1
|
Talayev V, Svetlova M, Zaichenko I, Voronina E, Babaykina O, Neumoina N, Perfilova K. CCR6 + T helper cells and regulatory T cells in the blood and gastric mucosa during Helicobacter pylori infection. Helicobacter 2024; 29:e13097. [PMID: 38819071 DOI: 10.1111/hel.13097] [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: 01/23/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Helicobacter pylori (H. pylori) can evade the host's immune response and persist for a long time on the gastric mucosa. T helper (Th) cells appear to be involved in the control of H. pylori bacteria but promote mucosal inflammation. In contrast, regulatory T cells (Tregs) may reduce inflammation but promote H. pylori persistence. CC motif chemokine receptor 6 (CCR6) is involved in the migration of various cells into inflamed gastric mucosa. In this study, we examined CCR6+ Th cells and CCR6+ Tregs during H. pylori infection in humans. MATERIALS AND METHODS Isolation of cells from blood and mucosal biopsies, magnetic separation of В cells, CD4+ and CD4+CCR6+CD45RO+ T cells, antigen-specific activation, B cell response in vitro, flow cytometry, determination of CD4+CD25hiFoxP3+ Tregs and various groups of Th cells. RESULTS CD4+CCR6+ blood lymphocytes from healthy donors included Th cells and Tregs. These CCR6+ Th cells produced proinflammatory cytokines and also stimulated plasma cell maturation and antibody production in vitro. H. pylori gastritis and peptic ulcer disease were associated with an increase in the number of circulate CD4+CCR6+CD45RO+ cells and the percentage of Th1, Th17 and Th1/17 cells in this lymphocyte subgroup. In H. pylori-positive patients, circulating CD4+CCR6+ cells contained a higher proportion of H. pylori-specific cells compared with their CD4+CCR6- counterparts. H. pylori infection strongly increased the content of CD4+ lymphocytes in the inflamed gastric mucosa, with the majority of these CD4+ lymphocytes expressing CCR6. CD4+CCR6+ lymphocytes from H. pylori-infected stomach included Tregs and in vivo activated T cells, some of which produced interferon-γ without ex vivo stimulation. CONCLUSION H. pylori infection causes an increase in the number of mature CD4+CCR6+ lymphocytes in the blood, with a pro-inflammatory shift in their composition and enrichment of the gastric mucosa with CD4+CCR6+ lymphocytes, including CCR6+ Th1 cells and Tregs.
Collapse
Affiliation(s)
- Vladimir Talayev
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor, Nizhny Novgorod, Russia
| | - Maria Svetlova
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor, Nizhny Novgorod, Russia
| | - Irina Zaichenko
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor, Nizhny Novgorod, Russia
| | - Elena Voronina
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor, Nizhny Novgorod, Russia
| | - Olga Babaykina
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor, Nizhny Novgorod, Russia
| | - Natalia Neumoina
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor, Nizhny Novgorod, Russia
| | - Ksenia Perfilova
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor, Nizhny Novgorod, Russia
| |
Collapse
|
2
|
Domínguez-Martínez DA, Fontes-Lemus JI, García-Regalado A, Juárez-Flores Á, Fuentes-Pananá EM. IL-8 Secreted by Gastric Epithelial Cells Infected with Helicobacter pylori CagA Positive Strains Is a Chemoattractant for Epstein-Barr Virus Infected B Lymphocytes. Viruses 2023; 15:651. [PMID: 36992360 PMCID: PMC10054738 DOI: 10.3390/v15030651] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Helicobacter pylori and EBV are considered the main risk factors in developing gastric cancer. Both pathogens establish life-lasting infections and both are considered carcinogenic in humans. Different lines of evidence support that both pathogens cooperate to damage the gastric mucosa. Helicobacter pylori CagA positive virulent strains induce the gastric epithelial cells to secrete IL-8, which is a potent chemoattractant for neutrophils and one of the most important chemokines for the bacterium-induced chronic gastric inflammation. EBV is a lymphotropic virus that persists in memory B cells. The mechanism by which EBV reaches, infects and persists in the gastric epithelium is not presently understood. In this study, we assessed whether Helicobacter pylori infection would facilitate the chemoattraction of EBV-infected B lymphocytes. We identified IL-8 as a powerful chemoattractant for EBV-infected B lymphocytes, and CXCR2 as the main IL-8 receptor whose expression is induced by the EBV in infected B lymphocytes. The inhibition of expression and/or function of IL-8 and CXCR2 reduced the ERK1/2 and p38 MAPK signaling and the chemoattraction of EBV-infected B lymphocytes. We propose that IL-8 at least partially explains the arrival of EBV-infected B lymphocytes to the gastric mucosa, and that this illustrates a mechanism of interaction between Helicobacter pylori and EBV.
Collapse
Affiliation(s)
- Diana A. Domínguez-Martínez
- Research Unit on Virology and Cancer, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| | - José I. Fontes-Lemus
- Research Unit on Virology and Cancer, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| | - Alejandro García-Regalado
- Research Unit on Virology and Cancer, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| | - Ángel Juárez-Flores
- Research Unit on Virology and Cancer, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico
| | - Ezequiel M. Fuentes-Pananá
- Research Unit on Virology and Cancer, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
3
|
Sharafutdinov I, Ekici A, Vieth M, Backert S, Linz B. Early and late genome-wide gastric epithelial transcriptome response during infection with the human carcinogen Helicobacterpylori. CELL INSIGHT 2022; 1:100032. [PMID: 37193047 PMCID: PMC10120309 DOI: 10.1016/j.cellin.2022.100032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 05/18/2023]
Abstract
Infection of the stomach by Helicobacter pylori is a major risk factor for the development of gastric cancer. Colonization of the gastric epithelium leads to the activation of multiple disease-related signaling pathways. Serine protease HtrA represents an important secreted virulence factor that mediates cleavage of cellular junctions. However, its potential role in nuclear responses is unknown. Here, we performed a genome-wide RNA-seq analysis of polarized gastric epithelial cells infected by wild-type (wt) and ΔhtrA mutant bacteria. Fluorescence microscopy showed that H. pylori wt, but not ΔhtrA bacteria, preferably localized at cellular junctions. Our results pinpointed early (2 h) and late (6 h) transcriptional responses, with most differentially expressed genes at 6 h post infection. The transcriptomes revealed HtrA-dependent targeting of genes associated with inflammation and apoptosis (e.g. IL8, ZFP36, TNF). Accordingly, infection with the ΔhtrA mutant induced increased apoptosis rates in host cells, which was associated with reduced H. pylori CagA expression. In contrast, transcription of various carcinogenesis-associated genes (e.g. DKK1, DOCK8) was affected by H. pylori independent of HtrA. These findings suggest that H. pylori disturbs previously unknown molecular pathways in an HtrA-dependent and HtrA-independent manner, and provide valuable new insights of this significant pathogen in humans and thus potential targets for better controlling the risk of malignant transformation.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, Friedrich Alexander Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058, Erlangen, Germany
| | - Arif Ekici
- Institute of Human Genetics, University Hospital, Friedrich Alexander Universität Erlangen-Nürnberg, Schwabachanlage 10, D-91054, Erlangen, Germany
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Klinikum Bayreuth, Preuschwitzer Str 101, D-95445, Bayreuth, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich Alexander Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058, Erlangen, Germany
| | - Bodo Linz
- Department of Biology, Division of Microbiology, Friedrich Alexander Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058, Erlangen, Germany
| |
Collapse
|
4
|
Korona B, Korona D, Zhao W, Wotherspoon AC, Du MQ. CCR6 activation links innate immune responses to mucosa-associated lymphoid tissue lymphoma development. Haematologica 2022; 107:1384-1396. [PMID: 35142152 PMCID: PMC9152962 DOI: 10.3324/haematol.2021.280067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
The genesis of extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue (MALT) is driven by oncogenic co-operation among immunological stimulations and acquired genetic changes. We previously identified recurrent CCR6 mutations in MALT lymphoma, with majority predicted to result in truncated proteins lacking the phosphorylation motif important for receptor desensitization. Functional consequences of these mutational changes, the molecular mechanisms of CCR6 activation and how this receptor signaling contributes to MALT lymphoma development remain to be investigated. In the present study, we demonstrated that these mutations impaired CCR6 receptor internalization and were activating changes, being more potent in apoptosis resistance, malignant transformation, migration and intracellular signaling, particularly in the presence of the ligands CCL20, HBD2 (human b defensin 2) and HD5 (human a defensin 5). CCR6 was highly expressed in malignant B cells irrespective of the lymphoma sites. HBD2 and CCL20 were constitutively expressed by the duct epithelial cells of salivary glands, and also those involved in lymphoepithelial lesions (LEL) in salivary gland MALT lymphoma. While in the gastric setting, HBD2, and HD5, to a less extent CCL20, were highly expressed in epithelial cells of pyloric and intestinal metaplasia respectively including those involved in LEL, which are adaptive responses to chronic Helicobacter pylori infection. These findings suggest that CCR6 signaling is most likely active in MALT lymphoma, independent of its mutation status. The observations explain why the emergence of malignant B cells and their clonal expansion in MALT lymphoma are typically around LEL, linking the innate immune responses to lymphoma genesis.
Collapse
Affiliation(s)
- Boguslawa Korona
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge
| | - Dagmara Korona
- Department of Genetics, University of Cambridge, Cambridge
| | - Wanfeng Zhao
- The Human Research Tissue Bank, Cambridge University Hospitals NHS Foundation Trust, Cambridge
| | | | - Ming-Qing Du
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK; Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge.
| |
Collapse
|
5
|
Infiltration of Immunoinflammatory Cells and Related Chemokine/Interleukin Expression in Different Gastric Immune Microenvironments. J Immunol Res 2020; 2020:2450569. [PMID: 33426088 PMCID: PMC7774301 DOI: 10.1155/2020/2450569] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 01/23/2023] Open
Abstract
Gastric mucosal immune microenvironment plays an important role in the occurrence and development of diseases such as inflammation and cancer. In the present study, single-sample gene set enrichment analysis (ssGSEA) was used to evaluate the expression of cytokines and the degree of immune cell infiltration in four different gastric mucosa tissues from normal gastric mucosa, simple gastritis, and atrophic gastritis to gastric cancer. Here, we show the immune microenvironments of these four gastric mucosae were significantly different. From inflammation to gastric cancer, most immunoinflammatory cells showed a downward trend such as central memory CD4 T cell. Instead, several cells showed an upward trend such as macrophage. Additionally, we found some chemokines/interleukins were illustrated to be low expressed (or highly expressed) in precancerous stage and highly expressed (or low expressed) in postcancerous stage, which demonstrated an opposite expression characteristic in pre-/postcancerous stage.
Collapse
|
6
|
Shen Q, Polom K, Williams C, de Oliveira FMS, Guergova-Kuras M, Lisacek F, Karlsson NG, Roviello F, Kamali-Moghaddam M. A targeted proteomics approach reveals a serum protein signature as diagnostic biomarker for resectable gastric cancer. EBioMedicine 2019; 44:322-333. [PMID: 31151932 PMCID: PMC6606959 DOI: 10.1016/j.ebiom.2019.05.044] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/08/2019] [Accepted: 05/18/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is the third leading cause of cancer death. Early detection is a key factor to reduce its mortality. METHODS We retrospectively collected pre- and postoperative serum samples as well as tumour tissues and adjacent normal tissues from 100 GC patients. Serum samples from non-cancerous patients were served as controls (n = 50). A high-throughput protein detection technology, multiplex proximity extension assays (PEA), was applied to measure levels of over 300 proteins. Alteration of each protein was analysed by univariate analysis. Elastic-net logistic regression was performed to select serum proteins into the diagnostic model. FINDINGS We identified 19 serum proteins (CEACAM5, CA9, MSLN, CCL20, SCF, TGF-alpha, MMP-1, MMP-10, IGF-1, CDCP1, PPIA, DDAH-1, HMOX-1, FLI1, IL-7, ZBTB-17, APBB1IP, KAZALD-1, and ADAMTS-15) that together distinguish GC cases from controls with a diagnostic sensitivity of 93%, specificity of 100%, and area under receiver operating characteristic curve (AUC) of 0·99 (95% CI: 0·98-1). Moreover, the 19-serum protein signature provided an increased diagnostic capacity in patients at TNM I-II stage (sensitivity 89%, specificity 100%, AUC 0·99) and in patients with high microsatellite instability (MSI) (91%, 98%, and 0·99) compared to individual proteins. These promising results will inspire a large-scale independent cohort study to be pursued for validating the proposed protein signature. INTERPRETATION Based on targeted proteomics and elastic-net logistic regression, we identified a 19-serum protein signature which could contribute to clinical GC diagnosis, especially for patients at early stage and those with high MSI. FUND: This study was supported by a European H2020-Marie Skłodowska-Curie Innovative Training Networks grant (316,929, GastricGlycoExplorer). Funder had no influence on trial design, data evaluation, and interpretation.
Collapse
Affiliation(s)
- Qiujin Shen
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden.
| | - Karol Polom
- Department of General Surgery and Surgical Oncology, University of Siena, Italy; Department of Surgical Oncology, Gdansk Medical University, Gdansk, Poland.
| | | | | | | | - Frederique Lisacek
- Proteome Informatics Group, SIB Swiss Institute of Bioinformatics, Geneva, Switzerland; Computer Science Department and Section of Biology, University of Geneva, Switzerland.
| | - Niclas G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden.
| | - Franco Roviello
- Department of General Surgery and Surgical Oncology, University of Siena, Italy.
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden.
| |
Collapse
|
7
|
Mejías-Luque R, Zöller J, Anderl F, Loew-Gil E, Vieth M, Adler T, Engler DB, Urban S, Browning JL, Müller A, Gerhard M, Heikenwalder M. Lymphotoxin β receptor signalling executes Helicobacter pylori-driven gastric inflammation in a T4SS-dependent manner. Gut 2017; 66:1369-1381. [PMID: 27196595 DOI: 10.1136/gutjnl-2015-310783] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 03/14/2016] [Accepted: 03/21/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Lymphotoxin β receptor (LTβR) signalling has been implicated in inflammation-associated tumour development in different tissues. We have analysed the role of LTβR and alternative NF-κB signalling in Helicobacter pylori-mediated gastric inflammation and pathology. DESIGN We analysed several ligands and receptors of the alternative NF-κB pathway, RelB, p52 nuclear translocation and target genes in tissue samples of H. pylori-infected patients with different degrees of gastritis or early gastric tumours by in situ hybridisation, immunohistochemistry, Western blot and real-time PCR analyses. Molecular mechanisms involved in LTβR activation by H. pylori were assessed in vitro using human gastric cancer cell lines and distinct H. pylori isolates. The effects of blocking or agonistically activating LTβR on gastric pathology during challenge with a human pathogenic H. pylori strain were studied in a mouse model. RESULTS Among the tested candidates, LT was significantly increased and activated alternative NF-κB signalling was observed in the gastric mucosa of H. pylori-infected patients. H. pyloriinduced LTβR-ligand expression in a type IV secretion system-dependent but CagA-independent manner, resulting in activation of the alternative NF-κB pathway, which was further enhanced by blocking canonical NF-κB during infection. Blocking LTβR signalling in vivo suppressed H. pylori-driven gastritis, whereas LTβR activation in gastric epithelial cells of infected mice induced a broadened pro-inflammatory chemokine milieu, resulting in exacerbated pathology. CONCLUSIONS LTβR-triggered activation of alternative NF-κB signalling in gastric epithelial cells executes H. pylori-induced chronic gastritis, representing a novel target to restrict gastric inflammation and pathology elicited by H. pylori, while exclusively targeting canonical NF-κB may aggravate pathology by enhancing the alternative pathway.
Collapse
Affiliation(s)
- Raquel Mejías-Luque
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany.,German Centre for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Jessica Zöller
- Institut für Virologie, Technische Universität München, Helmholtz Zentrum München, Neuherberg, Germany
| | - Florian Anderl
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Elena Loew-Gil
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Michael Vieth
- Institut für Pathologie, Klinikum Bayreuth, Bayreuth, Germany
| | - Thure Adler
- Immunology Screen, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Daniela B Engler
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Sabine Urban
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | | | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Markus Gerhard
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany.,German Centre for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Mathias Heikenwalder
- Institut für Virologie, Technische Universität München, Helmholtz Zentrum München, Neuherberg, Germany.,Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
8
|
A Gain-Of-Function Mutation in the Plcg2 Gene Protects Mice from Helicobacter felis-Induced Gastric MALT Lymphoma. PLoS One 2016; 11:e0150411. [PMID: 26966907 PMCID: PMC4788355 DOI: 10.1371/journal.pone.0150411] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 02/13/2016] [Indexed: 01/04/2023] Open
Abstract
Gastric mucosa-associated lymphoid tissue (MALT) lymphomas develop from a chronic Helicobacter infection. Phospholipase C gamma 2 (PLCG2) is important for B-cell survival and proliferation. We used BALB/c mice with a gain-of-function mutation in the Plcg2 gene (Ali5) to analyze its role in the development of gastric MALT lymphoma. Heterozygous BALB/c Plcg2Ali5/+ and wildtype (WT) mice were infected with Helicobacter felis (H. felis) and observed up to 16 months for development of gastric MALT lymphomas. In contrast to our initial hypothesis, Plcg2Ali5/+ mice developed MALT lymphomas less frequently than their WT littermates after long-term infection of 16 months. Infected Plcg2Ali5/+ mice showed downregulation of proinflammatory cytokines and decreased H. felis-specific IgG1 and IgG2a antibody responses. These results suggested a blunted immune response of Plcg2Ali5/+ mice towards H. felis infection. Intriguingly, Plcg2Ali5/+ mice harboured higher numbers of CD73 expressing regulatory T cells (Tregs), possibly responsible for impaired immune response towards Helicobacter infection. We suggest that Plcg2Ali5/+ mice may be protected from developing gastric MALT lymphomas as a result of elevated Treg numbers, reduced response to H. felis and decrease of proinflammatory cytokines.
Collapse
|
9
|
Tsai HF, Hsu PN. Modulation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis by Helicobacter pylori in immune pathogenesis of gastric mucosal damage. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2016; 50:4-9. [PMID: 26947589 DOI: 10.1016/j.jmii.2016.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 12/20/2015] [Accepted: 01/17/2016] [Indexed: 12/28/2022]
Abstract
Helicobacter pylori infection is associated with chronic gastritis, peptic ulcer, gastric carcinoma, and gastric mucosa-associated lymphoid tissue lymphomas. Apoptosis induced by microbial infections is implicated in the pathogenesis of H. pylori infection. Enhanced gastric epithelial cell apoptosis during H. pylori infection was suggested to play an important role in the pathogenesis of chronic gastritis and gastric pathology. In addition to directly triggering apoptosis, H. pylori induces sensitivity to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis in gastric epithelial cells. Human gastric epithelial cells sensitized to H. pylori confer susceptibility to TRAIL-mediated apoptosis via modulation of death-receptor signaling. The induction of TRAIL sensitivity by H. pylori is dependent upon the activation of caspase-8 and its downstream pathway. H. pylori induces caspase-8 activation via enhanced assembly of the TRAIL death-inducing signaling complex through downregulation of cellular FLICE-inhibitory protein. Moreover, H. pylori infection induces infiltration of T lymphocytes and triggers inflammation to augment apoptosis. In H. pylori infection, significant increases in CCR6+ CD3+ T cell infiltration in the gastric mucosa was observed, and the CCR6 ligand, CCL20 chemokine, was selectively expressed in inflamed gastric tissues. These mechanisms initiate chemokine-mediated T lymphocyte trafficking into inflamed epithelium and induce mucosal injury during Helicobacter infection. This article will review recent findings on the interactions of H. pylori with host-epithelial signaling pathways and events involved in the initiation of gastric pathology, including gastric inflammation and mucosal damage.
Collapse
Affiliation(s)
- Hwei-Fang Tsai
- Department of Internal Medicine, Taipei Medical University Shuang Ho Hospital, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ping-Ning Hsu
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
10
|
Santos AM, Lopes T, Oleastro M, Gato IV, Floch P, Benejat L, Chaves P, Pereira T, Seixas E, Machado J, Guerreiro AS. Curcumin inhibits gastric inflammation induced by Helicobacter pylori infection in a mouse model. Nutrients 2015; 7:306-20. [PMID: 25569625 PMCID: PMC4303841 DOI: 10.3390/nu7010306] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/09/2014] [Indexed: 12/17/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection triggers a sequence of gastric alterations starting with an inflammation of the gastric mucosa that, in some cases, evolves to gastric cancer. Efficient vaccination has not been achieved, thus it is essential to find alternative therapies, particularly in the nutritional field. The current study evaluated whether curcumin could attenuate inflammation of the gastric mucosa due to H. pylori infection. Twenty-eight C57BL/6 mice, were inoculated with the H. pylori SS1 strain; ten non-infected mice were used as controls. H. pylori infection in live mice was followed-up using a modified 13C-Urea Breath Test (13C-UBT) and quantitative real-time polymerase chain reaction (PCR). Histologically confirmed, gastritis was observed in 42% of infected non-treated mice at both 6 and 18 weeks post-infection. These mice showed an up-regulation of the expression of inflammatory cytokines and chemokines, as well as of toll-like receptors (TLRs) and MyD88, at both time points. Treatment with curcumin decreased the expression of all these mediators. No inflammation was observed by histology in this group. Curcumin treatment exerted a significant anti-inflammatory effect in H. pylori-infected mucosa, pointing to the promising role of a nutritional approach in the prevention of H. pylori induced deleterious inflammation while the eradication or prevention of colonization by effective vaccine is not available.
Collapse
Affiliation(s)
- António M Santos
- Serviço de Medicina 4-Hospital de Santa Marta/Centro Hospitalar de Lisboa Central, Rua de Santa Marta, 50, 1169-024 Lisboa, Portugal.
| | - Teresa Lopes
- CEDOC-Nova Medical School-Faculdade de Ciências Médicas Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal.
| | - Mónica Oleastro
- Departamento de Doenças Infecciosas, Instituto Nacional de Saúde Dr. Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal.
| | - Inês Vale Gato
- Departamento de Doenças Infecciosas, Instituto Nacional de Saúde Dr. Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal.
| | - Pauline Floch
- Bacteriology Laboratory, Bordeaux University, 146 rue Léo Saignat F-33000 Bordeaux, France.
| | - Lucie Benejat
- Bacteriology Laboratory, Bordeaux University, 146 rue Léo Saignat F-33000 Bordeaux, France.
| | - Paula Chaves
- Serviço de Anatomia Patológica-Instituto Português de Oncologia Dr. Francisco Gentil, R. Prof. Lima Basto, 1099-023 Lisboa, Portugal.
| | - Teresa Pereira
- Serviço de Anatomia Patológica-Instituto Português de Oncologia Dr. Francisco Gentil, R. Prof. Lima Basto, 1099-023 Lisboa, Portugal.
| | - Elsa Seixas
- CEDOC-Nova Medical School-Faculdade de Ciências Médicas Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal.
| | - Jorge Machado
- Departamento de Doenças Infecciosas, Instituto Nacional de Saúde Dr. Ricardo Jorge, Avenida Padre Cruz, 1649-016 Lisboa, Portugal.
| | - António S Guerreiro
- Serviço de Medicina 4-Hospital de Santa Marta/Centro Hospitalar de Lisboa Central, Rua de Santa Marta, 50, 1169-024 Lisboa, Portugal.
| |
Collapse
|
11
|
Cook KW, Letley DP, Ingram RJM, Staples E, Skjoldmose H, Atherton JC, Robinson K. CCL20/CCR6-mediated migration of regulatory T cells to the Helicobacter pylori-infected human gastric mucosa. Gut 2014; 63:1550-9. [PMID: 24436142 PMCID: PMC4173663 DOI: 10.1136/gutjnl-2013-306253] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Helicobacter pylori-induced peptic ulceration is less likely to occur in patients with a strong gastric anti-inflammatory regulatory T cell (Treg) response. Migration of Tregs into the gastric mucosa is therefore important. OBJECTIVE To identify the homing receptors involved in directing Tregs to the gastric mucosa, and investigate how H pylori stimulates the relevant chemokine responses. DESIGN Gastric biopsy samples and peripheral blood were donated by 84 H pylori-infected and 46 uninfected patients. Luminex assays quantified gastric biopsy chemokine concentrations. Flow cytometry was used to characterise homing receptors on CD4(+)CD25(hi) Tregs. H pylori wild-type and isogenic mutants were used to investigate the signalling mechanisms behind CCL20 and IL-8 induction in gastric epithelial cell lines. Transwell assays were used to quantify Treg migration towards chemokines in vitro. RESULTS CCL20, CXCL1-3 and IL-8 concentrations were significantly increased in gastric biopsy samples from H pylori-infected patients. CCR6 (CCL20 receptor), CXCR1 and CXCR2 (IL-8 and CXCL1-3 receptors) were expressed by a higher proportion of peripheral blood Tregs in infected patients. Most gastric Tregs expressed these receptors. H pylori induced CCL20 production by gastric epithelial cells via cag pathogenicity island (cagPAI)-dependent NF-κB signalling. Foxp3(+), but not Foxp3(-), CD4 cells from infected mice migrated towards recombinant CCL20 in vitro. CONCLUSIONS As well as increasing Treg numbers, H pylori infection induces a change in their characteristics. Expression of CCR6, CXCR1 and CXCR2 probably enables their migration towards CCL20 and IL-8 in the infected gastric mucosa. Such qualitative changes may also explain how H pylori protects against some extragastric inflammatory disorders.
Collapse
Affiliation(s)
- Katherine W Cook
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Darren P Letley
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Richard J M Ingram
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Emily Staples
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Helle Skjoldmose
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - John C Atherton
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Karen Robinson
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, The University of Nottingham, Nottingham, UK
| |
Collapse
|
12
|
Chen JP, Wu MS, Kuo SH, Liao F. IL-22 negatively regulates Helicobacter pylori-induced CCL20 expression in gastric epithelial cells. PLoS One 2014; 9:e97350. [PMID: 24824519 PMCID: PMC4019584 DOI: 10.1371/journal.pone.0097350] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 04/18/2014] [Indexed: 12/30/2022] Open
Abstract
Helicobacter pylori is a Gram-negative bacterium that infects the human gastric mucosa and causes various gastric diseases. H. pylori infection induces the production of inflammatory chemokine CCL20 in gastric mucosa and leads to gastric inflammation. Given that the IL-22/IL-22R axis plays a critical role in the regulation of homeostasis and inflammation of epithelial cells at barrier surfaces, we investigated the effect of IL-22 on CCL20 expression induced by H. pylori. We demonstrated that H. pylori infection of the gastric epithelia-derived AGS cells significantly induced CCL20 expression and the induction was inhibited by IL-22. Functional analysis of the CCL20 promoter revealed that the H. pylori-induced CCL20 expression required the activation of NF-κB, and that IL-22 inhibited the induction by attenuating NF-κB activation. Knockdown of endogenous STAT3 by either short interfering RNAs or a short hairpin RNA significantly reduced the inhibitory effect of IL-22. Furthermore, STAT3 phosphorylation elicited by IL-22 was crucial for the inhibition of H. pylori-induced CCL20 expression. Consistent with the in vitro data showing that IL-22 negatively regulated H. pylori-induced CCL20 expression in gastric epithelial cells, studies on the tissue sections from patients with H. pylori infection also revealed an inverse association of IL-22 expression and CCL20 expression in vivo. Together, our findings suggest that IL-22 plays a role in the control of overproduction of the inflammatory chemokine and thus may protect the gastric mucosa from inflammation-mediated damage.
Collapse
Affiliation(s)
- Jia-Perng Chen
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Hsin Kuo
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Cancer Research Center and Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Fang Liao
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
13
|
Klein M, Brouwer MC, Angele B, Geldhoff M, Marquez G, Varona R, Häcker G, Schmetzer H, Häcker H, Hammerschmidt S, van der Ende A, Pfister HW, van de Beek D, Koedel U. Leukocyte attraction by CCL20 and its receptor CCR6 in humans and mice with pneumococcal meningitis. PLoS One 2014; 9:e93057. [PMID: 24699535 PMCID: PMC3974727 DOI: 10.1371/journal.pone.0093057] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/28/2014] [Indexed: 01/10/2023] Open
Abstract
We previously identified CCL20 as an early chemokine in the cerebrospinal fluid (CSF) of patients with pneumococcal meningitis but its functional relevance was unknown. Here we studied the role of CCL20 and its receptor CCR6 in pneumococcal meningitis. In a prospective nationwide study, CCL20 levels were significantly elevated in the CSF of patients with pneumococcal meningitis and correlated with CSF leukocyte counts. CCR6-deficient mice with pneumococcal meningitis and WT mice with pneumococcal meningitis treated with anti-CCL20 antibodies both had reduced CSF white blood cell counts. The reduction in CSF pleocytosis was also accompanied by an increase in brain bacterial titers. Additional in vitro experiments showed direct chemoattractant activity of CCL20 for granulocytes. In summary, our results identify the CCL20-CCR6 axis as an essential component of the innate immune defense against pneumococcal meningitis, controlling granulocyte recruitment.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Antibodies, Monoclonal/pharmacology
- Blotting, Western
- Brain/immunology
- Brain/metabolism
- Brain/microbiology
- Case-Control Studies
- Cells, Cultured
- Chemokine CCL20/antagonists & inhibitors
- Chemokine CCL20/immunology
- Chemokine CCL20/metabolism
- Chemotaxis, Leukocyte/immunology
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Humans
- Immunoenzyme Techniques
- Male
- Meningitis, Pneumococcal/cerebrospinal fluid
- Meningitis, Pneumococcal/immunology
- Meningitis, Pneumococcal/metabolism
- Meningitis, Pneumococcal/microbiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Prognosis
- Prospective Studies
- Receptors, CCR6/physiology
- Survival Rate
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Matthias Klein
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
- * E-mail:
| | - Matthijs C. Brouwer
- Department of Neurology, University of Amsterdam, Amsterdam, The Netherlands
- Center of Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - Barbara Angele
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | - Madelijn Geldhoff
- Department of Neurology, University of Amsterdam, Amsterdam, The Netherlands
- Center of Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Rosa Varona
- Departamento de Immunologia y Oncologia, Centro National de Biotecnologia, Madrid, Spain
| | - Georg Häcker
- Institute for Medical Microbiology and Hygiene, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Helga Schmetzer
- Medical Department III, Ludwig-Maximilians-University, Munich, Germany
| | - Hans Häcker
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, University of Greifswald, Greifswald, Germany
| | - Arie van der Ende
- Center of Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
- Department of Medical Microbiology, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Diederik van de Beek
- Department of Neurology, University of Amsterdam, Amsterdam, The Netherlands
- Center of Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - Uwe Koedel
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
14
|
Jafarzadeh A, Nemati M, Rezayati MT, Khoramdel H, Nabizadeh M, Hassanshahi G, Abdollahi H. Lower circulating levels of chemokine CXCL10 in Helicobacter pylori-infected patients with peptic ulcer: Influence of the bacterial virulence factor CagA. IRANIAN JOURNAL OF MICROBIOLOGY 2013; 5:28-35. [PMID: 23467184 PMCID: PMC3577565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND AND OBJECTIVES Alterations in CXCL10 (a Th1 chemokine) expression have been associated with various diseases. The aim of this study was to evaluate the serum CXCL10 levels in H. pylori-infected patients with peptic ulcer (PU), H. pylori-infected asymptomatic (AS) subjects and healthy H. pylori-negative subjects, and also to determine its association with bacterial virulence factor cytotoxin-associated gene A (CagA). MATERIALS AND METHODS Serum samples from 90 H. pylori infected patients with PU (70 were anti-CagA(+), 20 were anti-CagA(-)), 65 AS carriers (40 were anti-CagA(+), 25 were anti-CagA(-)) and 30 healthy H. pylori-negative subjects (as a control group) were tested for concentrations of CXCL10 by using the ELISA method. RESULTS The mean serum levels of CXCL10 in PU patients (96.64 ± 20.85 pg/mL) were significantly lower than those observed in AS subjects (162.16 ± 53.31 pg/mL, P < 0.01) and the control group (193.93 ± 42.14 pg/mL, P < 0.02). In the PU group, the serum levels of CXCL10 in anti-CagA(+) subjects was significantly higher in comparison to anti-CagA(-) patients (P < 0.04). CONCLUSION These results showed that the mean concentrations of CXCL10 in H. pylori-infected-PU patients was lower than AS carriers and control group. In the PU group, the serum levels of CXCL10 were associated with bacterial factor CagA.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Microbiology and Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran,Immunology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran,Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran, Corresponding author: Abdollah Jafarzadeh, Address: Department of Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran. Tel: +98-391-5234003. Fax: +98-391-522 5209. E-mail:
| | - Maryam Nemati
- Department of Microbiology and Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Taghi Rezayati
- Department of Microbiology and Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossain Khoramdel
- Immunology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mansooreh Nabizadeh
- Department of Microbiology and Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Hamid Abdollahi
- Department of Microbiology and Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
15
|
Deutsch AJA, Steinbauer E, Hofmann NA, Strunk D, Gerlza T, Beham-Schmid C, Schaider H, Neumeister P. Chemokine receptors in gastric MALT lymphoma: loss of CXCR4 and upregulation of CXCR7 is associated with progression to diffuse large B-cell lymphoma. Mod Pathol 2013; 26:182-94. [PMID: 22936065 DOI: 10.1038/modpathol.2012.134] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chemokine receptors have a crucial role in the development and progression of lymphoid neoplasms. To determine the chemokine receptor expression profile in gastric mucosa-associated lymphoid tissue (MALT) lymphoma, we performed an expression analysis of 19 chemokine receptors at mRNA levels by using real-time RT-PCR, as well as of five chemokine receptors--CCR8, CCR9, CXCR4, CXCR6 and CXCR7--by immunohistochemistry on human tissue samples of Helicobacter pylori-associated gastritis, gastric MALT lymphoma and gastric extranodal diffuse large B-cell lymphoma originating from MALT lymphoma (transformed MALT lymphoma). Following malignant transformation from H. pylori-associated gastritis to MALT lymphoma, an upregulation of CCR7, CXCR3 and CXCR7, and a loss of CXCR4 were detected. The transformation of gastric MALT lymphomas to gastric extranodal diffuse large B-cell lymphoma was accompanied by upregulation of CCR1, CCR5, CCR7, CCR8, CCR9, CXCR3, CXCR6, CXCR7 and XCR1. Remarkably, CXCR4 expression was exclusively found in nodal marginal B-cell lymphomas and nodal diffuse large B-cell lymphomas but not at extranodal manifestation sites, ie, in gastric MALT lymphomas or gastric extranodal diffuse large B-cell lymphomas. Furthermore, the incidence of bone marrow infiltration (16/51 with bone marrow involvement vs 35/51 with bone marrow involvement; Spearman ρ=0467 P<0.001) positively correlated with CXCR4 expression. CXCL12, the ligand of CXCR4 and CXCR7, was expressed by epithelial, endothelial and inflammatory cells, MALT lymphoma cells and was most strongly expressed by extranodal diffuse large B-cell lymphoma cells, suggesting at least in part an autocrine signaling pathway. Our data indicate that CXCR4 expression is associated with nodal manifestation and a more advanced stage of lymphomas and hence, might serve as useful clinical prognostic marker.
Collapse
Affiliation(s)
- Alexander J A Deutsch
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Serelli-Lee V, Ling KL, Ho C, Yeong LH, Lim GK, Ho B, Wong SBJ. Persistent Helicobacter pylori specific Th17 responses in patients with past H. pylori infection are associated with elevated gastric mucosal IL-1β. PLoS One 2012; 7:e39199. [PMID: 22761739 PMCID: PMC3382622 DOI: 10.1371/journal.pone.0039199] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 05/16/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ongoing Helicobacter pylori (HP) infection triggers a chronic active gastritis. Eradicating HP reduces gastric inflammation, but does not eliminate it. We sought to characterize this persistent gastritis, and demonstrate the persistence of HP-specific Th17 responses in individuals previously infected with HP but who no longer had evidence of ongoing infection. METHODOLOGY/PRINCIPAL FINDINGS Study subjects were divided into 3 groups 55 individuals had active HP infection (group A), 41 were diagnosed with previous HP infection (group P), and 59 were naïve to HP (group N). Blood and gastric tissue were obtained with written informed consent from all subjects, and immune responses were evaluated using flow cytometry, semi-quantitative real time PCR, immunofluorescent staining, ELISA, and multiplex cytometric bead array for cytokine quantification. Elevated IL-17A responses were observed in patients from group A compared to group N. Interestingly, IL-17A responses remained persistently elevated in the blood and gastric mucosa of individuals from group P, despite the absence of ongoing HP infection. Using purified CD4(+) T cells as effectors and antibodies that blocked antigen presentation by MHC Class II, we showed that these persistent IL-17A responses were mediated primarily by HP-specific Th17 cells, rather than other immune cells that have also been described to secrete IL-17A. Gastric mucosal IL-1β levels were also persistently elevated in group P, and neutralisation of IL-1β reduced the HP-specific IL-17A response of purified CD4(+) T cells to autologous HP-pulsed antigen presenting cells in vitro, suggesting a functional association between IL-1β and the persistent Th17 response in group P patients. CONCLUSIONS/SIGNIFICANCE Despite lack of ongoing HP infection, HP-specific Th17 cells persist in the blood and gastric mucosa of individuals with past HP infection. We speculate that this persistent inflammation might contribute to gastric mucosal pathology, for example, persistent increased gastric cancer risk despite eradication of HP.
Collapse
Affiliation(s)
- Victoria Serelli-Lee
- Department of Microbiology, National University of Singapore, Singapore, Republic of Singapore
| | - Khoon Lin Ling
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Cassandra Ho
- Department of Microbiology, National University of Singapore, Singapore, Republic of Singapore
| | - Lai Han Yeong
- Department of Microbiology, National University of Singapore, Singapore, Republic of Singapore
| | - Gek Keow Lim
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Republic of Singapore
| | - Bow Ho
- Department of Microbiology, National University of Singapore, Singapore, Republic of Singapore
| | - Soon Boon Justin Wong
- Department of Microbiology, National University of Singapore, Singapore, Republic of Singapore
- Immunology Programme, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
17
|
Interweaving microRNAs and proinflammatory cytokines in gastric mucosa with reference to H. pylori infection. J Clin Immunol 2011; 32:290-9. [PMID: 22161133 DOI: 10.1007/s10875-011-9626-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Accepted: 11/24/2011] [Indexed: 12/19/2022]
Abstract
Using endoscopic biopsies, gastric mucosal expression levels of interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor-alpha (TNF-α) messenger RNA (mRNA) and microRNAs (miRNAs) that were differentially expressed in association with Helicobacter pylori were assessed by quantitative reverse-transcriptase polymerase chain reaction. Among the H. pylori-positive mucosa, 17 out of 29 miRNAs had significant correlations with at least one of the four proinflammatory cytokines in expression. Among the 17 miRNAs, 15 were associated with the degree of neutrophil infiltration and, more prominently, the degree of mononuclear cell infiltration, according to the updated Sydney system. Persistent H. pylori infection may affect the mucosal expression profiles of miRNAs via chronic inflammation mediated by proinflammatory cytokines. There were significant positive correlations between certain miRNAs including the microRNA-200 family and IL-1β, IL-6, or TNF-α mRNA in H. pylori-negative gastric mucosa. Underscoring the causal association between miRNAs and proinflammatory cytokines may provide insights into the pathogenesis of H. pylori-associated gastritis linking to gastric carcinogenesis.
Collapse
|
18
|
Enhanced expression of CXCL13 in human Helicobacter pylori-associated gastritis. Dig Dis Sci 2011; 56:2887-94. [PMID: 21647655 DOI: 10.1007/s10620-011-1717-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 04/08/2011] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND AIMS Chemokine CXC ligand 13 (CXCL13) and CXC receptor type 5 (CXCR5) are constitutively expressed in tertiary lymphoid follicles where the CXCL13/CXCR5 system regulates B lymphocytes homing. In this study, we sought to examine CXCL13 expression in the H. pylori-infected and -uninfected gastric mucosa and to elucidate the implication in the pathogenesis of HAG in humans. METHODS Using endoscopic biopsies taken from the gastric antrum of 29 subjects infected with Helicobacter pylori and 22 uninfected subjects, mucosal CXCL13 mRNA and protein levels were measured by real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. RESULTS The CXCL13 expression levels were significantly more elevated in H. pylori-positive patients than uninfected ones. The CXCL13 expression levels correlated with the degree of chronic gastritis and bacterial colonization. Immunohistochemistry and in vitro infection assay showed that CXCL13 was not produced by the gastric epithelium, but the α-smooth muscle antigen expressing mesenchymal cells were the possible source of CXCL13 within H. pylori-infected gastric mucosa. CXCR5 immunostaining was seen in the CD20-positive lymphoid aggregates. CONCLUSIONS The enhanced induction of CXCL13 may be involved in the pathogenesis of H. pylori-associated gastritis.
Collapse
|
19
|
Helicobacter pylori VacA reduces the cellular expression of STAT3 and pro-survival Bcl-2 family proteins, Bcl-2 and Bcl-XL, leading to apoptosis in gastric epithelial cells. Dig Dis Sci 2011; 56:999-1006. [PMID: 20927590 DOI: 10.1007/s10620-010-1420-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 08/31/2010] [Indexed: 01/20/2023]
Abstract
BACKGROUND Helicobacter pylori vacuolating cytotoxin, VacA, stimulates apoptosis via a mitochondria-dependent pathway. VacA induces apoptosis via activation of the pro-apoptotic B-cell lymphoma (Bcl)-2 family proteins, Bcl-2-associated X protein (Bax) and Bcl-2 homologous antagonist/killer (Bak), while the implication of such pro-survival Bcl-2 family members as Bcl-2 and Bcl-XL in the VacA-induced apoptosis remains unknown. Signal transduction and activator of transcription 3 (STAT3) is a pivotal transcription factor that upregulates Bcl-2 and Bcl-XL. AIMS This study was conducted to elicit the implication of STAT3 and pro-survival Bcl-2 and Bcl-XL in the intrinsic apoptosis. METHODS Immunoblot and reverse transcriptase real-time polymerase chain reaction (RT-PCR) were employed to assess the cellular expression of STAT3, Bcl-2, and Bcl-XL in response to purified VacA in gastric adenocarcinoma cell lines. VacA-induced apoptosis was quantitated morphologically following knockdown by each specific small interfering RNA (siRNA) or in the presence of pharmacological inhibitors. RESULTS VacA reduced STAT3, Bcl-2, and Bcl-XL expression in a dose-dependent manner. Knockdown of STAT3, Bcl-2, and Bcl-XL by siRNA induced apoptosis to a similar extent in the case of sufficient VacA inoculation. The VacA-mediated reduction of STAT3 expression was independent of cellular vacuolization, since a vacuolar-type ATPase inhibitor, bafilomycin A1, did not inhibit VacA-induced reduction of STAT3, Bcl-2, and Bcl-XL expression. Instead, a c-JUN NH2-terminal kinase (JNK) inhibitor, SP600125, restored the VacA-induced reduction of STAT3 expression to the basal level. CONCLUSIONS VacA-induced apoptosis may be, in part, implicated in the reduction of STAT3 linking to the downregulation of Bcl-2 and Bcl-XL, in association with JNK activity.
Collapse
|
20
|
He C, Zhang SL, Hu CJ, Tong DW, Li YZ. Higher levels of CCL20 expression on peripheral blood mononuclear cells of chinese patients with inflammatory bowel disease. Immunol Invest 2010; 39:16-26. [PMID: 20064082 DOI: 10.3109/08820130903380732] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study aimed at characterizing the levels of CCL20 mRNA transcripts in peripheral mononuclear blood cells (PMBC) of 56 Chinese patients with inflammatory bowel disease (IBD), 30 other intestinal diseases and 30 healthy controls by quantitative real time polymerase chain reaction. The levels of CCL20 mRNA transcripts in PBMC of patients with IBD were significantly higher than that of patients with non-IBD intestinal diseases and healthy controls (p < 0.01) and the CCL20 expression in active IBD patients was significantly higher than that in remission patients (p < 0.01). Importantly, the levels of CCL20 expression in PBMC were significantly correlated with the degrees of disease severity, the levels of erythrocyte sedimentation rate and C-reaction protein, but not hemoglobin, in patients with IBD (p < 0.01). Furthermore, the levels of CCL20 expression in active IBD patients after treatment with salazosulphapyridine or prednisone were significantly reduced, as compared with before treatment (p < 0.01). Therefore, analysis of CCL20 expression in PBMC may be used as a surrogate measure for evaluation of IBD activity, disease progression and therapeutic efficacy in Chinese IBD patients.
Collapse
Affiliation(s)
- Chun He
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | | | | | | | | |
Collapse
|
21
|
Norimura D, Isomoto H, Nakayama T, Hayashi T, Suematsu T, Nakashima Y, Inoue N, Matsushima K, Yamaguchi N, Ohnita K, Mizuta Y, Inoue K, Shikuwa S, Nakao K, Kohno S. Magnifying endoscopic observation with narrow band imaging for specialized intestinal metaplasia in barrett's esophagus with special reference to light blue crests. Dig Endosc 2010; 22:101-6. [PMID: 20447202 DOI: 10.1111/j.1443-1661.2010.00940.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Barrett's esophagus (BE) with specialized intestinal metaplasia (SIM) is at high risk of esophageal adenocarcinoma. Magnified endoscopy with narrow band imaging (ME-NBI) can be useful for detecting this condition. In addition to pit patterns, light blue crests (LBC), blue-whitish patchy areas on the metaplastic epithelia of the stomach, can predict SIM in BE under ME-NBI observation. METHODS A total of 54 patients with BE underwent ME-NBI to identify IM pits (tubular and villous pits) and LBC. Biopsy samples were taken for histological evaluation of IM, immunohistochemical staining for CD10, MUC2 and MUC5AC antigen, transmission electron microscopy and real-time polymerase chain reaction (RT-PCR) analysis of CD10 mRNA expression. RESULTS IM pit pattern with ME-NBI for the diagnosis of IM yielded acceptable sensitivity, specificity and accuracy at 92%, 77% and 83%, respectively. However, the sensitivity, specificity and accuracy of LBC with ME-NBI for IM were comparably high at 79%, 97% and 89%, respectively. Upon immunohistochemistry, all 19 metaplastic epithelia of LBC-positive BE showed immunoreactivity against anti-MUC2 antibody, whereas CD10 antigen was identified in 11 of the 19 LBC-positive BE. Brush borders were seen on IM epithelia using electron microscopy. On real-time PCR analysis, CD10 mRNA levels in the LBC-positive BE were higher compared to those in the LBC-negative BE. CONCLUSION The appearance of LBC can be an accurate sign to predict SIM in BE and may be associated with high CD10 expression, possibly along with brush borders.
Collapse
Affiliation(s)
- Daisuke Norimura
- Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Science, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Matsushima K, Isomoto H, Inoue N, Nakayama T, Hayashi T, Nakayama M, Nakao K, Hirayama T, Kohno S. MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int J Cancer 2010; 128:361-70. [PMID: 20333682 DOI: 10.1002/ijc.25348] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Accepted: 03/09/2010] [Indexed: 02/06/2023]
Abstract
The study was conducted to determine expression patterns of microRNA (miRNA), a non-coding RNA that controls gene expression mainly through translational repression, in gastric mucosa infected with Helicobacter pylori. Using endoscopic biopsy specimens, miRNA expression patterns in H. pylori-infected gastric mucosa were determined by microarray. The differentially expressed miRNAs were quantitated by real-time reverse-transcriptase polymerase chain reaction (RT-PCR). An in vitro infection model was assessed to monitor the regulation of miRNAs in gastric epithelium in response to H. pylori. The comprehensive method unraveled the expression profiles; among 470 human miRNAs loaded, 55 were differentially expressed between H. pylori-positive and -negative subjects. The expression levels were significantly decreased in 30 miRNAs, whereas hsa-miRNA-223 was the only miRNA to be overexpressed on quantitative RT-PCR. Eight miRNAs enabled discrimination of H. pylori status with acceptable accuracy. Gastritis scores of activity and chronic inflammation according to the updated Sydney system correlated significantly with the expression levels of diverse miRNAs. Cure of the infection with an anti-H. pylori regimen restored decreased expression in 14 of the 30 miRNAs. Expression levels of some miRNAs, including let-7 family members, were significantly altered following infection with a virulent H. pylori strain carrying intact cag pathogenicity island including cagA but not isogenic mutants. These results provide insights into miRNA involvement in the pathogenesis of H. pylori-associated gastritis. cagA may be involved in cellular regulation of certain miRNAs in the gastric epithelium.
Collapse
Affiliation(s)
- Kayoko Matsushima
- Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Interplay between Helicobacter pylori and immune cells in immune pathogenesis of gastric inflammation and mucosal pathology. Cell Mol Immunol 2010; 7:255-9. [PMID: 20190789 DOI: 10.1038/cmi.2010.2] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori infection is associated with an inflammatory response in the gastric mucosa, leading to chronic gastritis, peptic ulcers, gastric carcinoma and gastric mucosa-associated lymphoid tissue (MALT) lymphomas. Recent studies have shown that apoptosis of gastric epithelial cells is increased during H. pylori infection. Apoptosis induced by microbial infections are factors implicated in the pathogenesis of H. pylori infection. The enhanced gastric epithelial cell apoptosis in H. pylori infection has been suggested to play an important role in the pathogenesis of chronic gastritis and gastric pathology. In addition to directly triggering apoptosis, H. pylori induces sensitivity to tumor-necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis in gastric epithelial cells via modulation of TRAIL apoptosis signaling. Moreover, H. pylori infection induces infiltration of T lymphocytes and triggers inflammation to augment apoptosis. In H. pylori infection, there was significantly increased CCR6(+)CD3(+ )T-cell infiltration in the gastric mucosa, and the CCR6 ligand, CCL20 chemokine, was selectively expressed in inflamed gastric tissues. These results implicate that the interaction between CCL20 and CCR6 may play a role in recruiting T cells to the sites of inflammation in the gastric mucosa during Helicobacter infection. Through these mechanisms, chemokine-mediated T lymphocyte trafficking into inflamed epithelium is initiated and the mucosal injury in Helicobacter infection is induced. This article will review the recent novel findings on the interactions of H. pylori with diverse host epithelial signaling pathways and events involved in the initiation of gastric pathology, including gastric inflammation, mucosal damage and development of MALT lymphomas.
Collapse
|
24
|
Helicobacter pylori promotes the production of thymic stromal lymphopoietin by gastric epithelial cells and induces dendritic cell-mediated inflammatory Th2 responses. Infect Immun 2009; 78:108-14. [PMID: 19841072 DOI: 10.1128/iai.00762-09] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Helicobacter pylori colonizes the stomach and induces strong, specific local and systemic humoral and cell-mediated immunity, resulting in the development of chronic gastritis in humans. Although H. pylori-induced chronic atrophic gastritis is characterized by marked infiltration of T helper type 1 (Th1) cytokine-producing CD4(+) T cells, almost all of the inflamed gastric mucosae also contain focal lymphoid aggregates with germinal centers. In addition, typical H. pylori-induced chronic gastritis in children, called follicular gastritis, is characterized by B-cell follicle formation in the gastric mucosa. The aim of this study was to examine whether thymic stromal lymphopoietin (TSLP), an epithelial-cell-derived cytokine inducing a dendritic cell (DC)-mediated inflammatory Th2 response, is involved in Th2 responses triggering B-cell activation in H. pylori-induced gastritis. Here, we show that H. pylori triggered human gastric epithelial cells to produce TSLP, together with the DC-attracting chemokine MIP-3alpha and the B-cell-activating factor BAFF. After DCs were incubated with supernatants from H. pylori-infected epithelial cells, the conditioned cells expressed high levels of costimulatory molecules, such as CD80, and triggered naïve CD4(+) T cells to produce high levels of the Th2 cytokines interleukin-4 and interleukin-13 and of the inflammatory cytokines tumor necrosis factor alpha and gamma interferon. In contrast, after incubation of the supernatants with the neutralizing antibodies to TSLP, the conditioned DCs did not prime T cells to produce high levels of Th2 cytokines. These results, together with the finding that TSLP was expressed by the epithelial cells of human follicular gastritis, suggest that H. pylori can directly trigger epithelial cells to produce TSLP. It also suggests that TSLP-mediated DC activation may be involved in Th2 responses triggering B-cell activation in H. pylori-induced gastritis.
Collapse
|