1
|
Takeuchi C, Sato J, Yamashita S, Sasaki A, Akahane T, Aoki R, Yamamichi M, Liu YY, Ito M, Furuta T, Nakajima S, Sakaguchi Y, Takahashi Y, Tsuji Y, Niimi K, Tomida S, Fujishiro M, Yamamichi N, Ushijima T. Autoimmune gastritis induces aberrant DNA methylation reflecting its carcinogenic potential. J Gastroenterol 2022; 57:144-155. [PMID: 35034200 DOI: 10.1007/s00535-021-01848-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Autoimmune gastritis (AIG) is a chronic inflammatory condition in gastric mucosa and is associated with increased cancer risk, though not as high as that by Helicobacter pylori (H. pylori)-associated gastritis (HPG). Although aberrant DNA methylation is induced by HPG and the level correlates with the risk of gastric cancer, DNA methylation induction by AIG is unknown. METHODS Gastric mucosa samples from the corpus were collected from 12 people with AIG without H. pylori infection, 10 people with HPG, and eight healthy volunteers. Genome-wide DNA methylation analysis was conducted using Infinium Methylation EPIC array. Gene expression was analyzed by quantitative RT-PCR. RESULTS The AIG samples had extensive aberrant DNA methylation but presented unique methylation profiles against the HPG samples after correction of leucocyte fractions. Comparison between the AIG and HPG samples showed that AIG induced methylation, but less than HPG, in overall CpG sites and also in promoter CpG islands. Promoter CpG islands of tumor-suppressor genes in the pathway of cell cycle, cell adhesion, p53, and WNT were highly methylated in the AIG samples, but more so in the HPG samples. The expression levels of IL1B and IL8, secreted by macrophage, were significantly lower in the AIG samples than in the HPG samples, suggesting that a difference in inflammatory response affected the degree and patterns of aberrant DNA methylation. CONCLUSIONS AIG induced aberrant DNA methylation in gastric mucosa. However, the degree of DNA methylation was less than that by HPG, which reflected carcinogenic risk.
Collapse
Affiliation(s)
- Chihiro Takeuchi
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Junichi Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Satoshi Yamashita
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Akiko Sasaki
- Department of Gastroenterology, Medicine Center, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Takemi Akahane
- Department of Gastroenterology, Nara Medical University, Nara, Japan
| | - Rika Aoki
- Tokushima Health Screening Center, Tokushima, Japan
| | - Mitsue Yamamichi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yu-Yu Liu
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Masayoshi Ito
- Department of Gastroenterology, Yotsuya Medical Cube, Tokyo, Japan
| | - Takahisa Furuta
- Center for Clinical Research, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Shigemi Nakajima
- Department of General Medicine, Consortium for Community Medicine, Japan Community Healthcare Organization Shiga Hospital, Shiga University of Medical Science, Shiga, Japan
| | - Yoshiki Sakaguchi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yu Takahashi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Tsuji
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keiko Niimi
- Center for Epidemiology and Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuta Tomida
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobutake Yamamichi
- Center for Epidemiology and Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshikazu Ushijima
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
2
|
Cen Q, Gao T, Ren Y, Lu X, Lei H. Immune evaluation of a Saccharomyces cerevisiae-based oral vaccine against Helicobacter pylori in mice. Helicobacter 2021; 26:e12772. [PMID: 33219579 DOI: 10.1111/hel.12772] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is a common human pathogenic bacterium that is associated with gastric diseases. The current leading clinical therapy is combination antibiotics, but this treatment has safety issues, especially the induction of drug resistance. Therefore, developing a safe and effective vaccine against H. pylori is one of the best alternatives. OBJECTIVE To develop Saccharomyces cerevisiae (S. cerevisiae)-based oral vaccines and then demonstrate the feasibility of this platform for preventing H. pylori infection in the absence of a mucosal adjuvant. MATERIALS AND METHODS Saccharomyces cerevisiae (S. cerevisiae)-based oral vaccines, including EBY100/pYD1-UreB and EBY100/pYD1-VacA, were generated and analyzed by Western blot, Immunofluorescence analysis, flow cytometric assay, and indirect enzyme-link immunosorbent assay (ELISA). Further, antibody responses induced by oral administration of EBY100/pYD1-UreB, EBY100/pYD1-VacA, or EBY100/pYD1-UreB + EBY100/pYD1-VacA were measured in a mouse model. Lastly, the vaccinated mice were infected with H. pylori SS1, and colonization in the stomach were evaluated. RESULTS Saccharomyces cerevisiae-based H. pylori oral vaccines were successfully constructed. Mice orally administered with EBY100/pYD1-UreB, EBY100/pYD1-VacA, or EBY100/pYD1-UreB + EBY100/pYD1-VacA exhibited a significant humoral immune response as well as a mucosal immune response. Importantly, S. cerevisiae-based oral vaccines could effectively reduce bacterial loads with statistical significance after H. pylori infection. CONCLUSIONS Our study shows that S. cerevisiae-based platforms can serve as an alternative approach for the future development of promising bacterial oral vaccine candidates.
Collapse
Affiliation(s)
- Qianhong Cen
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Tong Gao
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Yi Ren
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Xin Lu
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Han Lei
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
3
|
Natarajan V, Moar P, Kaur US, Venkatesh V, Kumar A, Chaturvedi R, Himanshu D, Tandon R. Helicobacter pylori Reactivates Human Immunodeficiency Virus-1 in Latently Infected Monocytes with Increased Expression of IL-1β and CXCL8. Curr Genomics 2020; 20:556-568. [PMID: 32581644 PMCID: PMC7290055 DOI: 10.2174/1389202921666191226091138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/08/2019] [Accepted: 12/08/2019] [Indexed: 12/13/2022] Open
Abstract
Background Helicobacter pylori are gram-negative bacteria, which colonize the human stomach. More than 50% of the world's population is infected by H. pylori. Based on the high prevalence of H. pylori, it is very likely that HIV and H. pylori infection may coexist. However, the molecular events that occur during HIV-H. pylori co-infection remain unclear. Latent HIV reservoirs are the major obstacle in HIV cure despite effective therapy. Here, we explored the effect of H. pylori stimulation on latently HIV-infected monocytic cell line U1. Methods High throughput RNA-Seq using Illumina platform was performed to analyse the change in transcriptome between unstimulated and H. pylori-stimulated latently HIV-infected U1 cells. Transcriptome analysis identified potential genes and pathways involved in the reversal of HIV latency using bioinformatic tools that were validated by real-time PCR. Results H. pylori stimulation increased the expression of HIV-1 Gag, both at transcription (p<0.001) and protein level. H. pylori stimulation also increased the expression of proinflammatory cytokines IL-1β, CXCL8 and CXCL10 (p<0.0001). Heat-killed H. pylori retained their ability to induce HIV transcription. RNA-Seq analysis revealed 197 significantly upregulated and 101 significantly downregulated genes in H. pylori-stimulated U1 cells. IL-1β and CXCL8 were found to be significantly upregulated using transcriptome analysis, which was consistent with real-time PCR data. Conclusion H. pylori reactivate HIV-1 in latently infected monocytes with the upregulation of IL-1β and CXCL8, which are prominent cytokines involved in the majority of inflammatory pathways. Our results warrant future in vivo studies elucidating the effect of H. pylori in HIV latency and pathogenesis.
Collapse
Affiliation(s)
- Vidhya Natarajan
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Preeti Moar
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Urvinder S Kaur
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Vimala Venkatesh
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Abhishek Kumar
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Rupesh Chaturvedi
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - D Himanshu
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Ravi Tandon
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| |
Collapse
|
4
|
Kato M, Toda A, Yamamoto‐Honda R, Arase Y, Sone H. Association between Helicobacter pylori infection, eradication and diabetes mellitus. J Diabetes Investig 2019; 10:1341-1346. [PMID: 30663265 PMCID: PMC6717903 DOI: 10.1111/jdi.13011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 12/17/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
AIMS/INTRODUCTION It is suspected that Helicobacter pylori is associated with extradigestive diseases including diabetes. So far, a number of studies have examined the association between H. pylori and diabetes, and the results were conflicting. The aim of the present study was to examine the association between H. pylori infection, eradication and diabetes. MATERIALS AND METHODS The present cross-sectional study was carried out using data from annual health checkups carried out at the Toranomon Hospital Health Management Center. The status of H. pylori infection, determined by serum antibodies and history of eradication, was categorized into three groups as "never," "current" and "past." The association between H. pylori infection and diabetes was examined using logistic regression. RESULTS Of 21,634 participants, 6,530 (30.2%) had a current or past history of H. pylori infection, and 1,184 (5.5%) were identified as having diabetes. Multivariate adjusted odds ratios for diabetes compared with the "never" group were 1.36 (95% confidence interval 1.10-1.67) for the "current" group and 0.92 (95% confidence interval 0.79-1.07) for the "past" group. The association between H. pylori infection and diabetes was also observed among participants without a history of eradication. CONCLUSIONS We found that current H. pylori infection was associated with an increased risk of diabetes, and the increased risk was not observed among participants after eradication. The results were concordant with the hypothesis that H. pylori infection increases the risk of diabetes. Further studies are necessary to validate the present results.
Collapse
Affiliation(s)
- Masayuki Kato
- Health Management CenterToranomon HospitalTokyoJapan
- Okinaka Memorial Institute for Medical ResearchToranomon HospitalTokyoJapan
| | - Akiko Toda
- Health Management CenterToranomon HospitalTokyoJapan
- Okinaka Memorial Institute for Medical ResearchToranomon HospitalTokyoJapan
| | - Ritsuko Yamamoto‐Honda
- Health Management CenterToranomon HospitalTokyoJapan
- Okinaka Memorial Institute for Medical ResearchToranomon HospitalTokyoJapan
| | - Yasuji Arase
- Health Management CenterToranomon HospitalTokyoJapan
- Okinaka Memorial Institute for Medical ResearchToranomon HospitalTokyoJapan
| | - Hirohito Sone
- Department of Internal Medicine (Metabolism)Faculty of MedicineNiigata UniversityNiigataJapan
| |
Collapse
|
5
|
The Relationship between Toll-like Receptors and Helicobacter pylori-Related Gastropathies: Still a Controversial Topic. J Immunol Res 2019; 2019:8197048. [PMID: 30863783 PMCID: PMC6378784 DOI: 10.1155/2019/8197048] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/02/2019] [Indexed: 12/13/2022] Open
Abstract
Innate immunity represents the first barrier against bacterial invasion. Toll-like receptors (TLRs) belong to the large family of pattern recognition receptors (PRRs), and their activation leads to the induction of inflammatory cytokines, chemokines, antigen-presenting molecules, and costimulatory molecules. Recent studies have focused on identifying the association between TLRs and Helicobacter pylori- (H. pylori-) related diseases. Therefore, this minireview focuses on assessing the role of these TLRs in the development of H. pylori-related gastropathies. Both TLR2 and TLR were found to be involved in H. pylori LPS recognition, with contradictory results most likely due to both the inability to obtain pure LPS in experimental studies and the heterogeneity of the bacterial LPS. In addition, TLR2 was found to be the most extensively expressed gene among all the TLRs in gastric tumors. High levels of TLR4 were also associated with a higher risk of gastric cancer. TLR5 was initially associated with the recognition of H. pylori flagellin, but it seems that this bacterium has developed mechanisms to escape this recognition representing an important factor involved in the persistence of this infection and subsequent carcinogenesis. TLR9, the only TLR with both anti- and proinflammatory roles, was involved in the recognition of H. pylori DNA. The dichotomous role of TLR9, promoting or suppressing the infection, depends on the gastric environment. Recently, TLR7 and TLR8 were shown to recognize purified H. pylori RNA, thereby inducing proinflammatory cytokines. TLR1 and TLR10 gene polymorphisms were associated with a higher risk for gastric cancer in H. pylori-infected individuals. Different gene polymorphisms of these TLRs were found to be associated with gastric cancer depending mostly on ethnicity. Further studies are required in order to develop preventive and therapeutic strategies against H. pylori infections based on the functions of TLRs.
Collapse
|
6
|
Maleki Kakelar H, Barzegari A, Dehghani J, Hanifian S, Saeedi N, Barar J, Omidi Y. Pathogenicity of Helicobacter pylori in cancer development and impacts of vaccination. Gastric Cancer 2019; 22:23-36. [PMID: 30145749 DOI: 10.1007/s10120-018-0867-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/14/2018] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori affect around 50% of the population worldwide. More importantly, the gastric infection induced by this bacterium is deemed to be associated with the progression of distal gastric carcinoma and gastric mucosal lymphoma in the human. H. pylori infection and its prevalent genotype significantly differ across various geographical regions. Based on numerous virulence factors, H. pylori can target different cellular proteins to modulate the variety of inflammatory responses and initiate numerous "hits" on the gastric mucosa. Such reactions lead to serious complications, including gastritis and peptic ulceration, gastric cancer and gastric mucosa-associated lymphoid structure lymphoma. Therefore, H. pylori have been considered as the type I carcinogen by the Global Firm for Research on Cancer. During the two past decades, different reports revealed that H. pylori possess oncogenic potentials in the gastric mucosa through a complicated interplay between the bacterial factors, various facets, and the environmental factors. Accordingly, numerous signaling pathways could be triggered in the development of gastrointestinal diseases (e.g., gastric cancer). Therefore, the main strategy for the treatment of gastric cancer is controlling the disease far before its onset using preventive/curative vaccination. Increasing the efficiency of vaccines may be achieved by new trials of vaccine modalities, which is used to optimize the cellular immunity. Taken all, H. pylori infection may impose severe complications, for resolving of which extensive researches are essential in terms of immune responses to H. pylori. We envision that H. pylori-mediated diseases can be controlled by advanced vaccines and immunotherapies.
Collapse
Affiliation(s)
- Hadi Maleki Kakelar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaber Dehghani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Hanifian
- Department of Food Science and Technology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Nazli Saeedi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran.
| |
Collapse
|
7
|
Morey P, Pfannkuch L, Pang E, Boccellato F, Sigal M, Imai-Matsushima A, Dyer V, Koch M, Mollenkopf HJ, Schlaermann P, Meyer TF. Helicobacter pylori Depletes Cholesterol in Gastric Glands to Prevent Interferon Gamma Signaling and Escape the Inflammatory Response. Gastroenterology 2018; 154:1391-1404.e9. [PMID: 29273450 DOI: 10.1053/j.gastro.2017.12.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 11/17/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Despite inducing an inflammatory response, Helicobacter pylori can persist in the gastric mucosa for decades. H pylori expression of cholesterol-α-glucosyltransferase (encoded by cgt) is required for gastric colonization and T-cell activation. We investigated how cgt affects gastric epithelial cells and the host immune response. METHODS MKN45 gastric epithelial cells, AGS cells, and human primary gastric epithelial cells (obtained from patients undergoing gastrectomy or sleeve resection or gastric antral organoids) were incubated with interferon gamma (IFNG) or interferon beta (IFNB) and exposed to H pylori, including cagPAI and cgt mutant strains. Some cells were incubated with methyl-β-cyclodextrin (to deplete cholesterol from membranes) or myriocin and zaragozic acid to prevent biosynthesis of sphingolipids and cholesterol and analyzed by immunoblot, immunofluorescence, and reverse transcription quantitative polymerase chain reaction analyses. We compared gene expression patterns among primary human gastric cells, uninfected or infected with H pylori P12 wt or P12Δcgt, using microarray analysis. Mice with disruption of the IFNG receptor 1 (Ifngr1-/- mice) and C57BL6 (control) mice were infected with PMSS1 (wild-type) or PMSS1Δcgt H pylori; gastric tissues were collected and analyzed by reverse transcription quantitative polymerase chain reaction or confocal microscopy. RESULTS In primary gastric cells and cell lines, infection with H pylori, but not cgt mutants, blocked IFNG-induced signaling via JAK and STAT. Cells infected with H pylori were depleted of cholesterol, which reduced IFNG signaling by disrupting lipid rafts, leading to reduced phosphorylation (activation) of JAK and STAT1. H pylori infection of cells also blocked signaling by IFNB, interleukin 6 (IL6), and IL22 and reduced activation of genes regulated by these signaling pathways, including cytokines that regulate T-cell function (MIG and IP10) and anti-microbial peptides such as human β-defensin 3 (hBD3). We found that this mechanism allows H pylori to persist in proximity to infected cells while inducing inflammation only in the neighboring, non-infected epithelium. Stomach tissues from mice infected with PMSS1 had increased levels of IFNG, but did not express higher levels of interferon-response genes. Expression of the IFNG-response gene IRF1 was substantially higher in PMSS1Δcgt-infected mice than PMSS1-infected mice. Ifngr1-/- mice were colonized by PMSS1 to a greater extent than control mice. CONCLUSIONS H pylori expression of cgt reduces cholesterol levels in infected gastric epithelial cells and thereby blocks IFNG signaling, allowing the bacteria to escape the host inflammatory response. These findings provide insight into the mechanisms by which H pylori might promote gastric carcinogenesis (persisting despite constant inflammation) and ineffectiveness of T-cell-based vaccines against H pylori.
Collapse
Affiliation(s)
- Pau Morey
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Lennart Pfannkuch
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Ervinna Pang
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Francesco Boccellato
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Michael Sigal
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Department of Hepatology and Gastroenterology, Charité University Medicine, Berlin, Germany
| | - Aki Imai-Matsushima
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Victoria Dyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Manuel Koch
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hans-Joachim Mollenkopf
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Philipp Schlaermann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany.
| |
Collapse
|
8
|
Keck J, Gupta R, Christenson LK, Arulanandam BP. MicroRNA mediated regulation of immunity against gram-negative bacteria. Int Rev Immunol 2017; 36:287-299. [PMID: 28800263 PMCID: PMC6904929 DOI: 10.1080/08830185.2017.1347649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Evidence over the last couple decades has comprehensively established that short, highly conserved, non-coding RNA species called microRNA (miRNA) exhibit the ability to regulate expression and function of host genes at the messenger RNA (mRNA) level. MicroRNAs play key regulatory roles in immune cell development, differentiation, and protective function. Intrinsic host immune response to invading pathogens rely on intricate orchestrated events in the development of innate and adaptive arms of immunity. We discuss the involvement of miRNAs in regulating these processes against gram negative pathogens in this review.
Collapse
Affiliation(s)
- Jonathon Keck
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| | - Lane K. Christenson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| |
Collapse
|
9
|
Wang F, Mao Z, Liu D, Yu J, Wang Y, Ye W, Lin D, Zhou N, Xie Y. Overexpression of Tim-3 reduces Helicobacter pylori-associated inflammation through TLR4/NFκB signaling in vitro. Mol Med Rep 2017; 15:3252-3258. [PMID: 28339054 DOI: 10.3892/mmr.2017.6346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 01/19/2017] [Indexed: 12/27/2022] Open
Abstract
The present study aimed to investigate the interaction between T-cell immunoglobulin and mucin-domain-containing molecule-3 (Tim-3) and Toll-like receptor 4 (TLR4)/nuclear factor κB (NF‑κB) signaling in Helicobacter pylori-infected RAW264.7 macrophage cells. RAW264.7 cells were co‑cultured with H. pylori SS1 at different bacteria/cell ratios, and subsequently the mRNA expression of Tim‑3, TLR4, and myeloid differentiation factor 88 (MyD88) was measured by reverse transcription-quantitative polymerase chain reaction (RT‑qPCR). Furthermore, the effect of Tim‑3 overexpression was examined by transfection of RAW264.7 with pLVX-IRES-ZsGreen-Tim-3 and co‑culturing with H. pylori. mRNA and protein expression levels were then analyzed for Tim‑3, TLR4, MyD88, and phosphorylated (p‑) NF‑κB by RT‑qPCR and western blot analysis respectively. The concentrations of pro‑inflammatory cytokines [tumor necrosis factor‑α (TNF‑α), interleukin 6 (IL-6), interferon‑γ (IFN‑γ) and interleukin 10 (IL‑10)] released in the culture supernatants were measured by ELISA. H. pylori stimulation resulted in a significant increase of Tim‑3, TLR4, and MyD88 mRNA expression in RAW264.7 cells. H. pylori stimulation upregulated Tim‑3 expression even in the Tim‑3‑overexpressing RAW264.7 cells compared with unstimulated cells. TLR4, MyD88, and pNF‑κB protein expression and pro‑inflammatory cytokines (TNF‑α, IL‑6, and IFN‑γ) release levels were increased in the control RAW264.7 cells following H. pylori infection, but not in the Tim-3-overexpressing RAW264.7 cells. By contrast, IL‑10 levels were decreased following H. pylori infection in both control and Tim‑3‑overexpressing RAW264.7 cells. Overexpression of Tim-3 reduced H. pylori-associated inflammation in RAW264.7 macrophages, by downregulating expression of proteins in the TLR4 pathway and release of pro‑inflammatory cytokines. These findings suggest that Tim‑3 serves a crucial role in the negative regulation of H. pylori-associated inflammation and may be a novel therapeutic target for H. pylori infection.
Collapse
Affiliation(s)
- Fucai Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Gastroenterology Institute of Jiangxi, Key Laboratory of Digestive Diseases of Jiangxi, Nanchang, Jiangxi 330006, P.R. China
| | - Zhirong Mao
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Gastroenterology Institute of Jiangxi, Key Laboratory of Digestive Diseases of Jiangxi, Nanchang, Jiangxi 330006, P.R. China
| | - Dongsheng Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Gastroenterology Institute of Jiangxi, Key Laboratory of Digestive Diseases of Jiangxi, Nanchang, Jiangxi 330006, P.R. China
| | - Jing Yu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Gastroenterology Institute of Jiangxi, Key Laboratory of Digestive Diseases of Jiangxi, Nanchang, Jiangxi 330006, P.R. China
| | - Youhua Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Gastroenterology Institute of Jiangxi, Key Laboratory of Digestive Diseases of Jiangxi, Nanchang, Jiangxi 330006, P.R. China
| | - Wen Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Gastroenterology Institute of Jiangxi, Key Laboratory of Digestive Diseases of Jiangxi, Nanchang, Jiangxi 330006, P.R. China
| | - Dongjia Lin
- Department of Immunology, Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Nanjin Zhou
- Institute of Immunology and Biological Therapy, Jiangxi Academy of Medical Sciences, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Gastroenterology Institute of Jiangxi, Key Laboratory of Digestive Diseases of Jiangxi, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
10
|
Mirzaei N, Poursina F, Moghim S, Rashidi N, Ghasemian Safaei H. The study of H. pylori putative candidate factors for single- and multi-component vaccine development. Crit Rev Microbiol 2017; 43:631-650. [PMID: 28581361 DOI: 10.1080/1040841x.2017.1291578] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori has grown to colonize inside the stomach of nearly half of the world's population, turning into the most prevalent infections in the universe. Medical care failures noticeably confirm the need for a vaccine to hinder or deal with H. pylori. This review is planned to discuss the most known factors as a vaccine candidate, including single (AhpC, BG, CagA, KatA, Fla, Hsp, HWC, Lpp, LPS, NAP, OMP, OMV, SOD, Tpx, Urease, VacA) and multi-component vaccines. Many promising results in the field of single and multivalent vaccine can be seen, but there is no satisfactory outcome and neither a prophylactic nor a therapeutic vaccine to treat or eradicate the infection in human has been acquired. Hence, selecting suitable antigen is an important factor as an appropriate adjuvant. Taken all together, the development of efficient anti-H. pylori vaccines relies on the fully understanding of the interactions between H. pylori and its host immune system. Therefore, more work should be done on epitope mapping, analysis of molecular structure, and determination of the antigen determinant region as well due to design a vaccine, preferably a multi-component vaccine to elicit specific CD4 T-cell responses that are required for H. pylori vaccine efficacy.
Collapse
Affiliation(s)
- Nasrin Mirzaei
- a Department of Microbiology , Tonekabon Branch, Islamic Azad University , Tonekabon , Iran
| | - Farkhondeh Poursina
- b Department of Microbiology , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Sharareh Moghim
- b Department of Microbiology , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Niloufar Rashidi
- c Department of Laboratory Sciences , Ahvaz University of Medical Sciences , Ahvaz , Iran
| | | |
Collapse
|
11
|
Surface expression of Helicobacter pylori HpaA adhesion antigen on Vibrio cholerae, enhanced by co-expressed enterotoxigenic Escherichia coli fimbrial antigens. Microb Pathog 2017; 105:177-184. [PMID: 28215587 DOI: 10.1016/j.micpath.2017.02.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 02/11/2017] [Accepted: 02/13/2017] [Indexed: 12/23/2022]
Abstract
Helicobacter pylori infection can cause peptic ulceration and is associated with gastric adenocarcinoma. This study aimed to construct and characterize a non-virulent Vibrio cholerae O1 strain, which grows more rapidly than H. pylori, as vector for H. pylori antigens for possible use as a vaccine strain against H. pylori. This was done by recombinant expression of the H. pylori adhesion antigen HpaA alone or, as a proof of principle, together with different colonization factor (CF) antigens of enterotoxigenic Escherichia coli (ETEC) which may enhance immune responses against HpaA. A recombinant V. cholerae strain co-expressing HpaA and a fimbrial CF antigens CFA/I or CS5, but not the non-fimbrial CF protein CS6, was shown to express larger amounts of HpaA on the surface when compared with the same V. cholerae strain expressing HpaA alone. Mutations in the CFA/I operon showed that the chaperon, possibly together with the usher, was involved in enhancing the surface expression of HpaA. Oral immunization of mice with formaldehyde-inactivated recombinant V. cholerae expressing HpaA alone or together with CFA/I induced significantly higher serum antibody responses against HpaA than mice similarly immunized with inactivated HpaA-expressing H. pylori bacteria. Our results demonstrate that a non-virulent V. cholerae strain can be engineered to allow strong surface expression of HpaA, and that the expression can be further increased by co-expressing it with ETEC fimbrial antigens. Such recombinant V. cholerae strains expressing HpaA, and possibly also other H. pylori antigens, may have the potential as oral inactivated vaccine candidates against H. pylori.
Collapse
|
12
|
Ma Z, Liu G, Zhang M, Li M, Liu Y, Yanfang J. Helicobacter pylori Infection Increases Frequency of PDCA-1(+) (CD317(+)) B-cell Subsets. Arch Med Res 2016; 47:96-104. [PMID: 27133710 DOI: 10.1016/j.arcmed.2016.04.003] [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] [Received: 11/10/2015] [Accepted: 04/11/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS As a newly discovered B-cell subset, PDCA-1(+) B cells have been shown to participate in the immune clearance of invading pathogens. The prominence of PDCA-1(+) B cell immunity in the pathogenesis of Helicobacter pylori infection prompted us to explore the potential role of this subset in gastric H. pylori infection. METHODS H. pylori infection was determined by (14)C-urea breath test and Western blot. The frequency of the different sub-compartments of PDCA-1(+) B cells and their relation to serum cytokines was determined in 33 H. pylori-infected and 14 uninfected patients and in 12 healthy controls (HC). RESULTS In comparison to uninfected individuals, there was a significantly increased frequency of PDCA-1(+) B cells, PDCA-1(+)IgM(+) B cells, CD93(+)PDCA-1(+) B cells, CD93(+)PDCA-1(+)IgM(+) B cells, CD137(+)PDCA-1(+) B cells and CD137(+)PDCA-1(+)IgM(+) B cells were detected in patients with H. pylori infection, corresponding to increased levels of serum IFN-α and IgM in this group. Compared with H. pylori-positive (HP(+)) chronic non-atrophic gastritis patients, a larger proportion of PDCA-1(+) B cells, CD93(+)PDCA-1(+) B cells and CD137(+)PDCA-1(+) B cells were observed in HP(+) patients suffering from atrophic gastritis or HP(+) peptic ulcers. CONCLUSIONS The frequency of the PDCA-1(+) B cell compartment is increased during H. pylori infection. Our data support the potential role of this B-cell subset in the pathogenesis of H. pylori-dependent gastritis.
Collapse
Affiliation(s)
- Zhaoyang Ma
- The First Hospital, Jilin University, Changchun, China
| | - Guangming Liu
- The First Hospital, Jilin University, Changchun, China
| | - Manli Zhang
- The First Hospital, Jilin University, Changchun, China
| | - Man Li
- The First Hospital, Jilin University, Changchun, China
| | - Yuanyuan Liu
- The First Hospital, Jilin University, Changchun, China.
| | - Jiang Yanfang
- The First Hospital, Jilin University, Changchun, China.
| |
Collapse
|
13
|
Immunodominant epitope-specific Th1 but not Th17 responses mediate protection against Helicobacter pylori infection following UreB vaccination of BALB/c mice. Sci Rep 2015; 5:14793. [PMID: 26434384 PMCID: PMC4593181 DOI: 10.1038/srep14793] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori (H. pylori) infects more than half of the world’s population, causing chronic gastritis, peptic ulcers and gastric cancer. Urease B subunit (UreB), a conserved protein of H. pylori, is capable of inducing specific CD4+ T-cell responses and provides protection against this infection. Previous studies have confirmed the effectiveness of rUreB subunit vaccines in generating CD4+ T-cell-mediated protection, but less is known regarding the roles of different subtypes of T-cell immunity, such as Th1, Th2 and Th17, particularly the immunodominant epitopes inducing specific CD4+ T-cell responses, in vaccine-mediated protection. In this study, we demonstrated that the vaccination of BALB/c mice with rUreB resulted in significant antigen-specific Th1 and Th17 immune responses. Importantly, two novel Th epitopes, UreB317–329 and UreB409–421, which are recognized by a major population of CD4+ T cells, were identified in immunized mice. Our results demonstrated that two novel epitopes can simultaneously induce Th1 and Th17 immune responses; however, only the epitope vaccine-induced CD4+ T-cells secreting IFN-γ mediated the protection against H. pylori; cells secreting IL-17A did not. Taken together, our results suggest that two novel immunodominant epitopes can induce Th1 and Th17 immune responses, but only the induced Th1 lymphocytes mediate protection against H. pylori.
Collapse
|
14
|
Zhou X, Liu W, Gu M, Zhou H, Zhang G. Helicobacter pylori infection causes hepatic insulin resistance by the c-Jun/miR-203/SOCS3 signaling pathway. J Gastroenterol 2015; 50:1027-40. [PMID: 25689935 DOI: 10.1007/s00535-015-1051-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/31/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Epidemiological studies have indicated that patients with chronic Helicobacter pylori infection have an increased risk of developing type 2 diabetes mellitus, but the underlying mechanisms remain largely unknown. This study aimed to investigate whether H. pylori infection contributes to the development of insulin resistance, as well as the underlying mechanisms both in vivo and in vitro. METHODS The effect of H. pylori infection on glucose metabolism was evaluated in humans and mouse models. The role of the c-Jun/miR-203/suppressor of cytokine signaling 3 (SOCS3) pathway in H. pylori-induced insulin resistance was determined in vitro and was validated in vivo. RESULTS Average fasting glucose levels were increased in patients (P = 0.012) and mice (P = 0.004) with H. pylori infection. Diabetic mice with H. pylori infection showed impaired glucose metabolism and insulin tolerance and hyperinsulinemia. Furthermore, H. pylori infection impaired insulin signaling in primary hepatocytes. H. pylori infection could upregulate SOCS3, a well-known insulin signaling inhibitor, by downregulating miR-203. SOCS3 overexpression interfered with insulin signaling proteins, and knockdown of SOCS3 alleviated H. pylori-induced impairment of insulin signaling. The transcription factor c-Jun, which affects gene expression, was induced by H. pylori infection and suppressed miR-203 expression. CONCLUSIONS Our results demonstrated that H. pylori infection induced hepatic insulin resistance by the c-Jun/miR-203/SOCS3 signaling pathway and provide possible implications with regard to resolving insulin resistance.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Wei Liu
- Department of Gastroenterology, The First People's Hospital of Suqian, Suqian, Jiangsu, China
| | - Min Gu
- Department of Pediatrics, Changzhou Children's Hospital, Changzhou, Jiangsu, China
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Guoxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
15
|
Yazbek PB, Trindade AB, Chin CM, Dos Santos JL. Challenges to the Treatment and New Perspectives for the Eradication of Helicobacter pylori. Dig Dis Sci 2015; 60:2901-12. [PMID: 25999247 DOI: 10.1007/s10620-015-3712-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/07/2015] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori (H. pylori) is one of the leading causes of gastric diseases such as chronic gastritis, peptic ulcer, and gastric adenocarcinoma. The current treatment of H. pylori infection with antibiotics and proton pump inhibitors has several limitations, including poor adherence and intrinsic patient-related factors, drug resistance, and the absence of adequate treatments. This review summarizes the current therapeutic approaches to eradicating H. pylori, the difficulties associated with its treatment, and several new perspectives aimed at improving existing treatment strategies.
Collapse
Affiliation(s)
- Priscila Baptistella Yazbek
- School of Pharmaceutical Science, Drugs and Medicines Department, State University of São Paulo "Júlio de Mesquita Filho", Rodovia Araraquara Jaú Km, 01 s/n, Araraquara, SP, 14801-902, Brazil
| | - Ariane Biolcati Trindade
- School of Pharmaceutical Science, Drugs and Medicines Department, State University of São Paulo "Júlio de Mesquita Filho", Rodovia Araraquara Jaú Km, 01 s/n, Araraquara, SP, 14801-902, Brazil
| | - Chung Man Chin
- School of Pharmaceutical Science, Drugs and Medicines Department, State University of São Paulo "Júlio de Mesquita Filho", Rodovia Araraquara Jaú Km, 01 s/n, Araraquara, SP, 14801-902, Brazil
| | - Jean Leandro Dos Santos
- School of Pharmaceutical Science, Drugs and Medicines Department, State University of São Paulo "Júlio de Mesquita Filho", Rodovia Araraquara Jaú Km, 01 s/n, Araraquara, SP, 14801-902, Brazil.
| |
Collapse
|
16
|
Nguyen TT, Kim SJ, Park JM, Hahm KB, Lee HJ. Repressed TGF-β signaling through CagA-Smad3 interaction as pathogenic mechanisms of Helicobacter pylori-associated gastritis. J Clin Biochem Nutr 2015; 57:113-20. [PMID: 26388668 PMCID: PMC4566024 DOI: 10.3164/jcbn.15-38] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 03/23/2015] [Indexed: 12/19/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection
causes chronic gastric inflammation, peptic ulceration, and gastric
carcinogenesis, in which H. pylori cytotoxin-associated gene A
(CagA) plays major pathogenic action. Since transforming growth factor-β
(TGF-β) and its signaling also are principally implicated in either
modulating gastric mucosal inflammatory responses or causing carcinogenesis and
are attenuated after H. pylori infection, we hypothesized that
dysregulated Smad signaling and repressed TGF-β might be core pathogenic
mechanism for H. pylori-associated gastritis or carcinogenesis.
Until now, no precise underlying mechanism how deranged TGF-β signaling
developed after H. pylori infection relevant to various
clinical manifestations remains unclear. In this study, we examined the
molecular mechanism about the inhibition of TGF-β signaling by H.
pylori CagA protein. H. pylori CagA significantly
suppressed TGF-β/Smad transcriptional responses through critical
inhibition of Smad3, though CagA interacted constitutively with Smad2, Smad3,
and Smad4. CagA inhibited TGF-β-induced suppression of proinflammatory
chemokines, such as IL-8, CXCL1 and CXCL3, as well as TGF-β-induced
transcription of target genes. In conclusion, repressed TGF-β signaling
associated with CagA-positive H. pylori infection could be an
important determinant for the outcome of H. pylori infection.
Therefore, TGF-β signaling is one of the important determinants to avoid
from H. pylori CagA pathogenicity.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Laboratory of Chemoprevention, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea
| | - Seong-Jin Kim
- CHA University Cancer Prevention Research Center, CHA Bio Complex, 335 Pangyo-ro, Gundang-gu, Seongnam 463-400, Korea
| | - Jong Min Park
- CHA University Cancer Prevention Research Center, CHA Bio Complex, 335 Pangyo-ro, Gundang-gu, Seongnam 463-400, Korea
| | - Ki Baik Hahm
- CHA University Cancer Prevention Research Center, CHA Bio Complex, 335 Pangyo-ro, Gundang-gu, Seongnam 463-400, Korea
| | - Ho-Jae Lee
- Laboratory of Chemoprevention, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 406-840, Korea
| |
Collapse
|
17
|
Yang J, Dai LX, Pan X, Wang H, Li B, Zhu J, Li MY, Shi XL, Wang BN. Protection against Helicobacter pylori infection in BALB/c mice by oral administration of multi-epitope vaccine of CTB-UreI-UreB. Pathog Dis 2015; 73:ftv026. [PMID: 25846576 DOI: 10.1093/femspd/ftv026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2015] [Indexed: 12/26/2022] Open
Abstract
Chronic gastric infection by the Gram-negative bacterium Helicobacter pylori (H. pylori) is strongly associated with gastritis, gastric ulcer and the development of distal gastric carcinoma and gastric mucosal lymphoma in humans. Antibiotic treatment of H. pylori is becoming less effective because of increasing antibiotic resistance; other treatment approaches such as specifically targeted methods, etc. to destroy this organism would be beneficial. An epitope vaccine is a promising option for protection against H. pylori infection. In this study, a multi-epitope vaccine was constructed by linking cholera toxin B subunit (CTB), two antigenic fragments of H. pylori urease I subunit (UreI20-29, UreI98-107) and four antigenic fragments of H. pylori urease B subunit (UreB12-23, UreB229-251, UreB327-400, UreB515-561), resulting in the recombinant CTB-UreI-UreB (BIB). Its protective effect against H. pylori infection was evaluated in BALB/c mice. Significant protection against H. pylori challenge was achieved in BALB/c mice immunized with BIB (15/18, 83.3%), rIB plus rCTB (6/18, 33.3%) and rIB (2/18, 11.1%) separately, while no protective effect was found in the mice immunized with either adjuvant rCTB alone or PBS. The induction of significant protection against H. pylori is possibly mediated by specific serum IgA and mucosal sIgA antibodies, and a mixed Th1/Th2/Th17 cells response. This multi-epitope vaccine might be a promising vaccine candidate that helps to control H. pylori infection.
Collapse
Affiliation(s)
- Jing Yang
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China Sichuan Vaccine Technology Co. Ltd, Chengdu, Sichuan 610041, People's Republic of China Department of Infectious Disease, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, People's Republic of China Department of Microbiology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Lv-xia Dai
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China Experiment Teaching Center of Clinical Medicine, Chengdu College of Medicine, Chengdu, Sichuan 610500, People's Republic of China
| | - Xing Pan
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China Sichuan Vaccine Technology Co. Ltd, Chengdu, Sichuan 610041, People's Republic of China
| | - Hongren Wang
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Bei Li
- Department of Infectious Disease, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, People's Republic of China Department of Microbiology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, People's Republic of China
| | - Jie Zhu
- Sichuan Vaccine Technology Co. Ltd, Chengdu, Sichuan 610041, People's Republic of China
| | - Ming-yuan Li
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Xin-Li Shi
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, People's Republic of China
| | - Bao-ning Wang
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
18
|
Cook KW, Crooks J, Hussain K, O'Brien K, Braitch M, Kareem H, Constantinescu CS, Robinson K, Gran B. Helicobacter pylori infection reduces disease severity in an experimental model of multiple sclerosis. Front Microbiol 2015; 6:52. [PMID: 25762984 PMCID: PMC4327743 DOI: 10.3389/fmicb.2015.00052] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/15/2015] [Indexed: 12/19/2022] Open
Abstract
Recent research has demonstrated that infection with the bacterial pathogen Helicobacter pylori is less common amongst patients with multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system (CNS). We aimed to compare the prevalence of H. pylori amongst MS patients and healthy controls, and also investigated the impact of this infection on an animal model for MS, experimental autoimmune encephalomyelitis (EAE). The H. pylori status of 71 MS patients and 42 healthy controls was determined by serology. Groups of C57BL/6 mice were infected with H. pylori, or given diluent alone as a placebo, prior to inducing EAE. Clinical scores were assessed for all mice, and spleens and spinal cord tissue were harvested. CD4+ T cell subsets were quantified by flow cytometry, and T cell proliferation assays were performed. In MS patients the seroprevalence of H. pylori was half that of healthy controls (p = 0.018). Over three independent experiments, prior H. pylori infection had a moderate effect in reducing the severity of EAE (p = 0.012). In line with this, the antigen-specific T cell proliferative responses of infected animals were significantly reduced (p = 0.001), and there was a fourfold reduction in the number of CD4+ cells in the CNS. CD4+ populations in both the CNS and the spleens of infected mice also contained greatly reduced proportions of IFNγ+, IL-17+, T-bet+, and RORγt+ cells, but the proportions of Foxp3+ cells were equivalent. There were no differences in the frequency of splenic CD4+cells expressing markers of apoptosis between infected and uninfected animals. H. pylori was less prevalent amongst MS patients. In mice, the infection exerted some protection against EAE, inhibiting both Th1 and Th17 responses. This could not be explained by the presence of increased numbers of Foxp3+ regulatory T cells, or T cell apoptosis. This is the first direct experimental evidence showing that H. pylori may provide protection against inflammatory demyelination in the CNS.
Collapse
Affiliation(s)
- Katherine W Cook
- Nottingham Digestive Diseases Biomedical Research Unit, Centre for Biomolecular Sciences, University of Nottingham School of Medicine Nottingham, UK
| | - James Crooks
- Clinical Neurology Research Group, Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, UK
| | - Khiyam Hussain
- Nottingham Digestive Diseases Biomedical Research Unit, Centre for Biomolecular Sciences, University of Nottingham School of Medicine Nottingham, UK
| | - Kate O'Brien
- Clinical Neurology Research Group, Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, UK
| | - Manjit Braitch
- Clinical Neurology Research Group, Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, UK
| | - Huner Kareem
- Clinical Neurology Research Group, Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, UK
| | - Cris S Constantinescu
- Clinical Neurology Research Group, Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, UK
| | - Karen Robinson
- Nottingham Digestive Diseases Biomedical Research Unit, Centre for Biomolecular Sciences, University of Nottingham School of Medicine Nottingham, UK
| | - Bruno Gran
- Clinical Neurology Research Group, Division of Clinical Neuroscience, University of Nottingham School of Medicine Nottingham, UK
| |
Collapse
|
19
|
Differential expression of microRNAs in preneoplastic gastric mucosa. Sci Rep 2015; 5:8270. [PMID: 25652892 PMCID: PMC4317705 DOI: 10.1038/srep08270] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/14/2015] [Indexed: 12/14/2022] Open
Abstract
Gastric carcinogenesis is a multifactorial H.pylori-triggered dynamic process that goes through a cascade of preneoplastic conditions. The expression of miRNAs in the stomach with regard to preneoplastic precursor conditions and H.pylori infection has not been investigated systematically. In this prospective proof-of-principle study, we evaluated the miRNA expression in gastric antrum and corpus mucosa from patients with chronic non-atrophic gastritis (CNAG), atrophic gastritis (AG), and GC compared to controls. Gastric normal mucosa shows a unique expression pattern for miR-21, miR-155 and miR-223, which is specific for different regions. In correlation with progression of Correa's cascade and H.pylori infection, we observed a gradual increase in miR-155 and miR-223 both in corpus and antrum and miR-21 only in the antrum mucosa. Using miRNA expression we calculated a score that allowed us to discriminate patients with AG from subjects with normal mucosa with high diagnostic accuracy in testing and validation cohorts reproducibly. In summary, the expression pattern of miRNAs in the gastric mucosa is gradually increased with progression of Correa's cascade and H.pylori infection, suggesting miRNAs as potential biomarkers for preneoplastic precursor conditions. However, differences of miRNA expression between the gastric antrum and the corpus need to be considered in future studies.
Collapse
|
20
|
Milani M, Sharifi Y, Rahmati-Yamchi M, Somi MH, Akbarzadeh A. Immunology and vaccines and nanovaccines for Helicobacter pylori infection. Expert Rev Vaccines 2015; 14:833-40. [PMID: 25645086 DOI: 10.1586/14760584.2015.1008460] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori infection is very common worldwide and is an important cause of gastritis, peptic ulcer disease, gastric mucosa-associated lymphoid tissue lymphoma, and gastric adenocarcinoma. Since the eradication requires treatment with multidrug regimens, prevention of primary infection by a suitable vaccine is attractive. Developing vaccines on the spot when and where an infection is breaking out might be possible, thanks to engineered nanoparticles. In this review, the nature of the host immune response to H. pylori infection is considered. We explain recent candidate vaccines and prophylactic or therapeutic immunization strategies for use against H. pylori. We also describe identification of different types of immune responses that may be related to protection against H. pylori infection. Thus, it seems that there is still a strong need to clarify the main protective immune response against H. pylori.
Collapse
Affiliation(s)
- Morteza Milani
- Liver and Gastrointestinal disease research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | |
Collapse
|
21
|
Yuan XG, Huang YJ, Hu HS. Role of Tregs in Helicobacter pylori infection. Shijie Huaren Xiaohua Zazhi 2014; 22:4714-4718. [DOI: 10.11569/wcjd.v22.i30.4714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of Foxp3+ regulatory T cells (Tregs) in Helicobacter pylori (H. pylori) infection.
METHODS: By using biopsies from 89 H. pylori-positive patients, 60 of whom underwent treatment for eradication of H. pylori, and 79 H. pylori-negative adults, immunohistochemistry was performed to semi-quantify the number of Tregs in the gastric mucosa.
RESULTS: The proportion of Tregs was 2.4% in H. pylori-negative individuals, and 8.9% in H. pylori-positive patients (P < 0.01). After eradication of H. pylori, the proportion of Tregs significantly declined (3.0%, P < 0.01).
CONCLUSION: Tregs actively participate in the immune response to H. pylori infection. H. pylori chronically colonizes the stomach and increases the frequency of Tregs in the gastric mucosa, thereby suppressing the immune response and contributing to the persistence of H. pylori infection. Tregs decrease in the gastric mucosa after eradication of H. pylori, and mucosal immune balance is then restored.
Collapse
|
22
|
Smith SM. Role of Toll-like receptors in Helicobacter pylori infection and immunity. World J Gastrointest Pathophysiol 2014; 5:133-146. [PMID: 25133016 PMCID: PMC4133513 DOI: 10.4291/wjgp.v5.i3.133] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/25/2014] [Accepted: 05/19/2014] [Indexed: 02/07/2023] Open
Abstract
The gram-negative bacterium Helicobacter pylori (H. pylori) infects the stomachs of approximately half of the world’s population. Although infection induces an immune response that contributes to chronic gastric inflammation, the response is not sufficient to eliminate the bacterium. H. pylori infection causes peptic ulcers, gastric cancer and mucosa-associated lymphoid tissue lymphoma. Disease outcome is linked to the severity of the host inflammatory response. Gastric epithelial cells represent the first line of innate immune defence against H. pylori, and respond to infection by initiating numerous cell signalling cascades, resulting in cytokine induction and the subsequent recruitment of inflammatory cells to the gastric mucosa. Pathogen recognition receptors of the Toll-like receptor (TLR) family mediate many of these cell signalling events. This review discusses recent findings on the role of various TLRs in the recognition of H. pylori in distinct cell types, describes the TLRs responsible for the recognition of individual H. pylori components and outlines the influence of innate immune activation on the subsequent development of the adaptive immune response. The mechanistic identification of host mediators of H. pylori-induced pathogenesis has the potential to reveal drug targets and opportunities for therapeutic intervention or prevention of H. pylori-associated disease by means of vaccines or immunomodulatory therapy.
Collapse
|
23
|
|
24
|
Hanada K, Graham DY. Helicobacter pylori and the molecular pathogenesis of intestinal-type gastric carcinoma. Expert Rev Anticancer Ther 2014; 14:947-54. [PMID: 24802804 DOI: 10.1586/14737140.2014.911092] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gastric carcinoma is an inflammation-related cancer caused by long-term infection with the human bacterial pathogen, Helicobacter pylori. The pattern of acute-on-chronic inflammation causes progressive mucosal damage which may result in atrophy with metaplastic epithelia and eventually gastric cancer. Recently, it has been recognized that H. pylori can also cause genetic instability such as double-stranded DNA breaks and can produce gene activation and silencing via epigenetic pathways. As genetic instability is the hallmark of cancer, we highlight recent progress in understanding the gastric carcinogenesis in relation to H. pylori-related inflammation, H. pylori-induced double-stranded DNA breakage and aberrant gene expression as well as the mechanisms and role of H. pylori-associated epigenetic change in gene expression.
Collapse
Affiliation(s)
- Katsuhiro Hanada
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Japan
| | | |
Collapse
|