori infection. Helicobacter 2022; 27:e12875. [PMID: 35092634 PMCID: PMC9285700 DOI: 10.1111/hel.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/22/2021] [Accepted: 01/16/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) colonizes the human gastric mucosa with a high worldwide prevalence. Currently, H. pylori is eradicated by the use of antibiotics. However, elevated antibiotic resistance suggests new therapeutic strategies need to be envisioned: one approach being prophylactic vaccination. Pre-clinical and clinical data show that a urease-based vaccine is efficient in decreasing H. pylori infection through the mobilization of T helper (Th) cells, especially Th17 cells. Th17 cells produce interleukins such as IL-22 and IL-17, among others, and are key players in vaccine efficacy. Recently, granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing Th17 cells have been identified. AIM This study explores the possibility that GM-CSF plays a role in the reduction of H. pylori infection following vaccination. RESULTS We demonstrate that GM-CSF+ IL-17+ Th17 cells accumulate in the stomach mucosa of H. pylori infected mice during the vaccine-induced reduction of H. pylori infection. Secondly, we provide evidence that vaccinated GM-CSF deficient mice only modestly reduce H. pylori infection. Conversely, we observe that an increase in GM-CSF availability reduces H. pylori burden in chronically infected mice. Thirdly, we show that GM-CSF, by acting on gastric epithelial cells, promotes the production of βdefensin3, which exhibits H. pylori bactericidal activities. CONCLUSION Taken together, we demonstrate a key role of GM-CSF, most probably originating from Th17 cells, in the vaccine-induced reduction of H. pylori infection.
Collapse
Affiliation(s)
- Laurie Vaillant
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Paul Oster
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Brynn McMillan
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Eulalia Orozco Fernandez
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| | - Dominique Velin
- Service of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
5
|
Xu N, Ruan G, Liu W, Hu C, Huang A, Zeng Z, Luo S, Zhang Z, Fan M, Ye F, Xi T, Xing Y. Vaccine-induced gastric CD4 + tissue-resident memory T cells proliferate in situ to amplify immune response against Helicobacter pylori insult. Helicobacter 2019; 24:e12652. [PMID: 31414552 DOI: 10.1111/hel.12652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tissue-resident memory T cells accelerate the clearance of pathogens during recall response. However, whether CD4+ TRM cells themselves can provide gastric immunity is unclear. MATERIALS AND METHODS We established a parabiosis model between the enhanced green fluorescent protein and wild-type mice that the circulation system was shared, and the wild-type partner was vaccinated with H pylori vaccine composed of CCF and silk fibroin in gastric subserous layer to induce gastric EGFP+ CD4+ TRM cells. Antigen-specific EGFP+ CD4+ T cells and proliferous TRM cells were analyzed by flow cytometry. The colonization of H pylori was detected by quantitative real-time PCR. EGFP+ CD4+ TRM cells and the inflammation of the stomach were observed by histology. RESULTS A parabiosis animal model was employed to identify the cells that introduced by vaccination in GSL. Antigen-specific EGFP+ CD4+ T cells could be detected at day 7 post-vaccination. Thirty days later, EGFP+ CD4+ TRM cells were established with a phenotype of CD69+ CD103- . Of note, we found that when circulating lymphocytes were depleted by FTY720 administration, these TRM cells could proliferate in situ and differentiate into effector Th1 cells after H pylori challenge. A decrease in H pylori colonization was observed in the vaccinated mice but not unvaccinated mice. Further, we found that although FTY720 was administrated, mounted pro-inflammatory myeloid cells still emerged in the stomach of the vaccinated mice, which might contribute to the reduction of H pylori colonization. CONCLUSIONS Our study reveals that H pylori vaccine-induced CD4+ TRM cells can proliferate and differentiate in situ to enhance gastric local immunity during recall response.
Collapse
Affiliation(s)
- Ningyin Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Guojing Ruan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Chupeng Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - An Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhenxing Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Menghui Fan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Feng Ye
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Mechanisms of Inflammasome Signaling, microRNA Induction and Resolution of Inflammation by Helicobacter pylori. Curr Top Microbiol Immunol 2019; 421:267-302. [PMID: 31123893 DOI: 10.1007/978-3-030-15138-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammasome-controlled transcription and subsequent cleavage-mediated activation of mature IL-1β and IL-18 cytokines exemplify a crucial innate immune mechanism to combat intruding pathogens. Helicobacter pylori represents a predominant persistent infection in humans, affecting approximately half of the population worldwide, and is associated with the development of chronic gastritis, peptic ulcer disease, and gastric cancer. Studies in knockout mice have demonstrated that the pro-inflammatory cytokine IL-1β plays a central role in gastric tumorigenesis. Infection by H. pylori was recently reported to stimulate the inflammasome both in cells of the mouse and human immune systems. Using mouse models and in vitro cultured cell systems, the bacterial pathogenicity factors and molecular mechanisms of inflammasome activation have been analyzed. On the one hand, it appears that H. pylori-stimulated IL-1β production is triggered by engagement of the immune receptors TLR2 and NLRP3, and caspase-1. On the other hand, microRNA hsa-miR-223-3p is induced by the bacteria, which controls the expression of NLRP3. This regulating effect by H. pylori on microRNA expression was also described for more than 60 additionally identified microRNAs, indicating a prominent role for inflammatory and other responses. Besides TLR2, TLR9 becomes activated by H. pylori DNA and further TLR10 stimulated by the bacteria induce the secretion of IL-8 and TNF, respectively. Interestingly, TLR-dependent pathways can accelerate both pro- and anti-inflammatory responses during H. pylori infection. Balancing from a pro-inflammation to anti-inflammation phenotype results in a reduction in immune attack, allowing H. pylori to persistently colonize and to survive in the gastric niche. In this chapter, we will pinpoint the role of H. pylori in TLR- and NLRP3 inflammasome-dependent signaling together with the differential functions of pro- and anti-inflammatory cytokines. Moreover, the impact of microRNAs on H. pylori-host interaction will be discussed, and its role in resolution of infection versus chronic infection, as well as in gastric disease development.
Collapse
|
7
|
Lin LCW, Chattopadhyay S, Lin JC, Hu CMJ. Advances and Opportunities in Nanoparticle- and Nanomaterial-Based Vaccines against Bacterial Infections. Adv Healthc Mater 2018; 7:e1701395. [PMID: 29508547 DOI: 10.1002/adhm.201701395] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/22/2018] [Indexed: 02/06/2023]
Abstract
As the dawn of the postantibiotic era we approach, antibacterial vaccines are becoming increasingly important for managing bacterial infection and reducing the need for antibiotics. Despite the success of vaccination, vaccines remain unavailable for many pressing microbial diseases, including tuberculosis, chlamydia, and staphylococcus infections. Amid continuing research efforts in antibacterial vaccine development, the advancement of nanomaterial engineering has brought forth new opportunities in vaccine designs. With increasing knowledge in antibacterial immunity and immunologic adjuvants, innovative nanoparticles are designed to elicit the appropriate immune responses for effective antimicrobial defense. Rationally designed nanoparticles are demonstrated to overcome delivery barriers to shape the adaptive immunity. This article reviews the advances in nanoparticle- and nanomaterial-based antibacterial vaccines and summarizes the development of nanoparticulate adjuvants for immune potentiation against microbial pathogens. In addition, challenges and progress in ongoing antibacterial vaccine development are discussed to highlight the opportunities for future vaccine designs.
Collapse
Affiliation(s)
- Leon Chien-Wei Lin
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Saborni Chattopadhyay
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Jung-Chen Lin
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences; Academia Sinica; 128, Sec. 2, Academia Road Nangang District Taipei 11529 Taiwan
| |
Collapse
|
8
|
WATANABE T, ASANO N, KUDO M, STROBER W. Nucleotide-binding oligomerization domain 1 and gastrointestinal disorders. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:578-599. [PMID: 29021509 PMCID: PMC5743859 DOI: 10.2183/pjab.93.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 05/31/2017] [Indexed: 05/08/2023]
Abstract
Nucleotide-binding oligomerization domain 1 (NOD1) is an intracellular sensor that detects small peptides derived from the cell wall component of intestinal microflora. NOD1 is expressed in both non-hematopoietic cells such as epithelial cells and hematopoietic cells such as antigen-presenting cells. Detection of its ligand by NOD1 leads to innate immune responses through activation of nuclear factor kappa B and type I interferon as well as induction of autophagy. Innate immune responses through NOD1 activation play an indispensable role both in host defense against microbial infection and in the development of gastrointestinal disorders. Of particular importance, NOD1-mediated innate immune responses are associated with mucosal host defenses against Helicobacter pylori (H. pylori) infection of the stomach and with the development of pancreatitis. In this review, we discuss the molecular mechanisms by which NOD1 activation leads to the development of H. pylori-related gastric diseases and pancreatitis.
Collapse
Affiliation(s)
- Tomohiro WATANABE
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
- Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, U.S.A.
| | - Naoki ASANO
- Division of Gastroenterology and Hepatology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masatoshi KUDO
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Warren STROBER
- Mucosal Immunity Section, Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, U.S.A.
| |
Collapse
|
9
|
Konorev MR, Andronova TM, Matveenko ME. [Use of probiotics and probiotic-based immunomodulators as adjuvant therapy for Helicobacter pylori eradication]. TERAPEVT ARKH 2016. [PMID: 28635890 DOI: 10.17116/terarkh20168812140-148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
At present, Helicobacter pylori (Нр) infection is the most common chronic bacterial infection in humans, the pathogen of which colonizes approximately 50% of the world's population. Hp eradication is required to control complications of Hp-related diseases (gastric and duodenal ulcers). Nevertheless, a number of investigations have demonstrated widespread antibacterial therapy inefficiency due to Hp antibiotic resistance and patient non-compliance with treatment regimens. Due to the growing need to elaborate alternative eradication regimens, some researchers have drawn their attention to probiotics and immunomodulators derived from Lactobacillus in particular for eradication therapy in Нp-positive patients to enhance the effect of antibacterial drugs. The review analyzes the results of 10 meta-analyses of randomized clinical trials with a similar design, which were published in 2007 to 2015, and other clinical trials assessing the role of probiotics and probiotic-based immunomodulators as an adjuvant therapy for Hp eradication. The results of the analysis have established that Lactobacillus strain-containing probiotics, both monocomponent probiotics and those as part of multicomponent ones, when used as an adjunct to anti-Hp therapy, significantly increase the level of Нp eradication by 8.1-20.0% (p<0.05; Level of Evidence, 1A; Recommendation Grade A). The use of N-acetylglucosaminyl-N-acetylmuramyl dipeptide (Licopid, a Lactobacillus bulgaricus-based immunomodulator) 0.001 and 0.01 g/day as an adjuvant to first-line triple anti-Hp therapy was shown to increase the level of Hp eradication by 7.1-8.9%. The intake of licopid 0.001 and 0.01 g/day during 7-day triple anti-Hp therapy results in the absence of recurrent Hp infection, as compared with 7- and 14-day treatment protocols without licopid, and leads to a significantly low incidence of Hp reinfection within 2-5 years after successful bacterial eradication, as compared with the 7-day protocol without adjuvant therapy with glucosaminylmuramyl dipeptide (p<0.05).
Collapse
Affiliation(s)
- M R Konorev
- Vitebsk State Medical University, Vitebsk, Belarus
| | | | | |
Collapse
|
10
|
Raghavan S, Quiding-Järbrink M. Vaccination Against Helicobacter pylori Infection. HELICOBACTER PYLORI RESEARCH 2016:575-601. [DOI: 10.1007/978-4-431-55936-8_25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
11
|
Helicobacter pylori and T Helper Cells: Mechanisms of Immune Escape and Tolerance. J Immunol Res 2015. [PMID: 26525279 DOI: 10.1155/7015/981328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Helicobacter pylori colonizes the gastric mucosa of at least half of the human population, causing a worldwide infection that appears in early childhood and if not treated, it can persist for life. The presence of symptoms and their severity depend on bacterial components, host susceptibility, and environmental factors, which allow H. pylori to switch between commensalism and pathogenicity. H. pylori-driven interactions with the host immune system underlie the persistence of the infection in humans, since the bacterium is able to interfere with the activity of innate and adaptive immune cells, reducing the inflammatory response in its favour. Gastritis due to H. pylori results from a complex interaction between several T cell subsets. In particular, H. pylori is known to induce a T helper (Th)1/Th17 cell response-driven gastritis, whose impaired modulation caused by the bacterium is thought to sustain the ongoing inflammatory condition and the unsuccessful clearing of the infection. In this review we discuss the current findings underlying the mechanisms implemented by H. pylori to alter the T helper lymphocyte proliferation, thus facilitating the development of chronic infections and allowing the survival of the bacterium in the human host.
Collapse
|
12
|
Helicobacter pylori and T Helper Cells: Mechanisms of Immune Escape and Tolerance. J Immunol Res 2015; 2015:981328. [PMID: 26525279 PMCID: PMC4615206 DOI: 10.1155/2015/981328] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/29/2015] [Indexed: 12/18/2022] Open
Abstract
Helicobacter pylori colonizes the gastric mucosa of at least half of the human population, causing a worldwide infection that appears in early childhood and if not treated, it can persist for life. The presence of symptoms and their severity depend on bacterial components, host susceptibility, and environmental factors, which allow H. pylori to switch between commensalism and pathogenicity. H. pylori-driven interactions with the host immune system underlie the persistence of the infection in humans, since the bacterium is able to interfere with the activity of innate and adaptive immune cells, reducing the inflammatory response in its favour. Gastritis due to H. pylori results from a complex interaction between several T cell subsets. In particular, H. pylori is known to induce a T helper (Th)1/Th17 cell response-driven gastritis, whose impaired modulation caused by the bacterium is thought to sustain the ongoing inflammatory condition and the unsuccessful clearing of the infection. In this review we discuss the current findings underlying the mechanisms implemented by H. pylori to alter the T helper lymphocyte proliferation, thus facilitating the development of chronic infections and allowing the survival of the bacterium in the human host.
Collapse
|
13
|
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
|
14
|
Nedrud JG, Bagheri N, Schön K, Xin W, Bergroth H, Eliasson DG, Lycke NY. Subcomponent vaccine based on CTA1-DD adjuvant with incorporated UreB class II peptides stimulates protective Helicobacter pylori immunity. PLoS One 2013; 8:e83321. [PMID: 24391754 PMCID: PMC3877028 DOI: 10.1371/journal.pone.0083321] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 11/01/2013] [Indexed: 11/29/2022] Open
Abstract
A mucosal vaccine against Helicobacter pylori infection could help prevent gastric cancers and peptic ulcers. While previous attempts to develop such a vaccine have largely failed because of the requirement for safe and effective adjuvants or large amounts of well defined antigens, we have taken a unique approach to combining our strong mucosal CTA1-DD adjuvant with selected peptides from urease B (UreB). The protective efficacy of the selected peptides together with cholera toxin (CT) was first confirmed. However, CT is a strong adjuvant that unfortunately is precluded from clinical use because of its toxicity. To circumvent this problem we have developed a derivative of CT, the CTA1-DD adjuvant, that has been found safe in non-human primates and equally effective compared to CT when used intranasally. We genetically fused the selected peptides into the CTA1-DD plasmid and found after intranasal immunizations of Balb/c mice using purified CTA1-DD with 3 copies of an H. pylori urease T cell epitope (CTA1-UreB3T-DD) that significant protection was stimulated against a live challenge infection. Protection was, however, weaker than with the gold standard, bacterial lysate+CT, but considering that we only used a single epitope in nanomolar amounts the results convey optimism. Protection was associated with enhanced Th1 and Th17 immunity, but immunizations in IL-17A-deficient mice revealed that IL-17 may not be essential for protection. Taken together, we have provided evidence for the rational design of an effective mucosal subcomponent vaccine against H. pylori infection based on well selected protective epitopes from relevant antigens incorporated into the CTA1-DD adjuvant platform.
Collapse
Affiliation(s)
- John G. Nedrud
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Nayer Bagheri
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Karin Schön
- Mucosal Immunobiology and Vaccine Research Center (MIVAC) and the Department of Microbiolgy and Immunology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Wei Xin
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Hilda Bergroth
- Mucosal Immunobiology and Vaccine Research Center (MIVAC) and the Department of Microbiolgy and Immunology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Dubravka Grdic Eliasson
- Mucosal Immunobiology and Vaccine Research Center (MIVAC) and the Department of Microbiolgy and Immunology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| | - Nils Y. Lycke
- Mucosal Immunobiology and Vaccine Research Center (MIVAC) and the Department of Microbiolgy and Immunology, Institute of Biomedicine, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
15
|
Zawahir S, Czinn SJ, Nedrud JG, Blanchard TG. Vaccinating against Helicobacter pylori in the developing world. Gut Microbes 2013; 4:568-76. [PMID: 24253617 PMCID: PMC3928166 DOI: 10.4161/gmic.27093] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Helicobacter pylori infects more than half the world's population and in developing nations the incidence can be over 90%. The morbidity and mortality associated with H. pylori-associated diseases including ulcers and gastric cancer therefore, disproportionately impact the developing world. Mice have been used extensively to demonstrate the feasibility of developing a vaccine for H. pylori infection, and for testing antigens, routes of immunization, dose, and adjuvants. These successes however, have not translated well in clinical trials. Although there are examples where immune responses have been activated, there are few instances of achieving a reduced bacterial load. In vivo and in vitro analyses in both mice and humans demonstrates that the host responds to H. pylori infection through the activation of immunoregulatory mechanisms designed to suppress the anti-H. pylori response. Improved vaccine efficacy therefore, will require the inclusion of factors that over-ride or re-program these immunoregulatory rersponse mechanisms.
Collapse
Affiliation(s)
- Shamila Zawahir
- Department of Pediatrics; University of Maryland School of Medicine; Baltimore, MD USA
| | - Steven J Czinn
- Department of Pediatrics; University of Maryland School of Medicine; Baltimore, MD USA
| | - John G Nedrud
- Department of Pathology; Case Western Reserve University School of Medicine; Cleveland, OH USA
| | - Thomas G Blanchard
- Department of Pediatrics; University of Maryland School of Medicine; Baltimore, MD USA,Correspondence to: Thomas G Blanchard,
| |
Collapse
|
16
|
TLR2 mediates Helicobacter pylori-induced tolerogenic immune response in mice. PLoS One 2013; 8:e74595. [PMID: 24058595 PMCID: PMC3772856 DOI: 10.1371/journal.pone.0074595] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 08/03/2013] [Indexed: 12/11/2022] Open
Abstract
We have shown that Helicobacter pylori induces tolerogenic programming of dendritic cells and inhibits the host immune response. Toll-like receptors (TLRs) represent a class of transmembrane pattern recognition receptors essential for microbial recognition and control of the innate immune response. In this study, we examined the role of TLRs in mediating H. pylori tolerogenic programming of dendritic cells and their impact on anti-H. pylori immunity using C57BL/6 wild-type and TLR2-knockout (TLR2KO) mice. We analyzed the response of TLR2KO bone marrow-derived dendritic cells (BMDCs) to H. pylori SS1 stimulation and the outcome of chronic H. pylori infection in TLR2KO mice. We showed that H. pylori-stimulated BMDCs upregulated the expression of TLR2, but not TLR4, TLR5, or TLR9. H. pylori-stimulated BMDCs from TLRKO mice induced lower Treg and Th17 responses, but a higher IFN-γ response compared to H. pylori-stimulated BMDCs from wild-type mice. In vivo analyses following an H. pylori infection of 2 months duration showed a lower degree of gastric H. pylori colonization in TLR2KO mice and more severe gastric immunopathology compared to WT mice. The gastric mucosa of the infected TLR2KO mice showed a lower mRNA expression of Foxp3, IL-10, and IL-17A, but higher expression of IFN-γ compared to the gastric mRNA expression in infected wild-type mice. Moreover, the H. pylori-specific Th1 response was higher and the Treg and Th17 responses were lower in the spleens of infected TLR2KO mice compared to infected WT mice. Our data indicate that H. pylori mediates immune tolerance through TLR2-derived signals and inhibits Th1 immunity, thus evading host defense. TLR2 may be an important target in the modulation of the host response to H. pylori.
Collapse
|
17
|
Ding H, Nedrud JG, Blanchard TG, Zagorski BM, Li G, Shiu J, Xu J, Czinn SJ. Th1-mediated immunity against Helicobacter pylori can compensate for lack of Th17 cells and can protect mice in the absence of immunization. PLoS One 2013; 8:e69384. [PMID: 23874957 PMCID: PMC3708891 DOI: 10.1371/journal.pone.0069384] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 06/09/2013] [Indexed: 12/29/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection can be significantly reduced by immunization in mice. Th17 cells play an essential role in the protective immune response. Th1 immunity has also been demonstrated to play a role in the protective immune response and can compensate in the absence of IL-17. To further address the potential of Th1 immunity, we investigated the efficacy of immunization in mice deficient in IL-23p19, a cytokine that promotes Th17 cell development. We also examined the course of Helicobacter infection in unimmunized mice treated with Th1 promoting cytokine IL-12. C57BL/6, IL-12 p35 KO, and IL-23 p19 KO mice were immunized and challenged with H. pylori. Protective immunity was evaluated by CFU determination and QPCR on gastric biopsies. Gastric and splenic IL-17 and IFNγ levels were determined by PCR or by ELISA. Balb/c mice were infected with H. felis and treated with IL-12 therapy and the resulting gastric bacterial load and inflammatory response were assessed by histologic evaluation. Vaccine induced reductions in bacterial load that were comparable to wild type mice were observed in both IL-12 p35 and IL-23 p19 KO mice. In the absence of IL-23 p19, IL-17 levels remained low but IFNγ levels increased significantly in both immunized challenged and unimmunized/challenged mice. Additionally, treatment of H. felis-infected Balb/c mice with IL-12 resulted in increased gastric inflammation and the eradication of bacteria in most mice. These data suggest that Th1 immunity can compensate for the lack of IL-23 mediated Th17 responses, and that protective Th1 immunity can be induced in the absence of immunization through cytokine therapy of the infected host.
Collapse
Affiliation(s)
- Hua Ding
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - John G. Nedrud
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
| | - Thomas G. Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | | | - Guanghui Li
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jessica Shiu
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jinghua Xu
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Steven J. Czinn
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
18
|
Chen J, Lin L, Li N, She F. Enhancement of Helicobacter pylori outer inflammatory protein DNA vaccine efficacy by co-delivery of interleukin-2 and B subunit heat-labile toxin gene encoded plasmids. Microbiol Immunol 2012; 56:85-92. [PMID: 22150716 DOI: 10.1111/j.1348-0421.2011.00409.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Development of an effective vaccine for controlling H. pylori-associated infection, which is present in about half the people in the world, is a priority. The H. pylori outer inflammatory protein (oipA) has been demonstrated to be a potential antigen for a vaccine. In the present study, use of oipA gene encoded construct (poipA) for C57BL/6 mice vaccination was investigated. Whether co-delivery of IL-2 gene encoded construct (pIL-2) and B subunit heat-labile toxin of Escherichia coli gene encoded construct (pLTB) can modulate the immune response and enhance DNA vaccine efficacy was also explored. Our results demonstrated that poipA administered intradermally ('gene gun' immunization) promoted a strong Th2 immune response, whereas co-delivery of either pIL-2 or pLTB adjuvant elicited a Th1-biased immune response. PoipA administered with both pIL-2 and pLTB adjuvants promoted a strong Th1 immune response. Regardless of the different immune responses promoted by the various vaccination regimes, all immunized mice had smaller bacterial loads after H. pylori challenge than did PBS negative and pVAX1 mock controls. Co-delivery of adjuvant(s) enhances poipA DNA vaccine efficacy by shifting the immune response from being Th2 to being Th1-biased, which results in a greater reduction in bacterial load after H. pylori challenge. Both prophylactic and therapeutic vaccination can achieve sterile immunity in some subjects.
Collapse
Affiliation(s)
- Jiansen Chen
- Clinical Laboratory Department, Fujian Medical University Affiliated Union Hospital, Fuzhou 350001, Fujian, China
| | | | | | | |
Collapse
|
19
|
|
20
|
DeLyria ES, Nedrud JG, Ernst P, Alam MS, Redline RW, Ding H, Czinn SJ, Xu J, Blanchard T. Vaccine-induced immunity against Helicobacter pylori in the absence of IL-17A. Helicobacter 2011; 16:169-78. [PMID: 21585602 PMCID: PMC3107727 DOI: 10.1111/j.1523-5378.2011.00839.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is a gram negative bacterium that can cause diseases such as peptic ulcers and gastric cancer. IL-17A, a proinflammatory cytokine that can induce the production of CXC chemokines for neutrophil recruitment, has recently been shown to be elevated in both H. pylori-infected patients and mice. Furthermore, studies in mouse models of vaccination have reported levels significantly increased over infected, unimmunized mice and blocking of IL-17A during the challenge phase in immunized mice reduces protective immunity. Because many aspects of immunity had redundant or compensatory mechanisms, we investigated whether mice could be protectively immunized when IL-17A function is absent during the entire immune response using IL-17A and IL-17A receptor knockout (KO) mice immunized against H. pylori. MATERIALS AND METHODS Gastric biopsies were harvested from naïve, unimmunized/challenged, and immunized/challenged wild type (WT) and KO mice and analyzed for inflammation, neutrophil, and bacterial levels. Groups of IL-17A KO mice were also treated with anti-IFNγ or control antibodies. RESULTS Surprisingly, all groups of immunized KO mice reduced their bacterial loads comparably to WT mice. The gastric neutrophil counts did not vary significantly between IL-17A KO and WT mice, whereas IL-17RA KO mice had on average a four-fold decrease compared to WT. Additionally, we performed an immunization study with CXCR2 KO mice and observed significant gastric neutrophils and reduction in bacterial load. CONCLUSION These data suggest that there are compensatory mechanisms for protection against H. pylori and for neutrophil recruitment in the absence of an IL-17A-CXC chemokine pathway.
Collapse
Affiliation(s)
- Elizabeth S. DeLyria
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH
| | - John G. Nedrud
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH
| | - Peter Ernst
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Virginia, Charlottesville, VA
| | - M. Samiul Alam
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Raymond W. Redline
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH
| | - Hua Ding
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD
| | - Steven J. Czinn
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD
| | - Jinghua Xu
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD
| | - T.G. Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
21
|
Lewis ND, Asim M, Barry DP, de Sablet T, Singh K, Piazuelo MB, Gobert AP, Chaturvedi R, Wilson KT. Immune evasion by Helicobacter pylori is mediated by induction of macrophage arginase II. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:3632-41. [PMID: 21296975 PMCID: PMC3069806 DOI: 10.4049/jimmunol.1003431] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Helicobacter pylori infection persists for the life of the host due to the failure of the immune response to eradicate the bacterium. Determining how H. pylori escapes the immune response in its gastric niche is clinically important. We have demonstrated in vitro that macrophage NO production can kill H. pylori, but induction of macrophage arginase II (Arg2) inhibits inducible NO synthase (iNOS) translation, causes apoptosis, and restricts bacterial killing. Using a chronic H. pylori infection model, we determined whether Arg2 impairs host defense in vivo. In C57BL/6 mice, expression of Arg2, but not arginase I, was abundant and localized to gastric macrophages. Arg2(-/-) mice had increased histologic gastritis and decreased bacterial colonization compared with wild-type (WT) mice. Increased gastritis scores correlated with decreased colonization in individual Arg2(-/-) mice but not in WT mice. When mice infected with H. pylori were compared, Arg2(-/-) mice had more gastric macrophages, more of these cells were iNOS(+), and these cells expressed higher levels of iNOS protein, as determined by flow cytometry and immunofluorescence microscopy. There was enhanced nitrotyrosine staining in infected Arg2(-/-) versus WT mice, indicating increased NO generation. Infected Arg2(-/-) mice exhibited decreased macrophage apoptosis, as well as enhanced IFN-γ, IL-17a, and IL-12p40 expression, and reduced IL-10 levels consistent with a more vigorous Th1/Th17 response. These studies demonstrate that Arg2 contributes to the immune evasion of H. pylori by limiting macrophage iNOS protein expression and NO production, mediating macrophage apoptosis, and restraining proinflammatory cytokine responses.
Collapse
Affiliation(s)
- Nuruddeen D. Lewis
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37240
| | - Mohammad Asim
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
| | - Daniel P. Barry
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
| | - Thibaut de Sablet
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
| | - Kshipra Singh
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
| | - Maria B. Piazuelo
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
| | - Alain P. Gobert
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
- Institut National de la Recherche Agronomique, Unité de Microbiologie UR454, Saint-Genès-Champanelle, France
| | - Rupesh Chaturvedi
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
| | - Keith T. Wilson
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37240
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37240
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37212
| |
Collapse
|
22
|
Sundquist M, Quiding-Järbrink M. Helicobacter pylori and its effect on innate and adaptive immunity: new insights and vaccination strategies. Expert Rev Gastroenterol Hepatol 2010; 4:733-44. [PMID: 21108593 DOI: 10.1586/egh.10.71] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Infection with the gastric bacterium Helicobacter pylori invariably leads to active chronic gastritis, and is strongly correlated to peptic ulcer disease, gastric adenocarcinoma and mucosa-associated lymphoid tissue lymphoma. The infection leads to local accumulation of inflammatory cells and strong activation of B- and T-cell immunity. Still, the immune response can not eliminate the bacteria, and unless antibiotic treatment is used, the infection is usually lifelong. During the last few years, several immunomodulatory properties of H. pylori have been described, which probably contribute to the inability of the immune system to eradicate the bacterium. Another factor promoting bacterial persistence is probably the induction of a substantial regulatory T-cell response by the infection. Several different immunization schedules have resulted in protective immunity in animal models, while in humans no reliable vaccine is available as yet. In this article, we describe the innate and adaptive immune responses to H. pylori, and the attempts to create an effective vaccine.
Collapse
Affiliation(s)
- Malin Sundquist
- Department of Microbiology and Immunology, The Sahlgrenska Academy at the University of Gothenburg, P.O. Box 435, 405 30 Göteborg, Sweden
| | | |
Collapse
|
23
|
Allison CC, Ferrero RL. Role of virulence factors and host cell signaling in the recognition of Helicobacter pylori and the generation of immune responses. Future Microbiol 2010; 5:1233-55. [PMID: 20722601 DOI: 10.2217/fmb.10.84] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori colonizes a large proportion of the world's population, with infection invariably leading to chronic, lifelong gastritis. While the infection often persists undiagnosed and without causing severe pathology, there are a number of host, bacterial and environmental factors that can influence whether infection provokes a mild inflammatory response or results in significant morbidity. Intriguingly, the most virulent H. pylori strains appear to deliberately induce the epithelial signaling cascades responsible for activating the innate immune system. While the reason for this remains unclear, the resulting adaptive immune responses are largely ineffective in clearing the bacterium once infection has become established and, as a result, inflammation likely causes more damage to the host itself.
Collapse
Affiliation(s)
- Cody C Allison
- Centre for Innate Immunity & Infectious Diseases, Monash Institute of Medical Research, Clayton, Australia.
| | | |
Collapse
|
24
|
Quiding-Järbrink M, Raghavan S, Sundquist M. Enhanced M1 macrophage polarization in human helicobacter pylori-associated atrophic gastritis and in vaccinated mice. PLoS One 2010; 5:e15018. [PMID: 21124899 PMCID: PMC2990716 DOI: 10.1371/journal.pone.0015018] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 10/07/2010] [Indexed: 12/16/2022] Open
Abstract
Background Infection with Helicobacter pylori triggers a chronic gastric inflammation that can progress to atrophy and gastric adenocarcinoma. Polarization of macrophages is a characteristic of both cancer and infection, and may promote progression or resolution of disease. However, the role of macrophages and their polarization during H. pylori infection has not been well defined. Methodology/Principal Findings By using a mouse model of infection and gastric biopsies from 29 individuals, we have analyzed macrophage recruitment and polarization during H. pylori infection by flow cytometry and real-time PCR. We found a sequential recruitment of neutrophils, eosinophils and macrophages to the gastric mucosa of infected mice. Gene expression analysis of stomach tissue and sorted macrophages revealed that gastric macrophages were polarized to M1 after H. pylori infection, and this process was substantially accelerated by prior vaccination. Human H. pylori infection was characterized by a mixed M1/M2 polarization of macrophages. However, in H. pylori-associated atrophic gastritis, the expression of inducible nitric oxide synthase was markedly increased compared to uncomplicated gastritis, indicative of an enhanced M1 macrophage polarization in this pre-malignant lesion. Conclusions/Significance These results show that vaccination of mice against H. pylori amplifies M1 polarization of gastric macrophages, and that a similar enhanced M1 polarization is present in human H. pylori-induced atrophic gastritis.
Collapse
Affiliation(s)
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Malin Sundquist
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
25
|
Proinflammatory cytokine gene expression in the stomach correlates with vaccine-induced protection against Helicobacter pylori infection in mice: an important role for interleukin-17 during the effector phase. Infect Immun 2010; 79:879-86. [PMID: 21078851 DOI: 10.1128/iai.00756-10] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD4(+) T cells have been shown to be essential for vaccine-induced protection against Helicobacter pylori infection in mice. Less is known about the relative contributions of individual cell subpopulations, such as T(h)1 and T(h)17 cells, and their associated cytokines. The aim of the present study was to find immune correlates to vaccine-induced protection and further study their role in protection against H. pylori infection. Immunized and unimmunized mice were challenged with H. pylori, and immune responses were compared. Vaccine-induced protection was assessed by measuring H. pylori colonization in the stomach. Gastric gene expression of T(h)1- and/or T(h)17-associated cytokines was analyzed by quantitative PCR, and contributions of individual cytokines to protection were evaluated by antibody-mediated in vivo neutralization. By analyzing immunized and unimmunized mice, a significant inverse correlation between the levels of interleukin-12p40 (IL-12p40), tumor necrosis factor alpha (TNF), gamma interferon (IFN-γ), and IL-17 gene expression and the number of H. pylori bacteria in the stomachs of individual animals after challenge could be demonstrated. In a kinetic study, upregulation of TNF, IFN-γ, and IL-17 coincided with vaccine-induced protection at 7 days after H. pylori challenge and was sustained for at least 21 days. In vivo neutralization of these cytokines during the effector phase of the immune response revealed a significant role for IL-17, but not for IFN-γ or TNF, in vaccine-induced protection. In conclusion, although both T(h)1- and T(h)17-associated gene expression in the stomach correlate with vaccine-induced protection against H. pylori infection, our study indicates that mainly T(h)17 effector mechanisms are of critical importance to protection.
Collapse
|
26
|
Cytotoxic T cells in H. pylori-related gastric autoimmunity and gastric lymphoma. J Biomed Biotechnol 2010; 2010:104918. [PMID: 20617132 PMCID: PMC2896618 DOI: 10.1155/2010/104918] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 04/28/2010] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori infection is the major cause of gastroduodenal pathologies, but only a minority of infected patients develop gastric B-cell lymphoma, gastric autoimmunity, or other life threatening diseases, as gastric cancer or peptic ulcer. The type of host immune response against H. pylori, particularly the cytolytic effector functions of T cells, is crucial for the outcome of the infection. T cells are potentially able to kill a target via different mechanisms, such as perforins or Fas-Fas ligand interaction. In H. pylori-infected patients with gastric autoimmunity cytolytic T cells, that cross-recognize different epitopes of H. pylori proteins and H(+)K(+)-ATPase autoantigen, infiltrate the gastric mucosa and lead to gastric atrophy via long-lasting activation of Fas ligand-mediated appotosis and perforin-induced cytotoxicity. On the other hand, gastric T cells from MALT lymphoma exhibit defective perforin- and Fas-Fas ligand-mediated killing of B cells, with consequent abnormal help for B-cell proliferation, suggesting that deregulated and exhaustive H. pylori-induced T cell-dependent B-cell activation can support both the onset and the promotion of low-grade B-cell lymphoma.
Collapse
|
27
|
Abstract
A vaccination against Helicobacter pylori may represent both prophylactic and therapeutic approaches to the control of H. pylori infection. Different protective H. pylori-derived antigens, such as urease, vacuolating cytotoxin A, cytotoxin-associated antigen, neutrophil-activating protein and others can be produced at low cost in prokaryote expression systems and most of these antigens have already been administered to humans and shown to be safe. The recent development by Graham et al. of the model of H. pylori challenge in humans, the recent published clinical trials and the last insight generated in animal models of H. pylori infection regarding the immune mechanisms leading to vaccine-induced Helicobacter clearance will facilitate the evaluation of immunogenicity and efficacy of H. pylori vaccine candidates in Phase II and III clinical trials.
Collapse
Affiliation(s)
- Dominique Velin
- Service de Gastro-entérologie et d'Hépatologie, Centre Hospitalier Universitaire Vaudois and University of Lausanne, BH18-521, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland.
| | | |
Collapse
|
28
|
Kao JY, Zhang M, Miller MJ, Mills JC, Wang B, Liu M, Eaton KA, Zou W, Berndt BE, Cole TS, Takeuchi T, Owyang SY, Luther J. Helicobacter pylori immune escape is mediated by dendritic cell-induced Treg skewing and Th17 suppression in mice. Gastroenterology 2010; 138:1046-54. [PMID: 19931266 PMCID: PMC2831148 DOI: 10.1053/j.gastro.2009.11.043] [Citation(s) in RCA: 249] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 10/09/2009] [Accepted: 11/12/2009] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Helicobacter pylori infection increases gastric regulatory T cell (Treg) response, which may contribute to H pylori immune escape. We hypothesize that H pylori directs Treg skewing by way of dendritic cells (DCs) and thus inhibits interleukin-17(+) helper T cells (Th17) immunity. METHODS Two-photon microscopy was used to locate DCs in gastric lamina propria of mice. The induction of Th17 and Treg responses by bacteria-pulsed murine bone marrow-derived DCs was analyzed by cytokine production and stimulation of T-cell proliferation. The effect of VacA, CagA, transforming growth factor-beta (TGF-beta), and IL-10 on Th17/Treg balance was assessed. The in vivo significance of Tregs on the H pylori-specific Th17 response and H pylori density was determined by using anti-CD25 neutralizing antibodies to deplete Tregs in mice. RESULTS We showed that mucosal CD11c(+) DCs are located near the surface of normal gastric epithelium, and their number increased after H pylori infection. Study of the direct interaction of DCs with H pylori showed a Treg-skewed response. The Treg skewing was independent of H pylori VacA and CagA and dependent on TGF-beta and IL-10. In vivo Treg skewing by adoptive transfer of H pylori-pulsed DCs reduces the ratio of gastric IL-17/Foxp3 mRNA expressions. The depletion of CD25(+) Tregs results in early reduction of H pylori density, which is correlated with enhanced peripheral H pylori-specific Th17, but not Th1, response. CONCLUSIONS Overall, our study indicates that H pylori alters the DC-polarized Th17/Treg balance toward a Treg-biased response, which suppresses the effective induction of H pylori-specific Th17 immunity.
Collapse
Affiliation(s)
- John Y. Kao
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Min Zhang
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Mark J. Miller
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri 63110
| | - Jason C. Mills
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri 63110
| | - Baomei Wang
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri 63110
| | - Maochang Liu
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Kathyn A. Eaton
- Department of Microbiology and Immunology, University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Weiping Zou
- Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Bradford E. Berndt
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Tyler S. Cole
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Tomomi Takeuchi
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Stephanie Y. Owyang
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| | - Jay Luther
- Department of Internal Medicine (Division of Gastroenterology), University of Michigan Health System, Ann Arbor, Michigan 48109
| |
Collapse
|
29
|
Adjuvant effects for oral immunization provided by recombinant Lactobacillus casei secreting biologically active murine interleukin-1{beta}. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 17:43-8. [PMID: 19923575 DOI: 10.1128/cvi.00337-09] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Vaccine delivery systems using lactic acid bacteria are under development, but their efficiency is insufficient. Autologous cytokines, such as interleukin-1beta (IL-1beta), are potential adjuvants for mucosal vaccines and can be provided by recombinant lactic acid bacteria. The aim of this study was the construction and evaluation of recombinant Lactobacillus casei producing IL-1beta as an adjuvant delivery agent. The recombinant strain was constructed using an expression/secretion vector plasmid, including a mature IL-1beta gene from mouse. The biological activity of the cytokine was confirmed by IL-8 production from Caco-2 cells. In response to the recombinant L. casei secreting IL-1beta, expression of IL-6 was detected in vivo using a ligated-intestinal-loop assay. The release of IL-6 from Peyer's patch cells was also detected in vitro. Intragastric immunization with heat-killed Salmonella enterica serovar Enteritidis (SE) in combination with IL-1beta-secreting lactobacilli resulted in relatively high SE-specific antibody production. In this study, it was demonstrated that recombinant L. casei secreting bioactive murine IL-1beta provided adjuvant effects for intragastric immunization.
Collapse
|
30
|
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
|
31
|
Del Giudice G, Malfertheiner P, Rappuoli R. Development of vaccines against Helicobacter pylori. Expert Rev Vaccines 2009; 8:1037-49. [PMID: 19627186 DOI: 10.1586/erv.09.62] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori is a Gram-negative, microaerophilic bacterium adapted to survive in the stomach of humans where it can cause peptide ulcers and gastric cancer. Although effective antibiotic treatment exists, there is a consensus that vaccines are necessary to limit the severity of this infection. Great progress has been made since its discovery 25 years ago in understanding the virulence factors and several aspects of the pathogenesis of the H. pylori gastric diseases. Several key bacterial factors have been identified: urease, vacuolating cytotoxin, cytotoxin-associated antigen, the pathogenicity island, neutrophil-activating protein, and among others. These proteins, in their native or recombinant forms, have been shown to confer protection against infectious challenge with H. pylori in experimental animal models. It is not known, however, through which effector mechanisms this protection is achieved. Nevertheless, a number of clinical trials in healthy volunteers have been conducted using urease given orally as a soluble protein or expressed in bacterial vectors with limited results. Recently, a mixture of H. pylori antigens was reported to be highly immunogenic in H. pylori-negative volunteers following intramuscular administration of the vaccine with aluminium hydroxide as an adjuvant. These data show that vaccination against this pathogen is feasible. More research is required to understand the immunological mechanisms underlying immune-mediate protection.
Collapse
|
32
|
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: 140] [Impact Index Per Article: 8.8] [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
|
33
|
Velin D, Favre L, Bernasconi E, Bachmann D, Pythoud C, Saiji E, Bouzourene H, Michetti P. Interleukin-17 is a critical mediator of vaccine-induced reduction of Helicobacter infection in the mouse model. Gastroenterology 2009; 136:2237-2246.e1. [PMID: 19272385 DOI: 10.1053/j.gastro.2009.02.077] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Revised: 01/21/2009] [Accepted: 02/20/2009] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Despite the proven ability of immunization to reduce Helicobacter infection in mouse models, the precise mechanism of protection has remained elusive. This study explores the possibility that interleukin (IL)-17 plays a role in the reduction of Helicobacter infection following vaccination of wild-type animals or in spontaneous reduction of bacterial infection in IL-10-deficient mice. METHODS In mice, reducing Helicobacter infection, the levels and source of IL-17 were determined and the role of IL-17 in reduction of Helicobacter infection was probed by neutralizing antibodies. RESULTS Gastric IL-17 levels were strongly increased in mice mucosally immunized with urease plus cholera toxin and challenged with Helicobacter felis as compared with controls (654 +/- 455 and 34 +/- 84 relative units for IL-17 messenger RNA expression [P < .01] and 6.9 +/- 8.4 and 0.02 +/- 0.04 pg for IL-17 protein concentration [P < .01], respectively). Flow cytometry analysis showed that a peak of CD4(+)IL-17(+) T cells infiltrating the gastric mucosa occurred in immunized mice in contrast to control mice (4.7% +/- 0.3% and 1.4% +/- 0.3% [P < .01], respectively). Gastric mucosa-infiltrating CD4(+)IL-17(+) T cells were also observed in IL-10-deficient mice that spontaneously reduced H felis infection (4.3% +/- 2.3% and 2% +/- 0.6% [P < .01], for infected and noninfected IL-10-deficient mice, respectively). In wild-type immunized mice, intraperitoneal injection of anti-IL-17 antibodies significantly inhibited inflammation and the reduction of Helicobacter infection in comparison with control antibodies (1 of 12 mice vs 9 of 12 mice reduced Helicobacter infection [P < .01], respectively). CONCLUSIONS IL-17 plays a critical role in the immunization-induced reduction of Helicobacter infection from the gastric mucosa.
Collapse
Affiliation(s)
- Dominique Velin
- Division of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Chaouche-Drider N, Kaparakis M, Karrar A, Fernandez MI, Carneiro LAM, Viala J, Boneca IG, Moran AP, Philpott DJ, Ferrero RL. A commensal Helicobacter sp. of the rodent intestinal flora activates TLR2 and NOD1 responses in epithelial cells. PLoS One 2009; 4:e5396. [PMID: 19401779 PMCID: PMC2671595 DOI: 10.1371/journal.pone.0005396] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Accepted: 04/02/2009] [Indexed: 12/30/2022] Open
Abstract
Helicobacter spp. represent a proportionately small but significant component of the normal intestinal microflora of animal hosts. Several of these intestinal Helicobacter spp. are known to induce colitis in mouse models, yet the mechanisms by which these bacteria induce intestinal inflammation are poorly understood. To address this question, we performed in vitro co-culture experiments with mouse and human epithelial cell lines stimulated with a selection of Helicobacter spp., including known pathogenic species as well as ones for which the pathogenic potential is less clear. Strikingly, a member of the normal microflora of rodents, Helicobacter muridarum, was found to be a particularly strong inducer of CXC chemokine (Cxcl1/KC, Cxcl2/MIP-2) responses in a murine intestinal epithelial cell line. Time-course studies revealed a biphasic pattern of chemokine responses in these cells, with H. muridarum lipopolysaccharide (LPS) mediating early (24–48 h) responses and live bacteria seeming to provoke later (48–72 h) responses. H. muridarum LPS per se was shown to induce CXC chemokine production in HEK293 cells stably expressing Toll-like receptor 2 (TLR2), but not in those expressing TLR4. In contrast, live H. muridarum bacteria were able to induce NF-κB reporter activity and CXC chemokine responses in TLR2–deficient HEK293 and in AGS epithelial cells. These responses were attenuated by transient transfection with a dominant negative construct to NOD1, and by stable expression of NOD1 siRNA, respectively. Thus, the data suggest that both TLR2 and NOD1 may be involved in innate immune sensing of H. muridarum by epithelial cells. This work identifies H. muridarum as a commensal bacterium with pathogenic potential and underscores the potential roles of ill-defined members of the normal flora in the initiation of inflammation in animal hosts. We suggest that H. muridarum may act as a confounding factor in colitis model studies in rodents.
Collapse
Affiliation(s)
| | - Maria Kaparakis
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Abdulgader Karrar
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Maria-Isabel Fernandez
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, Paris, France
- INSERM U389, Paris, France
| | - Letitia A. M. Carneiro
- Institut Pasteur, Groupe d'Immunité Innée et Signalisation, Paris, France
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jérôme Viala
- Institut Pasteur, Groupe d'Immunité Innée et Signalisation, Paris, France
| | - Ivo Gomperts Boneca
- Institut Pasteur, Unité de Pathogénie Bactérienne des Muqueuses, Paris, France
- Institut Pasteur, Groupe de Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, Groupe Avenir, Paris, France
| | - Anthony P. Moran
- Department of Microbiology, National University of Ireland, Galway, Ireland
| | - Dana J. Philpott
- Institut Pasteur, Groupe d'Immunité Innée et Signalisation, Paris, France
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Richard L. Ferrero
- Institut Pasteur, Unité de Pathogénie Bactérienne des Muqueuses, Paris, France
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
35
|
DeLyria ES, Redline RW, Blanchard TG. Vaccination of mice against H pylori induces a strong Th-17 response and immunity that is neutrophil dependent. Gastroenterology 2009; 136:247-56. [PMID: 18948106 PMCID: PMC4960660 DOI: 10.1053/j.gastro.2008.09.017] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 08/07/2008] [Accepted: 09/11/2008] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Vaccine efficacy against gastric Helicobacter pylori infection has been shown in mice, but little is known about the mechanisms of bacterial clearance. Our aim was to investigate a possible T-cell/neutrophil pathway of vaccine-induced protection. METHODS Nonimmune and immunized mice were compared for their response to H pylori challenge. T-cell responses were assessed by recall assays. Interleukin (IL)-17-induced chemokine production was evaluated by cytokine enzyme-linked immunosorbent assay. In a kinetic study, biopsy specimens were collected at multiple time points postchallenge and assessed for bacterial load and inflammation. Relative levels of T cells, IL-17, interferon gamma, MIP-2, KC, and LIX were determined by quantitative polymerase chain reaction. The role of neutrophils was evaluated by antibody-mediated depletion of neutrophils following challenge. RESULTS Immunization induced strong interferon gamma- and IL-17-producing T-cell responses, and IL-17 was capable of inducing significant amounts of KC and MIP-2 from dendritic cells, macrophages, fibroblasts, and gastric epithelial cells. Challenge of immunized mice induced significantly greater gastritis than that of infected mice, preceding significantly lower bacterial loads by day 7. In immune mice, T-cell recruitment to the gastric mucosa correlated with a continuous rise in IL-17 and interferon gamma levels, followed by KC, MIP-2, and LIX production and the recruitment of significant numbers of neutrophils by day 5. Antibody-mediated depletion of neutrophils abrogated vaccine efficacy. CONCLUSIONS Vaccination of mice against H pylori results in a significant Th-17 cell recall response associated with increases in chemokines that attract neutrophils to the stomach, which are important for eradication of H pylori.
Collapse
Affiliation(s)
- Elizabeth S. DeLyria
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Raymond W. Redline
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Thomas G. Blanchard
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland,Corresponding author: Thomas Blanchard, Ph.D., Department of Pediatrics, University of Maryland School of Medicine, Bressler Research Building, 13-043, 655 West Baltimore Street, Baltimore, MD 21201, Voice: (410) 706-1772, Fax: (410) 328-1072,
| |
Collapse
|
36
|
Zhang M, Berndt BE, Eaton KA, Rathinavelu S, Pierzchala A, Kao JY. Helicobacter pylori-pulsed dendritic cells induce H. pylori-specific immunity in mice. Helicobacter 2008; 13:200-8. [PMID: 18466395 PMCID: PMC2871674 DOI: 10.1111/j.1523-5378.2008.00606.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The growing concern over the emergence of antibiotic-resistant Helicobacter pylori infection is propelling the development of an efficacious vaccine to control this highly adaptive organism. AIM We studied the use of a dendritic cell (DC)-based vaccine against H. pylori infection in mice. METHODS The cellular immune responses to murine bone marrow-derived DCs pulsed with phosphate-buffered saline (PBS-DC) or live H. pylori SS1 (HP-DC) were assessed in vitro and in vivo. The protective immunity against H. pylori SS1 oral challenge was compared between HP-DC or PBS-DC immunized mice. The effect of regulatory T-cell (Treg) depletion by anti-CD25 antibody on HP-DC vaccine efficacy was also evaluated. RESULTS HP-DC induced a Th1-dominant response in vitro. In vivo, HP-DC immunized mice were characterized by a mixed Th1/Th2 peripheral immune response. However, in the stomach, HP-DC immunized mice expressed a higher level of IFN-gamma compared to PBS-DC immunized mice; no difference was found for interleukin-5 expressions in the stomach. A lower bacterial colonization post-H. pylori challenge was observed in HP-DC immunized mice compared to PBS-DC immunized mice with no significant difference in gastritis severity. H. pylori-specific Th1 response and protective immunity were further enhanced in vivo by depletion of Treg with anti-CD25 antibody. CONCLUSION DC-based anti-H. pylori vaccine induced H. pylori-specific helper T-cell responses capable of limiting bacterial colonization. Our data support the critical role of effector cellular immune response in the development of H. pylori vaccine.
Collapse
Affiliation(s)
- Min Zhang
- Department of Internal Medicine (Gastroenterology Division), University of Michigan, Ann Arbor, Michigan 48109
| | - Bradford E. Berndt
- Department of Internal Medicine (Gastroenterology Division), University of Michigan, Ann Arbor, Michigan 48109
| | - Kathryn A. Eaton
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan 48109
| | - Sivaprakash Rathinavelu
- Department of Internal Medicine (Gastroenterology Division), University of Michigan, Ann Arbor, Michigan 48109
| | - Anna Pierzchala
- Department of Internal Medicine (Gastroenterology Division), University of Michigan, Ann Arbor, Michigan 48109
| | - John Y. Kao
- Department of Internal Medicine (Gastroenterology Division), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
37
|
Wehrens A, Aebischer T, Meyer TF, Walduck AK. Leptin receptor signaling is required for vaccine-induced protection against Helicobacter pylori. Helicobacter 2008; 13:94-102. [PMID: 18321299 DOI: 10.1111/j.1523-5378.2008.00591.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND A vaccine against Helicobacter pylori would be a desirable alternative to antibiotic therapy. Vaccination has been shown to be effective in animal models but the mechanism of protection is poorly understood. Previous studies investigating the gene expression in stomachs of vaccinated mice showed changes in adipokine expression correlated to a protective response. In this study, we investigate a well-characterized adipokine-leptin, and reveal an important role for leptin receptor signaling in vaccine-induced protection. MATERIALS AND METHODS Leptin receptor signaling-deficient (C57BL/Ks Lepr(db)), wild-type C57BL/Ks m littermates and C57BL/6 mice were vaccinated, and then challenged with H. pylori. Levels of bacterial colonization, antibody levels, and gastric infiltrates were compared. The local gene expression pattern in the stomach of leptin receptor signaling-deficient and wild-type mice was also compared using microarrays. RESULTS Interestingly, while vaccinated wild-type lean C57BL/6 and C57BL/Ks m mice were able to significantly reduce colonization compared to controls, vaccinated obese C57BL/Ks Lepr(db) were not. All mice responded to vaccination, i.e. developed infiltrates predominantly of T lymphocytes in the gastric mucosa, and made H. pylori-specific antibodies. A comparison of expression profiles in protected C57BL/6 and nonprotected C57BL/Ks Lepr(db) mice revealed a subset of inflammation-related genes that were more strongly expressed in nonprotected mice. CONCLUSIONS Our data suggest that functional leptin receptor signaling is required for mediating an effective protective response against H. pylori.
Collapse
Affiliation(s)
- Andrea Wehrens
- Department of Molecular Biology, Max-Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | | | | | | |
Collapse
|
38
|
Taylor JM, Ziman ME, Canfield DR, Vajdy M, Solnick JV. Effects of a Th1- versus a Th2-biased immune response in protection against Helicobacter pylori challenge in mice. Microb Pathog 2008; 44:20-7. [PMID: 17683897 PMCID: PMC2234601 DOI: 10.1016/j.micpath.2007.06.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2007] [Accepted: 06/13/2007] [Indexed: 12/16/2022]
Abstract
The roles that T helper type 1 (Th1) and T helper type 2 (Th2) Helicobacter pylori-specific immune responses play in protection from H. pylori challenge are poorly understood. It is expected that Th2 immune responses are required for protection against extracellular bacteria, such as H. pylori. However, recent studies have suggested that Th1 immunity is required for protection. The mechanisms by which this might occur are unknown. Our goal in this study was to more clearly define the effects of a Th1- versus a Th2-promoting H. pylori vaccine on immunity and protection. Therefore, we tested a Th1 vaccine consisting of an H. pylori sonicate and CpG oligonucleotides (CpG) and a Th2 vaccine consisting of a lipopolysaccharide (LPS)-depleted H. pylori sonicate combined with cholera toxin (CT). We demonstrate that although the Th2-promoting vaccine induced stronger systemic and local immune responses, only the Th1-promoting vaccine was protective.
Collapse
Affiliation(s)
- Jennifer M. Taylor
- Center for Comparative Medicine, University of California, Davis CA 95616
| | - Melanie E. Ziman
- Center for Comparative Medicine, University of California, Davis CA 95616
| | - Don R. Canfield
- Center for Comparative Medicine, University of California, Davis CA 95616
- California National Primate Research Center, University of California, Davis CA 95616
| | - Michael Vajdy
- Departments of Internal Medicine and Medical Microbiology and Immunology, University of California, Davis CA 95616
| | - Jay V. Solnick
- Center for Comparative Medicine, University of California, Davis CA 95616
- Departments of Internal Medicine and Medical Microbiology and Immunology, University of California, Davis CA 95616
| |
Collapse
|
39
|
Abstract
Nuclear factor kappaB (NF-kappaB) plays a key regulatory role in host cell responses to Helicobacter pylori infection in humans. Although mice are routinely used as a model to study H. pylori pathogenesis, the role of NF-kappaB in murine cell responses to helicobacters has not been studied in detail. We thus investigated the abilities of different Helicobacter isolates to induce NF-kappaB-dependent responses in murine gastric epithelial cells (GECs) and in transgenic mice harboring an NF-kappaB-responsive lacZ reporter gene. H. pylori and Helicobacter felis strains up-regulated the synthesis in mouse GECs of the NF-kappaB-dependent chemokines KC (CXCL1) and MIP-2 (CXCL2). These responses were cag pathogenicity island (cagPAI) independent and could be abolished by pretreatment with a pharmacological inhibitor of NF-kappaB. Consistent with the in vitro data, experimental Helicobacter infection of transgenic mice resulted in increased numbers of GECs with nuclear beta-galactosidase activity, which is indicative of specific NF-kappaB activation. The numbers of beta-galactosidase-positive cells in mice were significantly increased at day 1 postinoculation with wild-type H. pylori strains harboring or not harboring a functional cagPAI, compared to naive animals (P = 0.007 and P = 0.04, respectively). Strikingly, however, no differences were observed in the levels of gastric NF-kappaB activation at day 1 postinoculation with H. felis or at day 30 or 135 postinoculation with H. pylori. This work demonstrates for the first time the induction of NF-kappaB activation within gastric mucosal cells during acute H. pylori infection. Furthermore, the data suggest that helicobacters may be able to regulate NF-kappaB signaling during chronic infection.
Collapse
|
40
|
Takemoto K, Ogino K, Shibamori M, Gondo T, Hitomi Y, Takigawa T, Wang DH, Takaki J, Ichimura H, Fujikura Y, Ishiyama H. Transiently, paralleled upregulation of arginase and nitric oxide synthase and the effect of both enzymes on the pathology of asthma. Am J Physiol Lung Cell Mol Physiol 2007; 293:L1419-26. [PMID: 17890324 DOI: 10.1152/ajplung.00418.2006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Changes in the expression of arginase and their association with nitrosative stress were investigated using an asthmatic model previously established in NC/Nga mice with mite extract. Mite crude extract (100 microg/day) from Dermatophagoides farinae was administered intranasally for 5 consecutive days (day 0-4), and a single challenge was performed on day 11. On day 12, upregulation of the mRNA expression of inducible types of nitric oxide synthase (iNOS) and increases in immunohistochemical staining for iNOS and nitrotyrosine were observed. However, the level of nitrite + nitrate was unchanged. An increase in enzymatic activity, upregulation of mRNA expression, and immunostaining for arginase I was detected in the lung tissue and serum. Moreover, increases in both arginase I and II were revealed by immunoblotting. Goblet cell hyperplasia in bronchial epithelial cells and increasing collagen synthesis around the bronchus were also observed. These results suggested that an increase in arginase may lead to decreased availability of arginine for nitric oxide synthase and may contribute to the remodeling of the lung.
Collapse
Affiliation(s)
- Kei Takemoto
- Shin Nippon Biomedical Laboratories, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Wilson KT, Crabtree JE. Immunology of Helicobacter pylori: insights into the failure of the immune response and perspectives on vaccine studies. Gastroenterology 2007; 133:288-308. [PMID: 17631150 DOI: 10.1053/j.gastro.2007.05.008] [Citation(s) in RCA: 187] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Accepted: 05/02/2007] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori infects the stomach of half of the human population worldwide and causes chronic active gastritis, which can lead to peptic ulcer disease, gastric adenocarcinoma, and mucosa-associated lymphoid tissue lymphoma. The host immune response to the infection is ineffective, because the bacterium persists and the inflammation continues for decades. Bacterial activation of epithelial cells, dendritic cells, monocytes, macrophages, and neutrophils leads to a T helper cell 1 type of adaptive response, but this remains inadequate. The host inflammatory response has a key functional role in disrupting acid homeostasis, which impacts directly on the colonization patterns of H pylori and thus the extent of gastritis. Many potential mechanisms for the failure of the host response have been postulated, and these include apoptosis of epithelial cells and macrophages, inadequate effector functions of macrophages and dendritic cells, VacA inhibition of T-cell function, and suppressive effects of regulatory T cells. Because of the extent of the disease burden, many strategies for prophylactic or therapeutic vaccines have been investigated. The goal of enhancing the host's ability to generate protective immunity has met with some success in animal models, but the efficacy of potential vaccines in humans remains to be demonstrated. Aspects of H pylori immunopathogenesis are reviewed and perspectives on the failure of the host immune response are discussed. Understanding the mechanisms of immune evasion could lead to new opportunities for enhancing eradication and prevention of infection and associated disease.
Collapse
Affiliation(s)
- Keith T Wilson
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0252, USA
| | | |
Collapse
|
42
|
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
|
43
|
Algood HMS, Cover TL. Helicobacter pylori persistence: an overview of interactions between H. pylori and host immune defenses. Clin Microbiol Rev 2006; 19:597-613. [PMID: 17041136 PMCID: PMC1592695 DOI: 10.1128/cmr.00006-06] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori is a gram-negative bacterium that persistently colonizes more than half of the global human population. In order to successfully colonize the human stomach, H. pylori must initially overcome multiple innate host defenses. Remarkably, H. pylori can persistently colonize the stomach for decades or an entire lifetime despite development of an acquired immune response. This review focuses on the immune response to H. pylori and the mechanisms by which H. pylori resists immune clearance. Three main sections of the review are devoted to (i) analysis of the immune response to H. pylori in humans, (ii) analysis of interactions of H. pylori with host immune defenses in animal models, and (iii) interactions of H. pylori with immune cells in vitro. The topics addressed in this review are important for understanding how H. pylori resists immune clearance and also are relevant for understanding the pathogenesis of diseases caused by H. pylori (peptic ulcer disease, gastric adenocarcinoma, and gastric lymphoma).
Collapse
Affiliation(s)
- Holly M Scott Algood
- Division of Infectious Diseases, A2200 Medical Center North, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|
44
|
|
45
|
Kao JY, Rathinavelu S, Eaton KA, Bai L, Zavros Y, Takami M, Pierzchala A, Merchant JL. Helicobacter pylori-secreted factors inhibit dendritic cell IL-12 secretion: a mechanism of ineffective host defense. Am J Physiol Gastrointest Liver Physiol 2006; 291:G73-81. [PMID: 16469828 DOI: 10.1152/ajpgi.00139.2005] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Helicobacter pylori evades host immune defenses and causes chronic gastritis. Immunity against intestinal pathogens is largely mediated by dendritic cells, yet the role of dendritic cells in acute H. pylori infection is largely unknown. We observed the recruitment of dendritic cells to the gastric mucosa of H. pylori-infected mice. Bone marrow-derived dendritic cells from mice responded to live H. pylori by upregulating the expression of proinflammatory cytokine mRNA (i.e., IL-1alpha, IL-1beta, and IL-6). The supernatant from dendritic cells stimulated with H. pylori for 18 h contained twofold higher levels of IL-12p70 than IL-10 and induced the proliferation of syngeneic splenocytes and type 1 T helper cell cytokine release (IFN-gamma and TNF-alpha). These responses were significantly lower compared with those induced by Acinetobacter lwoffi, another gastritis-causing pathogen more susceptible to host defenses. Analysis of whole H. pylori sonicate revealed the presence of a heat-stable factor secreted from H. pylori that specifically inhibited IL-12 but not IL-10 release from dendritic cells activated by A. lwoffi. Our findings suggest that dendritic cells participate in the host immune response against H. pylori and that their suppression by H. pylori may explain why infected hosts fail to prevent bacterial colonization.
Collapse
Affiliation(s)
- John Y Kao
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109-0682, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Anderson KM, Czinn SJ, Redline RW, Blanchard TG. Induction of CTLA-4-mediated anergy contributes to persistent colonization in the murine model of gastric Helicobacter pylori infection. THE JOURNAL OF IMMUNOLOGY 2006; 176:5306-13. [PMID: 16621997 DOI: 10.4049/jimmunol.176.9.5306] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Helicobacter pylori infection induces gastric inflammation but the host fails to generate protective immunity. Therefore, we evaluated the immunologic mechanisms that contribute to the failure of the T cells to promote active immunity to H. pylori in the mouse model of H. pylori infection. Spleen cells from infected C57BL/6 mice underwent significantly less proliferation and cytokine production than cells from immune mice upon in vitro stimulation with H. pylori lysate. Similar results were observed when stimulating with Ag-pulsed macrophages demonstrating that hyporesponsiveness was not due to a direct effect of H. pylori virulence factors on the T cells. Ag-specific hyporesponsiveness could be reversed by the addition of high-dose IL-2 but not by removal of CD4(+)CD25(+) T cells, indicating that hyporesponsiveness was due to anergy and not due to active suppression. Cells from infected mice lacked significant suppressor activity as shown by the failure to reduce the recall response of cells from immune mice in coculture at physiologic ratios. Direct blockade of CTLA-4 using anti-CTLA-4 Fabs or indirect blockade using CTLA-4 Ig plus anti-CD28 Ab resulted in significantly increased T cell activation in vitro. The importance of CTLA-4 in establishing anergy was confirmed in an in vivo model of H. pylori infection in which mice that received anti-CTLA-4 Fabs responded to H. pylori challenge with significantly greater inflammation and significantly reduced bacterial load. These results suggest that CTLA-4 engagement induces and maintains functional inactivation of H. pylori-specific T cells during H. pylori infection resulting in a reduced immune response.
Collapse
Affiliation(s)
- Kathleen M Anderson
- Department of Pathology, School of Medicine, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
47
|
Hatzifoti C, Roussel Y, Harris AG, Wren BW, Morrow JW, Bajaj-Elliott M. Mucosal immunization with a urease B DNA vaccine induces innate and cellular immune responses against Helicobacter pylori. Helicobacter 2006; 11:113-22. [PMID: 16579841 DOI: 10.1111/j.1523-5378.2006.00385.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Helicobacter pylori is recognized as a major risk factor for recurrent gastroduodenal inflammatory diseases and gastric adenocarcinoma. The high prevalence of H. pylori infection worldwide, the risks of side-effects from antibiotic therapy, and increasing resistance to antibiotics are the main primers for the development of improved H. pylori vaccines. The antigenic potential of its urease enzyme, a critical virulence factor required for colonization of the gastric mucosa, has been demonstrated in animal and human studies. An important but controversial issue in H. pylori vaccine studies is the type of immune response required to control infection. A new approach in H. pylori vaccinology is the administration of DNA vaccines, which has included heat-shock protein and catalase DNA vaccines. MATERIALS AND METHODS The H. pylori urease subunit B construct or vector alone was administered to mice via the intranasal route. Spleens and stomachs were examined on day 0 and weeks 3, 6, and 12 after immunization. Proliferation of spleen cells was assessed using the carboxyfluorescein diacetate succinimidyl ester-based flow cytometry assay and cytokine secretion from cultured spleen cells was detected by ELISA, after stimulation with the urease subunit B recombinant antigen. Total RNA was isolated from stomach and spleen tissue and the expression of beta-defensin and cytokine genes was monitored by reverse transcription followed by polymerase chain reaction (RT-PCR). Immunized mice were challenged with H. pylori and bacterial DNA quantified by TaqMan PCR. RESULTS The urease B subunit DNA vaccine increased INF-gamma secretion and splenocyte proliferation without inducing adverse effects in the spleen. Increase in gastric beta-defensin 1 and marked induction in local IL-10 : IFN-gamma ratio up to 12 weeks post-immunization suggest a potential role for local innate immune responses in protection at the site of infection. Although significant bacterial reduction in the stomachs of urease B subunit DNA-immunized mice was observed, intermediate reduction was also noted in the vector group. Increased defensin expression and adjuvant effects of the cytosine preceding guanosine motifs may contribute to this phenomenon. Our data confirm that cytosine preceding guanosine motifs, even without coadministration with antigen, can reduce extracellular bacterial load. CONCLUSIONS In this study, a DNA construct encoding the urease B subunit was assessed for its immune profile and its ability to reduce bacterial colonization in the murine stomach. Our studies suggest that local innate immune responses may play a greater role than previously supposed in limiting H. pylori colonization in the gastric mucosa.
Collapse
Affiliation(s)
- Caterina Hatzifoti
- Department of Immunology, St. Bartholomew's and the Royal London School of Medicine and Dentistry, London, UK.
| | | | | | | | | | | |
Collapse
|
48
|
Akhiani AA, Stensson A, Schön K, Lycke N. The nontoxic CTA1-DD adjuvant enhances protective immunity against Helicobacter pylori infection following mucosal immunization. Scand J Immunol 2006; 63:97-105. [PMID: 16476008 DOI: 10.1111/j.1365-3083.2005.01713.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Safe and efficacious adjuvants are much needed to facilitate the development of mucosal vaccines. Here, we have asked whether our nontoxic vaccine adjuvant, CTA1-DD, can enhance protective immunity against Helicobacter pylori infection. Intranasal immunizations with H. pylori lysate together with CTA1-DD-adjuvant induced significant protection in C57Bl/6 mice, almost as strong as similar immunizations using cholera toxin (CT)-adjuvant. Protection remained strong even at 8 weeks postchallenge and the bacterial colonization was reduced by 20-fold compared to lysate-immunized controls. Although CTA1-DD was designed to bind to B cells, microMT mice developed significant, but lower, level of protection following immunization. Intranasal immunizations with CT adjuvant in C57Bl/6 mice resulted in the development of severe postimmunization gastritis at 2 and 8 weeks postchallenge, whereas the degree of gastritis was substantially lower in the CTA1-DD-immunized mice. Protection induced by both CTA1-DD- and CT adjuvant was associated with a strong local infiltration of CD4(+) T cells in the gastric mucosa, and recall responses to specific Ag elicited substantial IFN-gamma production, indicating Th1-dominance. These findings clearly demonstrate that CTA1-DD adjuvant is a promising candidate to be further exploited in the development of a mucosal vaccine against H. pylori infection.
Collapse
Affiliation(s)
- A A Akhiani
- Department of Clinical Immunology, Göteborg University, Göteborg, Sweden
| | | | | | | |
Collapse
|
49
|
Hansson M, Lundgren A, Elgbratt K, Quiding-Järbrink M, Svennerholm AM, Johansson EL. Dendritic cells express CCR7 and migrate in response to CCL19 (MIP-3β) after exposure to Helicobacter pylori. Microbes Infect 2006; 8:841-50. [PMID: 16500130 DOI: 10.1016/j.micinf.2005.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Revised: 09/02/2005] [Accepted: 10/10/2005] [Indexed: 12/28/2022]
Abstract
Helicobacter pylori infection induces chronic inflammation in the gastric mucosa with a marked increase in the number of lymphoid follicles consisting of infiltrating B and T cells, neutrophils, dendritic cells (DC) and macrophages. It has been suggested that an accumulation of mature DC in the tissue, resulting from a failure of DC to migrate to lymph nodes, may contribute to this chronic inflammation. Migration of DC to lymph nodes is regulated by chemokine receptor CCR7, expressed on mature DC, and the CCR7 ligands CCL19 and CCL21. In this study we analysed the maturation, in vitro migration and cytokine production of human DC after stimulation with live H. pylori. For comparison, DC responses to non-pathogenic Escherichia coli bacteria were also evaluated. Stimulation with H. pylori induced maturation of DC, i.e. up-regulation of the chemokine receptors CCR7 and CXCR4 and the maturation markers HLA-DR, CD80 and CD86. The H. pylori-stimulated DC also induced CD4(+) T-cell proliferation. DC stimulated with H. pylori secreted significantly more interleukin (IL)-12 compared to DC stimulated with E. coli, while E. coli-stimulated DC secreted more IL-10. Despite low surface expression of CCR7 protein following stimulation with H. pylori compared to E. coli, the DC migrated equally well towards CCL19 after stimulation with both bacteria. Thus, we could not detect any failure in the migration of H. pylori stimulated DC in vitro that may contribute to chronic gastritis in vivo, and our results suggest that H. pylori induces maturation and migration of DC to lymph nodes where they promote T cell responses.
Collapse
Affiliation(s)
- Malin Hansson
- Department of Medical Microbiology and Immunology and Göteborg University Vaccine Research Institute (GUVAX), Göteborg University, Box 435, 405 30 Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
50
|
Bergman M, Del Prete G, van Kooyk Y, Appelmelk B. Helicobacter pylori phase variation, immune modulation and gastric autoimmunity. Nat Rev Microbiol 2006; 4:151-9. [PMID: 16415930 DOI: 10.1038/nrmicro1344] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Helicobacter pylori can be regarded as a model pathogen for studying persistent colonization of humans. Phase-variable expression of Lewis blood-group antigens by H. pylori allows this microorganism to modulate the host T-helper-1-cell versus T-helper-2-cell response. We describe a model in which interactions between host lectins and pathogen carbohydrates facilitate asymptomatic persistence of H. pylori. This delicate balance, favourable for both the pathogen and the host, could lead to gastric autoimmunity in genetically susceptible individuals.
Collapse
Affiliation(s)
- Mathijs Bergman
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Room L-253, 3015 GD Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|