1
|
Yang W, Ou Y, Luo H, You L, Du H. Causal relationship between circulating immune cells and gastric cancer: a bidirectional Mendelian randomization analysis using UK Biobank and FinnGen datasets. Transl Cancer Res 2024; 13:4702-4713. [PMID: 39430856 PMCID: PMC11483344 DOI: 10.21037/tcr-24-480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/01/2024] [Indexed: 10/22/2024]
Abstract
Background The role of immune cells in cancer pathogenesis remains controversial due to conflicting reports, potentially arising from various confounding factors. Emerging evidence suggests that cancer can also influence immune cell populations and functions, making it challenging to investigate their causal relationship. Traditional observational studies often fail to eliminate all confounding factors and are prone to reverse causality. Therefore, we employ Mendelian randomization (MR) to determine the causal relationship between immune cells and cancer, as this method can identify causal relationships independent of confounding factors and avoid reverse causality. Methods Genome-wide association study (GWAS) summary statistics on immune traits, encompassing 310 immune cell phenotypes, were obtained from 3,757 European individuals, with peripheral blood immune cells tested using flow cytometry. GWAS summary statistics for gastric cancer were derived from 476,116 European individuals across two large-scale biobanks: the UK Biobank and FinnGen. Gastric cancer was identified by the International Classification of Diseases, 9th Revision (ICD-9), and 10th Revision (ICD-10) codes. Significant single nucleotide polymorphisms (SNPs) for immune traits were extracted at a threshold of P<1×10-5, while a threshold of P<5×10-8 was used for gastric cancer GWAS data. Linkage imbalance-based clumping was performed to obtain independent SNPs, and those with F<10 were excluded to mitigate weak instrument bias. Phenoscanner V2 was used to exclude SNPs directly associated with potential confounders or outcomes. Two-sample MR was conducted using five MR methods, with inverse-variance-weighted (IVW) as the primary analysis method. A false discovery rate (FDR) correction was used to reduce the likelihood of type 1 errors. In addition, we conducted MR-Egger intercept tests and Cochran's Q tests. Results The numbers of CD4-CD8- T cells and IgD-CD27- B cells were positively correlated with the development of gastric cancer, with odds ratios (ORs) of 1.15 [95% confidence interval (CI), 1.07-1.24; P<0.001; PFDR=0.041; IVW method] and 1.07 (95% CI, 1.03-1.11; P=0.001; PFDR=0.187; IVW method), respectively. However, the percentage of IgD+CD24- B cells in lymphocytes were negatively associated with the development of gastric cancer (OR =0.90; 95% CI, 0.84-0.96; P=0.002; PFDR=0.187; IVW method). MR analysis of the above three immune cell phenotypes showed no significant heterogeneity or horizontal pleiotropy. In the reverse MR analysis, gastric cancer was not causally associated with any of the immune cell phenotypes. Conclusions Circulating CD4-CD8- T cells and IgD-CD27- B cells are positively correlated with the development of gastric cancer, while the percentage of IgD+CD24- B cells in lymphocytes are negatively correlated. These findings provide insight into the relationship between immune cells and gastric cancer pathogenesis and may serve as a basis for the development of immunotherapies for gastric cancer.
Collapse
Affiliation(s)
- Weimin Yang
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, China
| | - Yang Ou
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, China
| | - Hui Luo
- Department of Gastrointestinal Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Lijuan You
- Department of Anesthesiology, Huanggang Central Hospital Affiliated to Yangtze University, Huanggang, China
| | - Heng Du
- Department of Gastrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Huanggang, China
| |
Collapse
|
2
|
Teng Y, Dong Q, Zhang S, Chen S, Li C. Clinical analysis of the effect of helicobacter pylori infection on immune function in children with peptic ulcer. Pak J Med Sci 2024; 40:1063-1066. [PMID: 38952501 PMCID: PMC11190406 DOI: 10.12669/pjms.40.6.7820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/06/2023] [Accepted: 02/21/2024] [Indexed: 07/03/2024] Open
Abstract
Objective To study whether children with peptic ulcer would have abnormalities in cellular and humoral immune functions, and whether Helicobacter pylori (Hp) infection would affect the immune function of children with peptic ulcer. Methods This is a retrospective study. The subjects of study were 72 children with diagnosed and cured peptic ulcer (ulcer group), and 50 healthy children with physical examination (control group) at Baoding Hospital, Beijing Children's Hospital Affiliated to Capital Medical University from June 2020 to December 2022. Further detection was conducted on T lymphocyte subsets (CD3+, CD4+, CD8+, and CD4+/CD8+ ratio) and immunoglobulin levels. Results Of the 72 children with peptic ulcer, 53(73.6%) were positive for Hp (Hp-positive group) and 19 (26.4%) were negative (Hp-negative group). The levels of CD3+, CD4+, and CD4+/CD8+ ratio in the control group were significantly higher than those in the ulcer group, with statistically significant difference (P<0.05); while the level of IgG in the control group was lower than that in the ulcer group, with statistically significant difference (P<0.05). Meanwhile, there were statistically significant differences in that the levels of CD3+, CD4+ and CD8+ were increased in Hp-positive group than those in Hp-negative group before treatment (P<0.05); while CD4+/CD8+ ratio was lower in the former group than that in the latter group, with statistically significant difference (P<0.05). Conclusion Hp infection can induce the elevation of T lymphocyte subsets. The development of peptic ulcer has an intimate association with the disorder of cellular and humoral immune functions.
Collapse
Affiliation(s)
- Yongnan Teng
- Yongnan Teng, Department of Gastroenterology, Baoding Hospital, Beijing Children’s Hospital Affiliated to Capital Medical University, Baoding, Hebei, 071000, P.R. China; Key Laborary of Clinical, Research on Respiratory Digestive Disease, Hebei Baoding, 071000, China
| | - Qingwei Dong
- Qingwei Dong, Department of Gastroenterology, Baoding Hospital, Beijing Children’s Hospital Affiliated to Capital Medical University, Baoding, Hebei, 071000, P.R. China; Key Laborary of Clinical, Research on Respiratory Digestive Disease, Hebei Baoding, 071000, China
| | - Sisi Zhang
- Sisi Zhang, Department of Gastroenterology, Baoding Hospital, Beijing Children’s Hospital Affiliated to Capital Medical University, Baoding, Hebei, 071000, P.R. China; Key Laborary of Clinical, Research on Respiratory Digestive Disease, Hebei Baoding, 071000, China
| | - Songsong Chen
- Songsong Chen, Department of Gastroenterology, Baoding Hospital, Beijing Children’s Hospital Affiliated to Capital Medical University, Baoding, Hebei, 071000, P.R. China; Key Laborary of Clinical, Research on Respiratory Digestive Disease, Hebei Baoding, 071000, China
| | - Chen Li
- Chen Li, Department of Gastroenterology, Baoding Hospital, Beijing Children’s Hospital Affiliated to Capital Medical University, Baoding, Hebei, 071000, P.R. China; Key Laborary of Clinical, Research on Respiratory Digestive Disease, Hebei Baoding, 071000, China
| |
Collapse
|
3
|
Skakic I, Taki AC, Francis JE, Dekiwadia C, Van TTH, Joe CCD, Phan T, Lovrecz G, Gorry PR, Ramsland PA, Walduck AK, Smooker PM. Nanocapsules Comprised of Purified Protein: Construction and Applications in Vaccine Research. Vaccines (Basel) 2024; 12:410. [PMID: 38675791 PMCID: PMC11053559 DOI: 10.3390/vaccines12040410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Nanoparticles show great promise as a platform for developing vaccines for the prevention of infectious disease. We have been investigating a method whereby nanocapsules can be formulated from protein, such that the final capsules contain only the cross-linked protein itself. Such nanocapsules are made using a silica templating system and can be customised in terms of size and porosity. Here we compare the construction and characteristics of nanocapsules from four different proteins: one a model protein (ovalbumin) and three from infectious disease pathogens, namely the influenza virus, Helicobacter pylori and HIV. Two of the nanocapsules were assessed further. We confirm that nanocapsules constructed from the urease A subunit of H. pylori can reduce subsequent infection in a vaccinated mouse model. Further, we show that capsules constructed from the HIV gp120 protein can be taken up by dendritic cells in tissue culture and can be recognised by antibodies raised against the virus. These results point to the utility of this method in constructing protein-only nanocapsules from proteins of varying sizes and isoelectric points.
Collapse
Affiliation(s)
- Ivana Skakic
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (I.S.); (J.E.F.); (T.T.H.V.); (P.A.R.)
| | - Aya C. Taki
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Jasmine E. Francis
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (I.S.); (J.E.F.); (T.T.H.V.); (P.A.R.)
| | - Chaitali Dekiwadia
- RMIT Microscopy & Microanalysis Facility, School of Science, RMIT University, Melbourne, VIC 3001, Australia;
| | - Thi Thu Hao Van
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (I.S.); (J.E.F.); (T.T.H.V.); (P.A.R.)
| | - Carina C. D. Joe
- CSIRO Manufacturing, Clayton, VIC 3169, Australia; (C.C.D.J.); (T.P.); (G.L.)
| | - Tram Phan
- CSIRO Manufacturing, Clayton, VIC 3169, Australia; (C.C.D.J.); (T.P.); (G.L.)
| | - George Lovrecz
- CSIRO Manufacturing, Clayton, VIC 3169, Australia; (C.C.D.J.); (T.P.); (G.L.)
| | - Paul R. Gorry
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute of Infection and Immunity, Parkville, VIC 3010, Australia;
| | - Paul A. Ramsland
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (I.S.); (J.E.F.); (T.T.H.V.); (P.A.R.)
- Department of Immunology, Monash University, Melbourne, VIC 3004, Australia
- Department of Surgery, Austin Health, The University of Melbourne, Heidelberg, VIC 3084, Australia
| | - Anna K. Walduck
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (I.S.); (J.E.F.); (T.T.H.V.); (P.A.R.)
| | - Peter M. Smooker
- School of Science, RMIT University, 264 Plenty Road, Bundoora, VIC 3083, Australia; (I.S.); (J.E.F.); (T.T.H.V.); (P.A.R.)
| |
Collapse
|
4
|
Engelsberger V, Gerhard M, Mejías-Luque R. Effects of Helicobacter pylori infection on intestinal microbiota, immunity and colorectal cancer risk. Front Cell Infect Microbiol 2024; 14:1339750. [PMID: 38343887 PMCID: PMC10853882 DOI: 10.3389/fcimb.2024.1339750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Infecting about half of the world´s population, Helicobacter pylori is one of the most prevalent bacterial infections worldwide and the strongest known risk factor for gastric cancer. Although H. pylori colonizes exclusively the gastric epithelium, the infection has also been associated with various extragastric diseases, including colorectal cancer (CRC). Epidemiological studies reported an almost two-fold increased risk for infected individuals to develop CRC, but only recently, direct causal and functional links between the chronic infection and CRC have been revealed. Besides modulating the host intestinal immune response, H. pylori is thought to increase CRC risk by inducing gut microbiota alterations. It is known that H. pylori infection not only impacts the gastric microbiota at the site of infection but also leads to changes in bacterial colonization in the distal large intestine. Considering that the gut microbiome plays a driving role in CRC, H. pylori infection emerges as a key factor responsible for promoting changes in microbiome signatures that could contribute to tumor development. Within this review, we want to focus on the interplay between H. pylori infection, changes in the intestinal microbiota, and intestinal immunity. In addition, the effects of H. pylori antibiotic eradication therapy will be discussed.
Collapse
Affiliation(s)
| | | | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
5
|
Elkoshi Z. The Eradication of Carcinogenic Viruses in Established Solid Cancers. J Inflamm Res 2023; 16:6227-6239. [PMID: 38145011 PMCID: PMC10749098 DOI: 10.2147/jir.s430315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/12/2023] [Indexed: 12/26/2023] Open
Abstract
Carcinogenic viruses (oncoviruses) can initiate cancer, but their impact on established cancer varies. Some of these viruses prolong survival while others shorten it. This study classifies oncoviruses into two categories: viruses which induce a strong CD8+T cell reaction in non-cancerous tissues, and viruses which induce a weak CD8+ T cell reaction in non-cancerous tissues. The classification proves useful in predicting the effect of oncoviruses on the prognosis of solid cancers. Therefore, while eliminating carcinogenic viruses in healthy individuals (for example by immunization) may be important for cancer prevention, this study suggests that only viruses which induce a weak CD8+ T cell reaction should be eradicated in established solid tumors. The model correctly predicts the effect of oncoviruses on survival for six out of seven known oncoviruses, indicating that immune modulation by oncoviruses has a prominent effect on prognosis. It seems that CD8+ T cell response to oncoviruses observed in infected benign tissues is retained in infected tumors. Clinical significance: the effect of oncoviruses on solid cancer prognosis can be predicted with confidence based on immunological responses when clinical data are unavailable.
Collapse
Affiliation(s)
- Zeev Elkoshi
- Research and Development Department, Taro Pharmaceutical Industries Ltd, Haifa, Israel
| |
Collapse
|
6
|
Zhang Z, Chen X, Li B, Xia T, Wu X, Wu C. Helicobacter pylori induces urease subunit B-specific CD8 + T cell responses in infected individuals via cytosolic pathway of cross-presentation. Helicobacter 2023; 28:e13005. [PMID: 37382428 DOI: 10.1111/hel.13005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Urease subunit B (UreB), a conserved and key virulence factor of Helicobacter pylori (H. pylori), can induce the host CD4+ T cell immune responses to provide protection, but less is known regarding CD8+ T cell responses. The characteristics of H. pylori-specific CD8+ T cell responses and the mechanism underlying antigen processing and presentation pathways remain unclear. This study was focus on protective antigen recombinant UreB (rUreb) to detect specific CD8+ T cell responses in vitro and elucidate the mechanism of UreB antigen processing and presentation. METHODS The peripheral blood mononuclear cells (PBMCs) collected from H. pylori-infected individuals were stimulated with rUreB in vitro to detect specific CD8+ T cell responses after co-culture with rUreB-pulsed autologous hMDCs. Through blocking assay, we investigated the potential pathway of UreB antigen processing and presentation via the cytosolic pathway or vacuolar pathway. The cytokines production of UreB specific CD8+ T cell were evaluated as well. RESULTS We demonstrated UreB can induce specific CD8+ T cell immune responses in H. pylori infected individuals. Importantly, we characterized that UreB were mainly processed by proteasome instead of lysosomal proteases and presented through cytosolic pathway of cross-presentation, which requires endoplasmic reticulum-Golgi transport and newly synthesized MHC-I molecules, to induce functional-specific CD8+ T cell (IFN-γ + TNF-α + Grz A+ Grz B+) responses. CONCLUSIONS These results suggest that H. pylori UreB induces specific CD8+ T cell responses through cytosolic pathway of cross-presentation in infected individuals.
Collapse
Affiliation(s)
- Zelin Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xingchi Chen
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bin Li
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tingting Xia
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaobin Wu
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
7
|
Tjandra D, Busuttil RA, Boussioutas A. Gastric Intestinal Metaplasia: Challenges and the Opportunity for Precision Prevention. Cancers (Basel) 2023; 15:3913. [PMID: 37568729 PMCID: PMC10417197 DOI: 10.3390/cancers15153913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
GIM is a persistent, premalignant lesion whereby gastric mucosa is replaced by metaplastic mucosa resembling intestinal tissue, arising in the setting of chronic inflammation, particularly in the context of Helicobacter pylori. While the overall rates of progression to gastric adenocarcinoma are low, estimated at from 0.25 to 2.5%, there are features that confer a much higher risk and warrant follow-up. In this review, we collate and summarise the current knowledge regarding the pathogenesis of GIM, and the clinical, endoscopic and histologic risk factors for cancer. We examine the current state-of-practice with regard to the diagnosis and management of GIM, which varies widely in the published guidelines and in practice. We consider the emerging evidence in population studies, artificial intelligence and molecular markers, which will guide future models of care. The ultimate goal is to increase the detection of early gastric dysplasia/neoplasia that can be cured while avoiding unnecessary surveillance in very low-risk individuals.
Collapse
Affiliation(s)
- Douglas Tjandra
- Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia;
- Department of Gastroenterology, The Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Rita A. Busuttil
- Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia;
- Department of Gastroenterology, The Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Alex Boussioutas
- Central Clinical School, Monash University, 99 Commercial Rd, Melbourne, VIC 3004, Australia;
- Department of Gastroenterology, The Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
8
|
Ahmed AAQ, Besio R, Xiao L, Forlino A. Outer Membrane Vesicles (OMVs) as Biomedical Tools and Their Relevance as Immune-Modulating Agents against H. pylori Infections: Current Status and Future Prospects. Int J Mol Sci 2023; 24:ijms24108542. [PMID: 37239888 DOI: 10.3390/ijms24108542] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Outer membrane vesicles (OMVs) are lipid-membrane-bounded nanoparticles that are released from Gram-negative bacteria via vesiculation of the outer membrane. They have vital roles in different biological processes and recently, they have received increasing attention as possible candidates for a broad variety of biomedical applications. In particular, OMVs have several characteristics that enable them to be promising candidates for immune modulation against pathogens, such as their ability to induce the host immune responses given their resemblance to the parental bacterial cell. Helicobacter pylori (H. pylori) is a common Gram-negative bacterium that infects half of the world's population and causes several gastrointestinal diseases such as peptic ulcer, gastritis, gastric lymphoma, and gastric carcinoma. The current H. pylori treatment/prevention regimens are poorly effective and have limited success. This review explores the current status and future prospects of OMVs in biomedicine with a special focus on their use as a potential candidate in immune modulation against H. pylori and its associated diseases. The emerging strategies that can be used to design OMVs as viable immunogenic candidates are discussed.
Collapse
Affiliation(s)
- Abeer Ahmed Qaed Ahmed
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100 Pavia, Italy
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100 Pavia, Italy
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-sen University, Shenzhen 518107, China
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
9
|
Koch MRA, Gong R, Friedrich V, Engelsberger V, Kretschmer L, Wanisch A, Jarosch S, Ralser A, Lugen B, Quante M, Vieth M, Vasapolli R, Schulz C, Buchholz VR, Busch DH, Mejías-Luque R, Gerhard M. CagA-specific Gastric CD8 + Tissue-Resident T Cells Control Helicobacter pylori During the Early Infection Phase. Gastroenterology 2023; 164:550-566. [PMID: 36587707 DOI: 10.1053/j.gastro.2022.12.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS Infection with Helicobacter pylori strongly affects global health by causing chronic gastritis, ulcer disease, and gastric cancer. Although extensive research into the strong immune response against this persistently colonizing bacterium exists, the specific role of CD8+ T cells remains elusive. METHODS We comprehensively characterize gastric H pylori-specific CD8+ T-cell responses in mice and humans by flow cytometry, RNA-sequencing, immunohistochemistry, and ChipCytometry, applying functional analyses including T-cell depletion, H pylori eradication, and ex vivo restimulation. RESULTS We define CD8+ T-cell populations bearing a tissue-resident memory (TRM) phenotype, which infiltrate the gastric mucosa shortly after infection and mediate pathogen control by executing antigen-specific effector properties. These induced CD8+ tissue-resident memory T cells (TRM cells) show a skewed T-cell receptor beta chain usage and are mostly specific for cytotoxin-associated gene A, the distinctive oncoprotein injected by H pylori into host cells. As the infection progresses, we observe a loss of the TRM phenotype and replacement of CD8+ by CD4+ T cells, indicating a shift in the immune response during the chronic infection phase. CONCLUSIONS Our results point toward a hitherto unknown role of CD8+ T-cell response in this bacterial infection, which may have important clinical implications for treatment and vaccination strategies against H pylori.
Collapse
Affiliation(s)
- Maximilian R A Koch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Ruolan Gong
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Verena Friedrich
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Veronika Engelsberger
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Lorenz Kretschmer
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Andreas Wanisch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Sebastian Jarosch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Anna Ralser
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Bob Lugen
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Michael Quante
- Technical University of Munich (TUM), School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, Munich, Germany; Department of Internal Medicine II, University Hospital Freiburg, University Freiburg, Freiburg, Germany
| | - Michael Vieth
- Institute of Pathology, Hospital Bayreuth, Friedrich Alexander University, Erlangen-Nuremberg, Bayreuth, Germany
| | - Riccardo Vasapolli
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany; Medical Department II, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Christian Schulz
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany; Medical Department II, University Hospital Großhadern, Ludwig-Maximilians-University, Munich, Germany
| | - Veit R Buchholz
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | - Dirk H Busch
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Raquel Mejías-Luque
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany.
| | - Markus Gerhard
- Technical University of Munich (TUM), School of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, Munich, Germany; German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany.
| |
Collapse
|
10
|
He T, Zhang F, Zhang J, Wei S, Ning J, Yuan H, Li B. UreB immunodominant epitope-specific CD8 + T-cell responses were beneficial in reducing gastric symptoms in Helicobacter pylori-infected individuals. Helicobacter 2023; 28:e12959. [PMID: 36828665 DOI: 10.1111/hel.12959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/18/2023] [Accepted: 02/06/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND AND AIMS Although Helicobacter pylori is recognized as an extracellular infection bacterium, it can lead to an increase in the number of CD8+ T cells after infection. At present, the characteristics of H. pylori antigen-specific CD8+ T cells and the epitope response have not been elucidated. This study was focused on putative protective antigen UreB to detect specific CD8+ T-cell responses in vitro and screen for predominant response epitopes. METHODS The PBMCs collected from H. pylori-infected individuals were stimulated by UreB peptide pools in vitro to identify the immunodominant CD8+ T-cell epitopes. Furthermore, their HLA restriction characteristics were detected accordingly by NGS. Finally, the relationship between immunodominant responses and appearance of gastric symptoms after H. pylori infection was conducted. RESULTS UreB-specific CD8+ T-cell responses were detected in H. pylori-infected individuals. Three of UreB dominant epitopes (A-2 (UreB443-451 : GVKPNMIIK), B-4 (UreB420-428 : SEYVGSVEV), and C-1 (UreB5-13 : SRKEYVSMY)) were firstly identified and mainly presented by HLA-A*1101, HLA-B*4001 and HLA-C*0702 alleles, respectively. C-1 responses were mostly occurred in H. pylori-infected subjects without gastric symptoms and may alleviate the degree of gastric inflammation. CONCLUSIONS The UreB dominant epitope-specific CD8+ T-cell response was closely related to the gastric symptoms after H. pylori infection, and the C-1 (UreB5-13 ) dominant peptides may be protective epitopes.
Collapse
Affiliation(s)
- Taojun He
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Fang Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jin Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shanshan Wei
- Department of Digestive Endoscopy Center, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jie Ning
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Hanmei Yuan
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bin Li
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
11
|
Fuchs S, Gong R, Gerhard M, Mejías-Luque R. Immune Biology and Persistence of Helicobacter pylori in Gastric Diseases. Curr Top Microbiol Immunol 2023; 444:83-115. [PMID: 38231216 DOI: 10.1007/978-3-031-47331-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Helicobacter pylori is a prevalent pathogen, which affects more than 40% of the global population. It colonizes the human stomach and persists in its host for several decades or even a lifetime, if left untreated. The persistent infection has been linked to various gastric diseases, including gastritis, peptic ulcers, and an increased risk for gastric cancer. H. pylori infection triggers a strong immune response directed against the bacterium associated with the infiltration of innate phagocytotic immune cells and the induction of a Th1/Th17 response. Even though certain immune cells seem to be capable of controlling the infection, the host is unable to eliminate the bacteria as H. pylori has developed remarkable immune evasion strategies. The bacterium avoids its killing through innate recognition mechanisms and manipulates gastric epithelial cells and immune cells to support its persistence. This chapter focuses on the innate and adaptive immune response induced by H. pylori infection, and immune evasion strategies employed by the bacterium to enable persistent infection.
Collapse
Affiliation(s)
- Sonja Fuchs
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Ruolan Gong
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany.
| |
Collapse
|
12
|
Iwamuro M, Takahashi T, Watanabe N, Abe M, Sakae H, Kono Y, Kanzaki H, Tanaka T, Kawano S, Otsuka F, Kawahara Y, Yanai H, Okada H. Site-specific differences in T lymphocyte composition of the gastric mucosa after Helicobacter pylori eradication. Medicine (Baltimore) 2022; 101:e30241. [PMID: 36042652 PMCID: PMC9410669 DOI: 10.1097/md.0000000000030241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
In our earlier work, we revealed that inflammation of the lesser curvature of the gastric body and antrum could constitute independent risk factors for gastric cancer development, while inflammation of the greater curvature was not. The aims of this study were as follows: first, to reveal the differences between T lymphocyte populations of the gastric antrum and the greater and lesser curvatures of the gastric body in patients after Helicobacter pylori eradication; second, to analyze the correlation between the composition of the stomach-resident T lymphocytes and time from H. pylori eradication; and third, to evaluate the sex differences in T lymphocyte subsets after H. pylori eradication. To investigate site-specific differences in stomach-resident T lymphocytes after H. pylori eradication, we performed flow cytometry analysis on samples taken from the gastric antrum, greater curvature of the gastric body, and lesser curvature of the gastric body of 20 patients. We also analyzed the correlation between the composition of the stomach-resident T lymphocytes and the time from H. pylori eradication. The lymphocyte subsets of the antrum and lesser curvature of the body were similar. In contrast, compared to those in the greater curvature of the gastric body, CD4+/CD3+ lymphocyte subsets (43.8 ± 19.4% vs 31.7 ± 14.6%) were elevated in the lesser curvature of the body, whereas CD8+/CD3+ (67.1 ± 21.3% vs 80.4 ± 12.0%), CD7+/CD3+ (91.2 ± 4.6% vs 93.7 ± 3.8%), CCR4+/CD3+ (7.7 ± 8.1% vs 10.4 ± 7.0%), CD45RA+/CD3+CD4+ (27.2 ± 24.8% vs 39.5 ± 20.8%), and CD45RA+/CD3+CD4- (14.2 ± 11.1% vs 18.7 ± 11.5) were lower. Linear regression analysis showed a negative correlation between the time after H. pylori eradication and CD4+/CD3+ (P < .05, R2 = 0.198). There were no significant differences between men and women with respect to the lymphocyte populations. These results indicate that there are site-specific differences in lymphocyte composition in the stomach after H. pylori eradication.
Collapse
Affiliation(s)
- Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
- *Correspondence: Masaya Iwamuro, Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan (e-mail: )
| | - Takahide Takahashi
- Division of Medical Support, Okayama University Hospital, Okayama, Japan
| | - Natsuki Watanabe
- Division of Medical Support, Okayama University Hospital, Okayama, Japan
| | - Makoto Abe
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Sakae
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshiyasu Kono
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromitsu Kanzaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Takehiro Tanaka
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Seiji Kawano
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshiro Kawahara
- Department of Practical Gastrointestinal Endoscopy, Okayama University Hospital, Okayama, Japan
| | - Hiroyuki Yanai
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
13
|
Sukri A, Hanafiah A, Kosai NR. The Roles of Immune Cells in Gastric Cancer: Anti-Cancer or Pro-Cancer? Cancers (Basel) 2022; 14:cancers14163922. [PMID: 36010915 PMCID: PMC9406374 DOI: 10.3390/cancers14163922] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Gastric cancer is still one of the leading causes of death caused by cancer in developing countries. The emerging role of immunotherapy in cancer treatment has led to more research to elucidate the roles of essential immune cells in gastric cancer prognosis. We reviewed the roles of immune cells including T cells, B cells, dendritic cells, macrophages and natural killer cells in gastric cancer. Although the studies conducted on the roles of immune cells in gastric cancer pathogenesis produced conflicting results, understanding the roles of immune cells in gastric cancer will help us to harness them for application in immunotherapy for better prognosis and management of gastric cancer patients. Abstract Despite the fact that the incidence of gastric cancer has declined over the last decade, it is still the world’s leading cause of cancer-related death. The diagnosis of early gastric cancer is difficult, as symptoms of this cancer only manifest at a late stage of cancer progression. Thus, the prognosis of gastric cancer is poor, and the current treatment for improving patients’ outcomes involves the application of surgery and chemotherapy. Immunotherapy is one of the most recent therapies for gastric cancer, whereby the immune system of the host is programmed to combat cancer cells, and the therapy differs based upon the patient’s immune system. However, an understanding of the role of immune cells, namely the cell-mediated immune response and the humoral immune response, is pertinent for applications of immunotherapy. The roles of immune cells in the prognosis of gastric cancer have yielded conflicting results. This review discusses the roles of immune cells in gastric cancer pathogenesis, specifically, T cells, B cells, macrophages, natural killer cells, and dendritic cells, as well as the evidence presented thus far. Understanding how cancer cells interact with immune cells is of paramount importance in designing treatment options for gastric cancer immunotherapy.
Collapse
Affiliation(s)
- Asif Sukri
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Shah Alam 43200, Malaysia
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
- Correspondence:
| | - Nik Ritza Kosai
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
14
|
Sun H, He T, Wu Y, Yuan H, Ning J, Zhang Z, Deng X, Li B, Wu C. Cytotoxin-Associated Gene A-Negative Helicobacter pylori Promotes Gastric Mucosal CX3CR1+CD4+ Effector Memory T Cell Recruitment in Mice. Front Microbiol 2022; 13:813774. [PMID: 35154057 PMCID: PMC8829513 DOI: 10.3389/fmicb.2022.813774] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background Helicobacter pylori can cause many kinds of gastric disorders, ranging from gastritis to gastric cancer. Cytotoxin-associated gene A (CagA)+H. pylori is more likely to cause gastric histopathologic damage than CagA–H. pylori. However, the underlying mechanism needs to be further investigated. Materials and methods Mice were intragastrically administered equal amounts of CagA+ or CagA–H. pylori. Four weeks later, 24 chemokines in stomachs were measured using a mouse chemokine array, and the phenotypes of the recruited gastric CD4+ T cells were analyzed. The migration pathway was evaluated. Finally, the correlation between each pair among the recruited CD4+ T cell sub-population, H. pylori colonization level, and histopathologic damage score were determined by Pearson correlation analysis. Results The concentration of chemokines, CCL3 and CX3CL1, were significantly elevated in CagA–H. pylori-infected gastric mucosa than in CagA+H. pylori-infected gastric mucosa. Among them, CX3CL1 secreted by gastric epithelial cells, which was elicited more effectively by CagA–H. pylori than by the CagA+ strain, dramatically promoted mucosal CD4+ T cell migration. The expression of CX3CR1, the only known receptor of CX3CL1, was upregulated on the surface of gastric CD4+ T cells in CagA–H. pylori-infected stomach. In addition, most of the CX3CR1-positive gastric CD4+ T cells were CD44+CD69–CCR7– effector memory T cells (Tem). Pearson correlation analysis showed that the recruited CX3CR1+CD4+ Tem cell population was negatively correlated with H. pylori colonization level and histopathologic damage score. Conclusion CagA–H. pylori promotes gastric mucosal CX3CR1+CD4+ Tem recruitment in mice.
Collapse
Affiliation(s)
- Heqiang Sun
- Department of Laboratory Medicine, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Taojun He
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yanan Wu
- Peking University People’s Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Hanmei Yuan
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jie Ning
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhenhua Zhang
- Department of Gastroenterology of the 305 Hospital of Chinese People’s Liberation Army, Beijing, China
| | - Xinli Deng
- Department of Laboratory Medicine, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Bin Li
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Bin Li,
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Chao Wu,
| |
Collapse
|
15
|
Ahmed AAQ, Qi F, Zheng R, Xiao L, Abdalla AME, Mao L, Bakadia BM, Liu L, Atta OM, Li X, Shi Z, Yang G. The impact of ExHp-CD (outer membrane vesicles) released from Helicobacter pylori SS1 on macrophage RAW 264.7 cells and their immunogenic potential. Life Sci 2021; 279:119644. [PMID: 34048813 DOI: 10.1016/j.lfs.2021.119644] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/02/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
Bacterial-derived extracellular vesicles could play a major role in attenuating and treating diseases. They play a major anti-infection role by modulating immune responses against pathogens and preventing infection by inhibiting pathogen localization and proliferation. In this study, outer membrane vesicles (ExHp-CD) released by Helicobacter pylori SS1 (H. pylori) and total antigens isolated from H. pylori SS1 (AgHp) were evaluated for their immunogenic potential and their effect on macrophage RAW 264.7 cells. Results demonstrated that both ExHp-CD and AgHp induced T helper 2 (Th2) immune response, which was reported to be important in immune protection against H. pylori infections. Both ExHp-CD and AgHp produced high levels of IL-10 and IL-4, while no significant levels of IL-12 p70 or IFN-γ were detected. However, ExHp-CD showed a better effect on macrophage RAW 264.7 cells compared to AgHp. Macrophage RAW 264.7 cells stimulated with 5, and 10 μg/mL of ExHp-CD showed an increased ratio of CD206 (M2 phenotype marker) and a decreased ratio of CD86 (M1 phenotype marker). Moreover, results suggested that the immunogenic effect that ExHp-CD possesses was attributed to their cargo of Epimerase_2 domain-containing protein (Epi_2D), Probable malate:quinone oxidoreductase (Pro_mqo), and Probable cytosol aminopeptidase (Pro_ca). Results demonstrated that ExHp-CD possesses an immunological activity to induce Th2 immune response against H. pylori infection with results comparable to AgHp. However, ExHp-CD showed higher efficacy regarding safety, biocompatibility, lack of toxicity, and hemocompatibility. Thus, it could serve as an immunogenic candidate with more desired characteristics.
Collapse
Affiliation(s)
- Abeer Ahmed Qaed Ahmed
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Fuyu Qi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Ruizhu Zheng
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Lin Xiao
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, China.
| | - Ahmed M E Abdalla
- Department of Biochemistry, College of Applied Science, University of Bahri, Khartoum 1660/11111, Sudan
| | - Lin Mao
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Bianza Moise Bakadia
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Li Liu
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Omar Mohammad Atta
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Xiaohong Li
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Zhijun Shi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China.
| |
Collapse
|
16
|
Chen X, Wang R, Bao C, Zhang J, Zhang J, Li R, Wu S, Wen J, Yang T, Wei S, Li H, Wei Y, Ren S, Zhao Y. Palmatine ameliorates Helicobacter pylori-induced chronic atrophic gastritis by inhibiting MMP-10 through ADAM17/EGFR. Eur J Pharmacol 2020; 882:173267. [PMID: 32569674 DOI: 10.1016/j.ejphar.2020.173267] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 01/15/2023]
Abstract
Palmatine (Pal), a plant-based isoquinoline alkaloid, was initially isolated from Coptidis Rhizoma (CR, Huanglian in Chinese) and considered to be a potential non-antibiotic therapeutic agent that can safely and effectively improve Helicobacter pylori (H. pylori) induced chronic atrophic gastritis (CAG). However, underlying mechanisms are unclear. In this study, we explored the protective effect of Pal on H. pylori induced CAG in vivo and in vitro. As a result, Pal alleviated the histological damage of gastric mucosa and the morphological changes of gastric epithelial cell (GES-1) caused by H. pylori. Furthermore, Pal significantly inhibited the expression of EGFR-activated ligand genes, including a disintegrin and metalloproteinase 17 (ADAM17) and heparin-binding epidermal growth factor-like growth factor (HB-EGF), and the proinflammatory factors, such as chemokine 16 (CXCL-16) and interleukin 8 (IL-8), were suppressed. In addition, Pal attenuated inflammatory infiltration of CD8+ T cells while promoted Reg3a expression to enhance host defense. Taken together, we concluded that Pal attenuated the MMP-10 dependent inflammatory response in the gastric mucosa by blocking ADAM17/EGFR signaling, which contributed to its gastrointestinal protective effect.
Collapse
Affiliation(s)
- Xing Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Ruilin Wang
- Integrative Medical Center, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Chunmei Bao
- Division of Clinical Microbiology, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Jianzhong Zhang
- Center of Disease Control and Prevention, National Institute for Communicable Disease Control and Prevention, Beijing, 102206, China
| | - Juling Zhang
- Division of Clinical Microbiology, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Ruisheng Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Shihua Wu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Jianxian Wen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Tao Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Shizhang Wei
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Haotian Li
- Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Ying Wei
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Sichen Ren
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Yanling Zhao
- Department of Pharmacy, Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
17
|
El-Zaatari M, Bishu S, Zhang M, Grasberger H, Hou G, Haley H, Humphries B, Syu LJ, Dlugosz AA, Luker K, Luker GD, Eaton K, Kamada N, Cascalho M, Kao JY. Aim2-mediated/IFN-β-independent regulation of gastric metaplastic lesions via CD8+ T cells. JCI Insight 2020; 5:94035. [PMID: 32053518 DOI: 10.1172/jci.insight.94035] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 02/12/2020] [Indexed: 01/24/2023] Open
Abstract
Development of gastric cancer is often preceded by chronic inflammation, but the immune cellular mechanisms underlying this process are unclear. Here we demonstrated that an inflammasome molecule, absent in melanoma 2 (Aim2), was upregulated in patients with gastric cancer and in spasmolytic polypeptide-expressing metaplasia of chronically Helicobacter felis-infected stomachs in mice. However, we found that Aim2 was not necessary for inflammasome function during gastritis. In contrast, Aim2 deficiency led to an increase in gastric CD8+ T cell frequency, which exacerbated metaplasia. These gastric CD8+ T cells from Aim2-/- mice were found to have lost their homing receptor expression (sphingosine-1-phosphate receptor 1 [S1PR1] and CD62L), a feature of tissue-resident memory T cells. The process was not mediated by Aim2-dependent regulation of IFN-β or by dendritic cell-intrinsic Aim2. Rather, Aim2 deficiency contributed to an increased production of CXCL16 by B cells, which could suppress S1PR1 and CD62L in CD8+ T cells. This study describes a potentially novel function of Aim2 that regulates CD8+ T cell infiltration and retention within chronically inflamed solid organ tissue. This function operates independent of the inflammasome, IFN-β, or dendritic cells. We provide evidence that B cells can contribute to this mechanism via CXCL16.
Collapse
Affiliation(s)
- Mohamad El-Zaatari
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Shrinivas Bishu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Min Zhang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Helmut Grasberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Guoqing Hou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Henry Haley
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Brock Humphries
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Li-Jyun Syu
- Department of Dermatology, School of Medicine
| | | | - Kathy Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | - Gary D Luker
- Department of Radiology, Microbiology and Immunology, and Biomedical Engineering
| | | | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - John Y Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine
| |
Collapse
|
18
|
Gonciarz W, Krupa A, Hinc K, Obuchowski M, Moran AP, Gajewski A, Chmiela M. The effect of Helicobacter pylori infection and different H. pylori components on the proliferation and apoptosis of gastric epithelial cells and fibroblasts. PLoS One 2019; 14:e0220636. [PMID: 31390383 PMCID: PMC6685636 DOI: 10.1371/journal.pone.0220636] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/19/2019] [Indexed: 12/16/2022] Open
Abstract
Background Helicobacter pylori colonizes the human gastric mucosa, causing chronic inflammation, peptic ulcers and gastric cancer. A cascade of harmful processes results from the interaction of these bacteria with the gastric epithelium. Aim To investigate these processes in terms of upregulation of oxidative stress and cell apoptosis and downregulation of the pro-regenerative activity of cells. Methods We employed an in vivo guinea pig model at 7 or 28 days postinoculation with H. pylori, corresponding to an acute or chronic stage of infection, respectively, and an in vitro model of guinea pig primary gastric epithelial cells and fibroblasts treated with bacterial components: glycine acid extract (GE), urease subunit A (UreA), cytotoxin-associated gene A protein (CagA) and lipopolysaccharide (LPS). Cells were evaluated for metabolic activity (MTT reduction), myeloperoxidase (MPO) and metalloproteinase (MMP-9) secretion, lipid peroxidation (4-hydroxynonenal (4HNE)), migration (wound healing), proliferation (Ki-67 antigen) and cell apoptosis (TUNEL assay; Bcl-xL, Bax, Bcl-2 expression; caspase 3 cleavage). Results Significant infiltration of the gastric mucosa by inflammatory cells in vivo in response to H. pylori was accompanied by oxidative stress and cell apoptosis, which were more intense 7 than 28 days after inoculation. The increase in cell proliferation was more intense in chronic than acute infection. H. pylori components GE, CagA, UreA, and LPS upregulated oxidative stress and apoptosis. Only H. pylori LPS inhibited cell migration and proliferation, which was accompanied by the upregulation of MMP-9. Conclusions H. pylori infection induces cell apoptosis in conjunction with increased oxidative stress. Elevated apoptosis protects against deleterious inflammation and neoplasia; however, it reduces cell integrity. Upregulation of cell migration and proliferation in response to injury in the milieu of GE, CagA or UreA facilitates tissue regeneration but increases the risk of neoplasia. By comparison, downregulation of cell regeneration by H. pylori LPS may promote chronic inflammation.
Collapse
Affiliation(s)
- Weronika Gonciarz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Łodz, Poland
| | - Agnieszka Krupa
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Łodz, Poland
| | - Krzysztof Hinc
- Laboratory of Molecular Bacteriology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdańsk, Gdansk, Poland
| | - Michał Obuchowski
- Laboratory of Molecular Bacteriology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdańsk, Gdansk, Poland
| | - Anthony P Moran
- Department of Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Adrian Gajewski
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Łodz, Poland
| | - Magdalena Chmiela
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Łodz, Poland
- * E-mail:
| |
Collapse
|
19
|
Lv YP, Cheng P, Zhang JY, Mao FY, Teng YS, Liu YG, Kong H, Wu XL, Hao CJ, Han B, Ma Q, Yang SM, Chen W, Peng LS, Wang TT, Zou QM, Zhuang Y. Helicobacter pylori-induced matrix metallopeptidase-10 promotes gastric bacterial colonization and gastritis. SCIENCE ADVANCES 2019; 5:eaau6547. [PMID: 30949574 PMCID: PMC6447374 DOI: 10.1126/sciadv.aau6547] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 02/08/2019] [Indexed: 05/31/2023]
Abstract
The interaction between gastric epithelium and immune response plays key roles in H. pylori-associated pathology. We demonstrated a procolonization and proinflammation role of MMP-10 in H. pylori infection. MMP-10 is elevated in gastric mucosa and is produced by gastric epithelial cells synergistically induced by H. pylori and IL-22 via the ERK pathway. Human gastric MMP-10 was correlated with H. pylori colonization and the severity of gastritis, and mouse MMP-10 from non-BM-derived cells promoted bacteria colonization and inflammation. H. pylori colonization and inflammation were attenuated in IL-22-/-, MMP-10-/-, and IL-22-/-MMP-10-/- mice. MMP-10-associated inflammation is characterized by the influx of CD8+ T cells, whose migration is induced via MMP-10-CXCL16 axis by gastric epithelial cells. Under the influence of MMP-10, Reg3a, E-cadherin, and zonula occludens-1 proteins decrease, resulting in impaired host defense and increased H. pylori colonization. Our results suggest that MMP-10 facilitates H. pylori persistence and promotes gastritis.
Collapse
Affiliation(s)
- Yi-pin Lv
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jin-yu Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Fang-yuan Mao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yong-sheng Teng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yu-gang Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hui Kong
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao-long Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chuan-jie Hao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Bin Han
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qiang Ma
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Shi-ming Yang
- Department of Gastroenterology, XinQiao Hospital, Third Military Medical University, Chongqing, China
| | - Weisan Chen
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria 3085, Australia
| | - Liu-sheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ting-ting Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Quan-ming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yuan Zhuang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
20
|
Immunization with Heat Shock Protein A and γ-Glutamyl Transpeptidase Induces Reduction on the Helicobacter pylori Colonization in Mice. PLoS One 2015; 10:e0130391. [PMID: 26102080 PMCID: PMC4478016 DOI: 10.1371/journal.pone.0130391] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/19/2015] [Indexed: 12/11/2022] Open
Abstract
The human gastric pathogen Helicobacter pylori (H. pylori) is a successful colonizer of the stomach. H. pylori infection strongly correlates with the development and progression of chronic gastritis, peptic ulcer disease, and gastric malignances. Vaccination is a promising strategy for preventing H. pylori infection. In this study, we evaluated the candidate antigens heat shock protein A (HspA) and H. pylori γ-glutamyl transpeptidase (GGT) for their effectiveness in development of subunit vaccines against H. pylori infection. rHspA, rGGT, and rHspA-GGT, a fusion protein based on HspA and GGT, were constructed and separately expressed in Escherichia coli and purified. Mice were then immunized intranasally with these proteins, with or without adjuvant. Immunized mice exhibited reduced bacterial colonization in stomach. The highest reduction in bacterial colonization was seen in mice immunized with the fusion protein rHspA-GGT when paired with the mucosal adjuvant LTB. Protection against H. pylori colonization was mediated by a strong systemic and localized humoral immune response, as well as a balanced Th1/Th2 cytokine response. In addition, immunofluorescence microscopy confirmed that rHspA-GGT specific rabbit antibodies were able to directly bind H. pylori in vitro. These results suggest antibodies are essential to the protective immunity associated with rHspA-GGT immunization. In summary, our results suggest HspA and GGT are promising vaccine candidates for protection against H. pylori infection.
Collapse
|
21
|
Kronsteiner B, Bassaganya-Riera J, Philipson N, Hontecillas R. Novel insights on the role of CD8+ T cells and cytotoxic responses during Helicobacter pylori infection. Gut Microbes 2014; 5:357-62. [PMID: 24755940 PMCID: PMC4153774 DOI: 10.4161/gmic.28899] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori chronically persists in 50% of the human population and causes serious gastric and duodenal pathologies in 15% of infected people. Research on the immune response to the infection has mainly focused on the induction of CD4+ T cell responses. Human studies emphasize the potential clinical relevance of CD8+ cytotoxic T lymphocytes, however this cell type has barely been reported in studies employing mouse or gerbil models. Traditionally characterized as an extracellular bacterium, H. pylori has been identified inside epithelial and immune cells. Similarly to other intracellular bacteria, H. pylori infection of macrophages can alter autophagy and phagosome processing. A novel animal model of H. pylori infection demonstrates for the first time the induction of cytotoxic CD8+ T cell responses in pigs and localization of intracellular H. pylori within lymphoid aggregates. Here, we discuss novel mechanisms of host-H. pylori interactions that could lead to the induction of cytotoxic responses.
Collapse
|
22
|
Helicobacter pylori infection in a pig model is dominated by Th1 and cytotoxic CD8+ T cell responses. Infect Immun 2013; 81:3803-13. [PMID: 23897614 DOI: 10.1128/iai.00660-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori infection is the leading cause for peptic ulcer disease and gastric adenocarcinoma. Mucosal T cell responses play an important role in mediating H. pylori-related gastric immunopathology. While induced regulatory T (iTreg) cells are required for chronic colonization without disease, T helper 1 (Th1) effector responses are associated with lower bacterial loads at the expense of gastric pathology. Pigs were inoculated with either H. pylori strain SS1 or J99. Phenotypic and functional changes in peripheral blood mononuclear cell (PBMC) populations were monitored weekly, and mucosal immune responses and bacterial loads were assessed up to 2 months postinfection. Both H. pylori strains elicited a Th1 response characterized by increased percentages of CD4(+)Tbet(+) cells and elevated gamma interferon (IFN-γ) mRNA in PBMCs. A subset of CD8(+) T cells expressing Tbet and CD16 increased following infection. Moreover, a significant increase in perforin and granzyme mRNA expression was observed in PBMCs of infected pigs, indicating a predominant cytotoxic immune response. Infiltration of B cells, myeloid cells, T cells expressing Treg- and Th17-associated transcription factors, and cytotoxic T cells was found in the gastric lamina propria of both infected groups. Interestingly, based on bacterial reisolation data, strain SS1 showed greater capacity to colonize and/or persist in the gastric mucosa than did strain J99. This novel pig model of infection closely mimics human gastric pathology and presents a suitable avenue for studying effector and regulatory responses toward H. pylori described in humans.
Collapse
|
23
|
Yang WC, Chen L, Li HB, Li B, Hu J, Zhang JY, Yang SM, Zou QM, Guo H, Wu C. Identification of two novel immunodominant UreB CD4(+) T cell epitopes in Helicobacter pylori infected subjects. Vaccine 2013; 31:1204-9. [PMID: 23306364 DOI: 10.1016/j.vaccine.2012.12.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 12/09/2012] [Accepted: 12/21/2012] [Indexed: 01/14/2023]
Abstract
An epitope-based vaccine is a promising option for treating Helicobacter pylori (H. pylori) infection. Epitope mapping is the first step in designing an epitope-based vaccine. A pivotal role of CD4(+) T cells in protection against H. pylori has been accepted, but few Th epitopes have been identified. In this study, two novel UreB CD4(+) T cell epitopes were identified using PBMCs obtained from two H. pylori infected subjects. We determined the restriction molecules by antibody blocking and used various Epstein-Barr virus-transformed B lymphocyte cell lines (BLCLs) with different HLA alleles as APCs to present peptides to CD4(+) T cells. These epitopes were DRB1*1404-restricted UreB(373-385) and DRB1*0803-restricted UreB(438-452). The T cells specific to these epitopes not only recognized autologous DCs loaded with recombinant UreB but also those pulsed with H. pylori whole cell lysates, suggesting that these epitope peptides are naturally processed. These epitopes have important value for designing an effective H. pylori vaccine.
Collapse
Affiliation(s)
- Wu-Chen Yang
- Department of Clinical Microbiology and Immunology, College of Medical Laboratory Science, Third Military Medical University, Chongqing 400038, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Martínez-Becerra F, Castillo-Rojas G, Ponce de León S, López-Vidal Y. IgG subclasses against Helicobacter pylori isolates: an important tool for disease characterization. Scand J Immunol 2012; 76:26-32. [PMID: 22686508 DOI: 10.1111/j.1365-3083.2012.02699.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Helicobacter pylori infects around 50% of the world's population and is associated with diverse pathologies. In the most severe cases, the bacterium causes peptic ulcers and gastric cancer. The interplay between H. pylori and the host's immune response may help to determine the specific outcome of the infection. To study the relationship between antibody subclasses and variation in immune recognition, we determined the immunoglobulin G1 and 2 (IgG1 and IgG2) titres of sera obtained from patients with different H. pylori-associated pathologies. IgG1 and IgG2 titres were determined by ELISA in 44 sera of patients with different H. pylori-associated diseases (peptic ulcer, bleeding peptic ulcers, gastric cancer and dyspepsia). Soluble proteins from lysates were obtained from 12 different clinical isolates from similar associated diseases. We found that soluble proteins from lysates of H. pylori strains (SPLHP) recognition patterns in these sera were highly variable. Overall, IgG2 titres were higher than the IgG1 titres in the infected patients. In particular, those with peptic ulcers showed marked elevation in IgG2/IgG1 ratios, while SPLHPs from dyspeptic patients resulted in high IgG1 titres. Our results reveal that correlation of antibody subclass titres with Th1/Th2 markers may aid pathology characterization and show a potential diagnosis that could be formally evaluated in other studies.
Collapse
Affiliation(s)
- F Martínez-Becerra
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | | |
Collapse
|
25
|
Toller IM, Altmeyer M, Kohler E, Hottiger MO, Müller A. Inhibition of ADP ribosylation prevents and cures helicobacter-induced gastric preneoplasia. Cancer Res 2010; 70:5912-22. [PMID: 20634404 DOI: 10.1158/0008-5472.can-10-0528] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gastric adenocarcinoma develops as a consequence of chronic inflammation of the stomach lining that is caused by persistent infection with the bacterium Helicobacter pylori. Gastric carcinogenesis progresses through a sequence of preneoplastic lesions that manifest histologically as atrophic gastritis, intestinal metaplasia, and dysplasia. We show here in several preclinical models of Helicobacter-induced atrophic gastritis, epithelial hyperplasia, and metaplasia that the inhibition of ADP ribosylation by the small-molecule inhibitor PJ34 not only prevents the formation of gastric cancer precursor lesions, but also efficiently reverses preexisting lesions. PJ34 exerts its chemopreventive and therapeutic effects by impairing Helicobacter-specific T-cell priming and T(H)1 polarization in the gut-draining mesenteric lymph nodes. The subsequent infiltration of pathogenic T cells into the gastric mucosa and the ensuing gastric T cell-driven immunopathology are prevented efficiently by PJ34. Our data indicate that PJ34 directly suppresses T-cell effector functions by blocking the IFN-gamma production of mesenteric lymph node T cells ex vivo. Upon exposure to PJ34, purified T cells failed to synthesize ADP-ribose polymers and to activate the transcription of genes encoding IFN-gamma, interleukin 2, and the interleukin 2 receptor alpha chain in response to stimuli such as CD3/CD28 cross-linking or phorbol 12-myristate 13-acetate/ionomycin. The immunosuppressive and chemoprotective effects of PJ34 therefore result from impaired T-cell activation and T(H)1 polarization, and lead to the protection from preneoplastic gastric immunopathology. In conclusion, ADP-ribosylating enzymes constitute novel targets for the treatment of Helicobacter-associated gastric lesions predisposing infected individuals to gastric cancer and may also hold promise for the treatment of other T cell-driven chronic inflammatory conditions and autoimmune pathologies.
Collapse
Affiliation(s)
- Isabella M Toller
- Institute of Molecular Cancer Research and Institute of Veterinary Biochemistry and Molecular Biology, University of Zürich, Zürich, Switzerland
| | | | | | | | | |
Collapse
|
26
|
Wang YH, Gorvel JP, Chu YT, Wu JJ, Lei HY. Helicobacter pylori impairs murine dendritic cell responses to infection. PLoS One 2010; 5:e10844. [PMID: 20523725 PMCID: PMC2877707 DOI: 10.1371/journal.pone.0010844] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 04/30/2010] [Indexed: 12/16/2022] Open
Abstract
Background Helicobacter pylori, a human pathogen associated with chronic gastritis, peptic ulcer and gastric malignancies, is generally viewed as an extracellular microorganism. Here, we show that H. pylori replicates in murine bone marrow derived-dendritic cells (BMDCs) within autophagosomes. Methodology/Principal Findings A 10-fold increase of CFU is found between 2 h and 6 h p.i. in H. pylori-infected BMDCs. Autophagy is induced around the bacterium and participates at late time points of infection for the clearance of intracellular H. pylori. As a consequence of infection, LC3, LAMP1 and MHC class II molecules are retained within the H. pylori-containing vacuoles and export of MHC class II molecules to cell surface is blocked. However, formalin-fixed H. pylori still maintain this inhibitory activity in BMDC derived from wild type mice, but not in from either TLR4 or TLR2-deficient mice, suggesting the involvement of H. pylori-LPS in this process. TNF-alpha, IL-6 and IL-10 expression was also modulated upon infection showing a TLR2-specific dependent IL-10 secretion. No IL-12 was detected favoring the hypothesis of a down modulation of DC functions during H. pylori infection. Furthermore, antigen-specific T cells proliferation was also impaired upon infection. Conclusions/Significance H. pylori can infect and replicate in BMDCs and thereby affects DC-mediated immune responses. The implication of this new finding is discussed for the biological life cycle of H. pylori in the host.
Collapse
Affiliation(s)
- Ya-Hui Wang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Jean-Pierre Gorvel
- Aix Marseille Université, Faculté des Sciences de Luminy, Centre d'Immunologie de Marseille-Luminy (CIML), UMR6546, Marseille, France
- Inserm, U631, Marseille, France
- CNRS, UMR6102, Marseille, France
| | - Yen-Ting Chu
- Departments of Microbiology and Immunology, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Jiunn-Jong Wu
- Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Huan-Yao Lei
- Departments of Microbiology and Immunology, National Cheng Kung University, Tainan, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
27
|
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: 133] [Impact Index Per Article: 8.9] [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
|