1
|
Li J, Chen X, Mao C, Xiong M, Ma Z, Zhu J, Li X, Chen W, Ma H, Ye X. Epiberberine ameliorates MNNG-induced chronic atrophic gastritis by acting on the EGFR-IL33 axis. Int Immunopharmacol 2025; 145:113718. [PMID: 39642571 DOI: 10.1016/j.intimp.2024.113718] [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] [Received: 10/07/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
Chronic atrophic gastritis (CAG) is a prevalent form of chronic gastritis that presents with chronic inflammation of the gastric mucosa, localised gastric mucosal glandular atrophy and intestinal metaplasia. Despite the existence of diagnostic criteria, effective therapeutic strategies for this condition remain to be developed. The objective of this study was to examine the potential therapeutic benefits of epiberberine in mitigating MNNG-induced CAG and to elucidate the underlying mechanisms. MNNG was employed to establish a CAG mouse model and a GES-1 cell model, and EPI was observed to be efficacious in ameliorating the gastric mucosal injury and inflammatory infiltration induced by MNNG in the CAG model mice, a finding that was subsequently validated in the GES-1 model cells. Bioinformatics analysis indicated that EPI may exert a direct effect on EGFR, thereby regulating the expression of IL-33 and thereby achieving the therapeutic effect of CAG. This hypothesis was also validated by molecular docking prediction, CETSA, and overexpression of EGFR in GES-1 model cells, using EGFR agonists and inhibitors to further demonstrate that EPI may act as an antagonist supplement to EGFR for the treatment of CAG.
Collapse
Affiliation(s)
- Juan Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Xiantao Chen
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Changxia Mao
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Mengyuan Xiong
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Jianyu Zhu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xuegang Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China.
| | - Hang Ma
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China.
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
2
|
Werawatganone P, Werawatganon D, Noonak N, Chayanupatkul M, Chatsuwan T, Klaikeaw N, Muangsiri W, Siriviriyakul P. Unveiling the Potency of Gardenia Extract Against H. pylori: Insights from In Vitro and In Vivo Studies. Biomedicines 2025; 13:92. [PMID: 39857676 PMCID: PMC11760463 DOI: 10.3390/biomedicines13010092] [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: 11/17/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND AND AIM Gardenia jasminoides (G. jasminoides) could treat various inflammatory diseases. This study aimed to investigate the effects of G. jasminoides fruit extract on gastric inflammation and protective mechanisms in Helicobacter pylori (H. pylori)-induced gastritis. Experimental procedure: G. jasminoides fruit extract was prepared and analyzed for geniposide content. The inhibitory effect of the extract on H. pylori growth was investigated using the disk diffusion method. The in vitro anti-inflammatory property of the extract was evaluated using the erythrocyte membrane stabilization method. Thirty-five male Sprague-Dawley rats were inoculated with H. pylori (108-1010 colony-forming unit/mL) and divided into five groups. Each group was treated with various doses of the extract (98-395 mg/kg). The serum and stomach tissue of the rats were evaluated using enzyme-linked immunosorbent assay, histopathology, and immunohistochemistry. RESULTS AND CONCLUSIONS The geniposide content in the dried extract was 8.12% ± 0.79% by dry weight. The inhibition zone was observed at the extract ≥ 1.97 mg/disk, and the extract presented anti-inflammatory potential. The H. pylori-inoculated rats had a significant increase in serum interleukin (IL)-17, IL-33, and gastric epidermal growth factor (EGF) levels and a significant decrease in serum prostaglandin E2 level (p < 0.05) in conjunction with the development of gastric inflammation on histopathology. The treatment of the extract could significantly decrease the serum IL-17, IL-33, and gastric EGF levels, significantly increase the serum PGE2 level (p < 0.05), and improve gastric histopathology. Thus, G. jasminoides fruit extract attenuated H. pylori-induced gastritis by inhibiting bacterial growth, reducing inflammation, and enhancing protective mechanisms.
Collapse
Affiliation(s)
- Pornpen Werawatganone
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (P.W.); (W.M.)
| | - Duangporn Werawatganon
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| | - Nattida Noonak
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| | - Maneerat Chayanupatkul
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Naruemon Klaikeaw
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Walaisiri Muangsiri
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (P.W.); (W.M.)
| | - Prasong Siriviriyakul
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand (M.C.)
| |
Collapse
|
3
|
Boziki M, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Tzitiridou-Chatzopoulou M, Doulberis M, Kazakos E, Deretzi G, Grigoriadis N, Kountouras J. Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut-Brain Axis and Helicobacter pylori Infection. Neurol Int 2024; 16:1750-1778. [PMID: 39728753 DOI: 10.3390/neurolint16060127] [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: 08/20/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND The innate immune response aims to prevent pathogens from entering the organism and/or to facilitate pathogen clearance. Innate immune cells, such as macrophages, mast cells (MCs), natural killer cells and neutrophils, bear pattern recognition receptors and are thus able to recognize common molecular patterns, such as pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs), the later occurring in the context of neuroinflammation. An inflammatory component in the pathology of otherwise "primary cerebrovascular and neurodegenerative" disease has recently been recognized and targeted as a means of therapeutic intervention. Activated MCs are multifunctional effector cells generated from hematopoietic stem cells that, together with dendritic cells, represent first-line immune defense mechanisms against pathogens and/or tissue destruction. METHODS This review aims to summarize evidence of MC implication in the pathogenesis of neurodegenerative diseases, namely, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. RESULTS In view of recent evidence that the gut-brain axis may be implicated in the pathogenesis of neurodegenerative diseases and the characterization of the neuroinflammatory component in the pathology of these diseases, this review also focuses on MCs as potential mediators in the gut-brain axis bi-directional communication and the possible role of Helicobacter pylori, a gastric pathogen known to alter the gut-brain axis homeostasis towards local and systemic pro-inflammatory responses. CONCLUSION As MCs and Helicobacter pylori infection may offer targets of intervention with potential therapeutic implications for neurodegenerative disease, more clinical and translational evidence is needed to elucidate this field.
Collapse
Affiliation(s)
- Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Nella
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Midwifery Department, School of Healthcare Sciences, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, 5001 Aarau, Switzerland
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgia Deretzi
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Department of Neurology, Papageorgiou General Hospital, 54629 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
4
|
Ismael AT, Abdulhameed RA, Hamdi BA, Tawfeeq RD, Ommar A. C-Kit Immunohistochemical Expression as a Complementary Method to Assess Mast Cell Density in Helicobacter pylori-Mediated Gastritis. Digestion 2024:1-7. [PMID: 39278202 DOI: 10.1159/000541387] [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: 04/30/2024] [Accepted: 08/24/2024] [Indexed: 09/18/2024]
Abstract
INTRODUCTION Chronic gastritis is a group of conditions commonly characterized by stomach lining inflammation. The study aimed to investigate the clinical and pathological aspects that play a role in its development. Additionally, the study examines the use of CD117 as an immunohistochemistry marker in evaluating mast cell density (MCD). METHODS This retrospective, cross-sectional study was conducted in Iraqi Kurdistan with a sample size of 380 patients. Patient data included gastritis type, neutrophil infiltration severity, mononuclear cell infiltration within the lamina propria, intestinal metaplasia, and glandular atrophy, which were categorized and given a score. The CD117 level was identified using an anti-human rabbit polyclonal antibody. RESULTS A statistically significant association was revealed between Helicobacter pylori-mediated gastritis and non-specific gastritis with age, activity, H. pylori and MCD, dysplasia, and malignancy. Meanwhile, no association was found with gender, inflammatory infiltrate, intestinal metaplasia, and glandular atrophy. C-Kit exhibited a marked increase in MCD in patients with H. pylori-mediated gastritis, intestinal metaplasia, atrophy, and gastric carcinoma. However, a significant decrease in MCD was observed on repeating endoscopy evaluations for patients after treatment. CONCLUSION Regions that exhibit severe inflammation, metaplasia, atrophy, and carcinoma demonstrated an increase in MCD with H. pylori-mediated gastritis. A detailed investigation in clinical practice to screen early diagnosis and treatment needs to be performed in high H. pylori prevalence.
Collapse
Affiliation(s)
- Ava T Ismael
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Rafal A Abdulhameed
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Bushra A Hamdi
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Rawaz D Tawfeeq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Aram Ommar
- Department of Clinical Pharmacy, College of Pharmacy, Al-Kitab University, Kirkuk, Iraq
| |
Collapse
|
5
|
Sadighi A, Aghamohammadpour Z, Sadeghpour Heravi F, Somi MH, Masnadi Shirazi Nezhad K, Hosseini S, Bahman Soufiani K, Ebrahimzadeh Leylabadlo H. The protective effects of Helicobacter pylori: A comprehensive review. JOURNAL OF RESEARCH IN CLINICAL MEDICINE 2024; 12:17. [DOI: 10.34172/jrcm.34509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2025] Open
Abstract
Previous reports have estimated that approximately half of the world’s population is infected with Helicobacter pylori, the most prevalent infectious agent responsible for gastrointestinal illnesses. Due to the life-threatening effects of H. pylori infections, numerous studies have focused on developing medical therapies for H. pylori infections, while the commensal relationship and positive impacts of this bacterium on overall human health have been largely overlooked. The inhibitory efficacy of H. pylori on the progression of several chronic inflammatory disorders and gastrointestinal diseases has recently raised concerns about whether this bacterium should be eradicated in affected individuals or maintained in an appropriate balance depending on the patient’s condition. This review investigates the beneficial effects of H. pylori in preventing various diseases and discusses the potential association of conditions such as inflammatory disorders with the absence of H. pylori.
Collapse
Affiliation(s)
- Ali Sadighi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Aghamohammadpour
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Samaneh Hosseini
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Katayoun Bahman Soufiani
- Department of Laboratory Sciences and Microbiology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | | |
Collapse
|
6
|
Kountouras J, Boziki M, Kazakos E, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Vardaka E, Mouratidou MC, Kyrailidi F, Tzitiridou-Chatzopoulou M, Orovou E, Giartza-Taxidou E, Deretzi G, Grigoriadis N, Doulberis M. Impact of Helicobacter pylori and metabolic syndrome on mast cell activation-related pathophysiology and neurodegeneration. Neurochem Int 2024; 175:105724. [PMID: 38508416 DOI: 10.1016/j.neuint.2024.105724] [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] [Received: 12/08/2023] [Revised: 03/03/2024] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Both Helicobacter pylori (H. pylori) infection and metabolic syndrome (MetS) are highly prevalent worldwide. The emergence of relevant research suggesting a pathogenic linkage between H. pylori infection and MetS-related cardio-cerebrovascular diseases and neurodegenerative disorders, particularly through mechanisms involving brain pericyte deficiency, hyperhomocysteinemia, hyperfibrinogenemia, elevated lipoprotein-a, galectin-3 overexpression, atrial fibrillation, and gut dysbiosis, has raised stimulating questions regarding their pathophysiology and its translational implications for clinicians. An additional stimulating aspect refers to H. pylori and MetS-related activation of innate immune cells, mast cells (MC), which is an important, often early, event in systemic inflammatory pathologies and related brain disorders. Synoptically, MC degranulation may play a role in the pathogenesis of H. pylori and MetS-related obesity, adipokine effects, dyslipidemia, diabetes mellitus, insulin resistance, arterial hypertension, vascular dysfunction and arterial stiffness, an early indicator of atherosclerosis associated with cardio-cerebrovascular and neurodegenerative disorders. Meningeal MC can be activated by triggers including stress and toxins resulting in vascular changes and neurodegeneration. Likewise, H.pylori and MetS-related MC activation is linked with: (a) vasculitis and thromboembolic events that increase the risk of cardio-cerebrovascular and neurodegenerative disorders, and (b) gut dysbiosis-associated neurodegeneration, whereas modulation of gut microbiota and MC activation may promote neuroprotection. This narrative review investigates the intricate relationship between H. pylori infection, MetS, MC activation, and their collective impact on pathophysiological processes linked to neurodegeneration. Through a comprehensive search of current literature, we elucidate the mechanisms through which H. pylori and MetS contribute to MC activation, subsequently triggering cascades of inflammatory responses. This highlights the role of MC as key mediators in the pathogenesis of cardio-cerebrovascular and neurodegenerative disorders, emphasizing their involvement in neuroinflammation, vascular dysfunction and, ultimately, neuronal damage. Although further research is warranted, we provide a novel perspective on the pathophysiology and management of brain disorders by exploring potential therapeutic strategies targeting H. pylori eradication, MetS management, and modulation of MC to mitigate neurodegeneration risk while promoting neuroprotection.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece.
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, 50100, Macedonia, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Maria Nella
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Elisabeth Vardaka
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, Alexander Campus, 57400, Macedonia, Greece
| | - Maria C Mouratidou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Foteini Kyrailidi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, 50100, Macedonia, Greece
| | - Eirini Orovou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, 50100, Macedonia, Greece
| | - Evaggelia Giartza-Taxidou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Georgia Deretzi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Department of Neurology, Papageorgiou General Hospital, Thessaloniki, Macedonia, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Gastroklinik, Private Gastroenterological Practice, 8810, Horgen, Switzerland; Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001, Aarau, Switzerland
| |
Collapse
|
7
|
Tzitiridou-Chatzopoulou M, Kazakos E, Orovou E, Andronikidi PE, Kyrailidi F, Mouratidou MC, Iatrakis G, Kountouras J. The Role of Helicobacter pylori and Metabolic Syndrome-Related Mast Cell Activation Pathologies and Their Potential Impact on Pregnancy and Neonatal Outcomes. J Clin Med 2024; 13:2360. [PMID: 38673633 PMCID: PMC11050948 DOI: 10.3390/jcm13082360] [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: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Helicobacter pylori infection, a significant global burden beyond the gastrointestinal tract, has long been implicated in various systemic pathologies. Rising evidence suggests that the bacterium's intricate relationship with the immune system and its potential to induce chronic inflammation impact diverse pathophysiological processes in pregnant women that may in turn affect the incidence of several adverse pregnancy and neonate outcomes. Helicobacter pylori infection, which has been linked to metabolic syndrome and other disorders by provoking pericyte dysfunction, hyperhomocysteinemia, galectin-3, atrial fibrillation, gut dysbiosis, and mast cell activation pathologies, may also contribute to adverse pregnancy and neonatal outcomes. Together with increasing our biological understanding of the individual and collective involvement of Helicobacter pylori infection-related metabolic syndrome and concurrent activation of mast cells in maternal, fetus, and neonatal health outcomes, the present narrative review may foster related research endeavors to offer novel therapeutic approaches and informed clinical practice interventions to mitigate relevant risks of this critical topic among pregnant women and their offspring.
Collapse
Affiliation(s)
- Maria Tzitiridou-Chatzopoulou
- School of Health Sciences, Department of Midwifery, University of Western Macedonia, 50100 Koila, Greece; (M.T.-C.); (E.K.); (E.O.)
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Evangelos Kazakos
- School of Health Sciences, Department of Midwifery, University of Western Macedonia, 50100 Koila, Greece; (M.T.-C.); (E.K.); (E.O.)
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Eirini Orovou
- School of Health Sciences, Department of Midwifery, University of Western Macedonia, 50100 Koila, Greece; (M.T.-C.); (E.K.); (E.O.)
| | - Paraskevi Eva Andronikidi
- Department of Nephrology, Aretaieion University Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Foteini Kyrailidi
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Maria C. Mouratidou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| | - Georgios Iatrakis
- Department of Midwifery, University of West Attica, 12243 Athens, Greece;
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, Macedonia, 54642 Thessaloniki, Greece; (F.K.); (M.C.M.)
| |
Collapse
|
8
|
Liu K, Huang H, Xiong M, Wang Q, Chen X, Feng Y, Ma H, Chen W, Li X, Ye X. IL-33 Accelerates Chronic Atrophic Gastritis through AMPK-ULK1 Axis Mediated Autolysosomal Degradation of GKN1. Int J Biol Sci 2024; 20:2323-2338. [PMID: 38617533 PMCID: PMC11008276 DOI: 10.7150/ijbs.93573] [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: 12/25/2023] [Accepted: 03/26/2024] [Indexed: 04/16/2024] Open
Abstract
Chronic atrophic gastritis (CAG) is a complex disease characterized by atrophy and inflammation in gastric mucosal tissue, especially with high expression of interleukins. However, the interaction and mechanisms between interleukins and gastric mucosal epithelial cells in CAG remain largely elusive. Here, we elucidate that IL-33 stands out as the predominant inflammatory factor in CAG, and its expression is induced by H. pylori and MNNG through the ROS-STAT3 signaling pathway. Furthermore, our findings reveal that the IL-33/ST2 axis is intricately involved in the progression of CAG. Utilizing phosphoproteomics mass spectrometry, we demonstrate that IL-33 enhances autophagy in gastric epithelial cells through the phosphorylation of AMPK-ULK1 axis. Notably, inhibiting autophagy alleviates CAG severity, while augmentation of autophagy exacerbates the disease. Additionally, ROS scavenging emerges as a promising strategy to ameliorate CAG by reducing IL-33 expression and inhibiting autophagy. Intriguingly, IL-33 stimulation promotes GKN1 degradation through the autolysosomal pathway. Clinically, the combined measurement of IL-33 and GKN1 in serum shows potential as diagnostic markers. Our findings unveil an IL-33-AMPK-ULK1 regulatory mechanism governing GKN1 protein stability in CAG, presenting potential therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Kewei Liu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hongxia Huang
- Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Mengyuan Xiong
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Qiaojiao Wang
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Xiantao Chen
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Yinqiong Feng
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400000, China
| | - Hang Ma
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, 400038, China
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400000, China
| | - Xuegang Li
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, 400038, China
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
9
|
Doulberis M, Papaefthymiou A, Polyzos SA, Boziki M, Kazakos E, Tzitiridou-Chatzopoulou M, Vardaka E, Hammrich C, Kulaksiz H, Riva D, Kiosses C, Linas I, Touloumtzi M, Stogianni A, Kountouras J. Impact of Helicobacter pylori and metabolic syndrome-related mast cell activation on cardiovascular diseases. FRONTIERS IN GASTROENTEROLOGY 2024; 3. [DOI: 10.3389/fgstr.2024.1331330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Helicobacter pylori, a widely renowned bacterium, has recently gained attention owing to its potential impact on extragastric health. The emergence of research linking H. pylori infection with metabolic syndrome (MetS)-related cardiovascular diseases (CVDs) has raised intriguing questions about the pathogenic linkage and its translational implications for clinicians. MetS encompasses a collection of metabolic abnormalities that considerably elevate the risk of CVDs and cerebrovascular diseases. Emerging evidence supports a potential pathogenetic role of H. pylori for MetS-related disorders through mechanisms implicating chronic smoldering inflammation, insulin resistance (IR), and modulation of immune responses. One intriguing aspect of this possible connection is the role of mast cells (MCs), a subset of immune cells representing innate immune system effector cells. They play a fundamental role in innate immune responses and the modulation of adaptive immunity. Activated MCs are commonly found in patients with MetS-related CVD. Recent studies have also suggested that H. pylori infection may activate MCs, triggering the release of pro-inflammatory mediators that contribute to IR and atherosclerosis. Understanding these intricate interactions at the cellular level provides new insights into the development of therapeutic strategies targeting both H. pylori infection and MetS-related MCs activation. This review investigates the current state of research regarding the potential impact of H. pylori infection and MetS-related MCs activation on the pathophysiology of CVD, thereby opening up new avenues for related research and paving the way for innovative approaches to prevention and treatment in clinical practice
Collapse
|
10
|
Lv Y, Tian W, Teng Y, Wang P, Zhao Y, Li Z, Tang S, Chen W, Xie R, Lü M, Zhuang Y. Tumor-infiltrating mast cells stimulate ICOS + regulatory T cells through an IL-33 and IL-2 axis to promote gastric cancer progression. J Adv Res 2024; 57:149-162. [PMID: 37086778 PMCID: PMC10918354 DOI: 10.1016/j.jare.2023.04.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/29/2023] [Accepted: 04/15/2023] [Indexed: 04/24/2023] Open
Abstract
INTRODUCTION In solid tumors, regulatory T cell (Treg) and mast cell perform different roles depending on the microenvironment. Nevertheless, mast cell and Treg-mediated interactions in gastric cancer (GC) are unclear, as are their regulation, function, and clinical significance. OBJECTIVE The present study demonstrated the mechanism of tumor-infiltrating mast cells stimulating ICOS+ regulatory T cells via the IL-33/IL-2 axis to promote the growth of gastric cancer. METHODS Analyses of 98 patients with GC were conducted to examine mast cell counts, ICOS+ Tregs, and the levels of IL-33 or IL-2. Isolated ICOS+ Treg and CD8+ T cell were stimulated, cultured and tested for their functional abilities in vitro and in vivo. RESULTS GC patients exhibited a significantly more production of IL-33 in tumors. Mast cell stimulated by tumor-derived IL-33 exhibited a prolonged lifespan through IL-33 mediated inhibition of apoptosis. Moreover, mast cells stimulated by tumor-derived IL-33 secreted IL-2, which induced Treg expansion. These inducible Tregs displayed an activated immunosuppressive phenotype with positive expression for the inducible T cell co-stimulator (ICOS). In vitro, IL-2 from IL to 33-stimulated mast cells induced increased numbers of ICOS+ Tregs with increased immunosuppressive activity against proliferation and effector function of CD8+ T cell. In vivo, ICOS+ Tregs were treated with anti-IL-2 neutralizing antibody followed by co-injection with CD8+ T cells in GC mouse model, which showed an increased CD8+ T cell infiltration and effector molecules production, meanwhile tumor growth and progression were inhibited. Besides, reduction in GC patient survival was associated with tumor-derived ICOS+ Tregs. CONCLUSION Our results highlight a crosstalk between GC-infiltrating mast cells and ICOS+ Tregs and provide a novel mechanism describing ICOS+ Treg expansion and induction by an IL-33/mast cell/IL-2 signaling axis in GC, and also provide functional evidence that the modulation of this immunosuppressive pathway can attenuate GC-mediated immune tolerance.
Collapse
Affiliation(s)
- Yipin Lv
- Department of Digestive Diseases, The General Hospital of Western Theater Command, Chengdu, Sichuan, China; National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| | - Wenqing Tian
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Teng
- The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, China
| | - Pan Wang
- The 940th Hospital of Joint Logistics Support Force of PLA, Lanzhou, China
| | - Yongliang Zhao
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhengyan Li
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shanhong Tang
- Department of Digestive Diseases, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Weisan Chen
- La Trobe Institute of Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Rui Xie
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China.
| | - Muhan Lü
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Yuan Zhuang
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China; Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.
| |
Collapse
|
11
|
Guo Y, Wang B, Gao H, He C, Xin S, Hua R, Liu X, Zhang S, Xu J. Insights into the Characteristics and Functions of Mast Cells in the Gut. GASTROENTEROLOGY INSIGHTS 2023; 14:637-652. [DOI: 10.3390/gastroent14040043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Mast cells have vital functions in allergic responses and parasite ejection, while the underlying mechanisms remain unclear. Meanwhile, MCs are essential for the maintenance of GI barrier function, and their interactions with neurons, immune cells, and epithelial cells have been related to various gastrointestinal (GI) disorders. An increasing number of investigations are being disclosed, with a lack of inner connections among them. This review aims to highlight their properties and categorization and further delve into their participation in GI diseases via interplay with neurons and immune cells. We also discuss their roles in diseases like inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS). Based on the evidence, we advocated for their potential application in clinical practices and advocated future research prospects.
Collapse
Affiliation(s)
- Yuexin Guo
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Boya Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
- Department of Clinical Laboratory, Aerospace Center Hospital, Peking University, Beijing 100049, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Rongxuan Hua
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaohui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Sitian Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
12
|
Guo X, Sun M, Yang P, Meng X, Liu R. Role of mast cells activation in the tumor immune microenvironment and immunotherapy of cancers. Eur J Pharmacol 2023; 960:176103. [PMID: 37852570 DOI: 10.1016/j.ejphar.2023.176103] [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] [Received: 06/08/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023]
Abstract
The mast cell is an important cellular component that plays a crucial role in the crosstalk between innate and adaptive immune responses within the tumor microenvironment (TME). Recently, numerous studies have indicated that mast cells related to tumors play a dual role in regulating cancers, with conflicting results seemingly determined by the degranulation medium. As such, mast cells are an ignored but very promising potential target for cancer immunotherapy based on their immunomodulatory function. In this review, we present a comprehensive overview of the roles and mechanisms of mast cells in diverse cancer types. Firstly, we evaluated the infiltration density and location of mast cells on tumor progression. Secondly, mast cells are activated by the TME and subsequently release a range of inflammatory mediators, cytokines, chemokines, and lipid products that modulate their pro-or anti-tumor functions. Thirdly, activated mast cells engage in intercellular communication with other immune or stromal cells to modulate the immune status or promote tumor development. Finally, we deliberated on the clinical significance of targeting mast cells as a therapeutic approach to restrict tumor initiation and progression. Overall, our review aims to provide insights for future research on the role of mast cells in tumors and their potential as therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Xinxin Guo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China; Xiangnan University, Chenzhou, China
| | - Mingjun Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Peiyan Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xingchen Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.
| |
Collapse
|
13
|
Sah P, Zenewicz LA. Modulation of innate lymphoid cells by enteric bacterial pathogens. Front Immunol 2023; 14:1219072. [PMID: 37483638 PMCID: PMC10358831 DOI: 10.3389/fimmu.2023.1219072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Innate lymphoid cells (ILCs) are key regulators of tissue homeostasis, inflammation, and immunity to infections. ILCs rapidly respond to environmental cues such as cytokines, microbiota and invading pathogens which regulate their function and phenotype. Even though ILCs are rare cells, they are enriched at barrier surfaces such as the gastrointestinal (GI) tract, and they are often critical to the host's immune response to eliminate pathogens. On the other side of host-pathogen interactions, pathogenic bacteria also have the means to modulate these immune responses. Manipulation or evasion of the immune cells is often to the pathogen's benefit and/or to the detriment of competing microbiota. In some instances, specific bacterial virulence factors or toxins have been implicated in how the pathogen modulates immunity. In this review, we discuss the recent progress made towards understanding the role of non-cytotoxic ILCs during enteric bacterial infections, how these pathogens can modulate the immune response, and the implications these have on developing new therapies to combat infection.
Collapse
Affiliation(s)
| | - Lauren A. Zenewicz
- Department of Microbiology and Immunology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
14
|
Wu E, Zhu J, Ma Z, Tuo B, Terai S, Mizuno K, Li T, Liu X. Gastric alarmin release: A warning signal in the development of gastric mucosal diseases. Front Immunol 2022; 13:1008047. [PMID: 36275647 PMCID: PMC9583272 DOI: 10.3389/fimmu.2022.1008047] [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: 07/31/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Alarmins exist outside cells and are early warning signals to the immune system; as such, alarmin receptors are widely distributed on various immune cells. Alarmins, proinflammatory molecular patterns associated with tissue damage, are usually released into the extracellular space, where they induce immune responses and participate in the damage and repair processes of mucosal diseases.In the stomach, gastric alarmin release has been shown to be involved in gastric mucosal inflammation, antibacterial defense, adaptive immunity, and wound healing; moreover, this release causes damage and results in the development of gastric mucosal diseases, including various types of gastritis, ulcers, and gastric cancer. Therefore, it is necessary to understand the role of alarmins in gastric mucosal diseases. This review focuses on the contribution of alarmins, including IL33, HMGB1, defensins and cathelicidins, to the gastric mucosal barrier and their role in gastric mucosal diseases. Here, we offer a new perspective on the prevention and treatment of gastric mucosal diseases.
Collapse
Affiliation(s)
- Enqin Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shuji Terai
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenichi Mizuno
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Xuemei Liu, ; Taolang Li,
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Xuemei Liu, ; Taolang Li,
| |
Collapse
|
15
|
Zhang Y, Wang M, Zhang K, Zhang J, Yuan X, Zou G, Cao Z, Zhang C. 6'-O-Galloylpaeoniflorin attenuates Helicobacter pylori-associated gastritis via modulating Nrf2 pathway. Int Immunopharmacol 2022; 111:109122. [PMID: 35964411 DOI: 10.1016/j.intimp.2022.109122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 12/14/2022]
Abstract
As a common disease of the digestive system, chronic gastritis is inflammation of the gastric mucosa caused by various factors. Helicobacter pylori (H. pylori) is one of the main causes of chronic gastritis, which can lead to gastric mucosal damage and gland atrophy, thereby promoting gastrocarcinogenesis. Oxidative stress and the inflammatory response are important mechanisms of H. pylori-induced gastritis. 6'-O-Galloylpaeoniflorin (GPF) is a substance isolated from peony root with antioxidant and anti-inflammatory activities. However, its role and mechanism in the pathogenesis of H. pylori-induced chronic gastritis remain unclear. This study explored the effects of GPF on H. pylori-induced gastric mucosal oxidative stress and inflammation using flow cytometry, western blotting, real-time quantitative PCR, and immunohistochemistry. We found that H. pylori infection increased oxidative stress and expression of inflammatory cytokines in vitro and in vivo and that these outcomes were inhibited by GPF. Furthermore, GPF activated nuclear factor erythroid-related factor-2 (Nrf2) and its downstream target genes in H. pylori-infected GES-1 cells and mice. The anti-inflammatory and antioxidant effects of GPF on H. pylori-infected cells were attenuated by an Nrf2 inhibitor. Taken together, these data suggest that GPF reduces H. pylori-induced gastric mucosa injury by activating Nrf2 signaling and that GPF is a potential candidate for the treatment of H. pylori-associated gastritis.
Collapse
Affiliation(s)
- Yun Zhang
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Maihuan Wang
- Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital, Beijing 100853, China
| | - Kebin Zhang
- Clinical Medical Research Center, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Junze Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Xinpu Yuan
- Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital, Beijing 100853, China
| | - Guijun Zou
- Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital, Beijing 100853, China
| | - Zhen Cao
- Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital, Beijing 100853, China.
| | - Chaojun Zhang
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of General Surgery, The First Medical Center of Chinese, PLA General Hospital, Beijing 100853, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
16
|
Levi-Schaffer F, Gibbs BF, Hallgren J, Pucillo C, Redegeld F, Siebenhaar F, Vitte J, Mezouar S, Michel M, Puzzovio PG, Maurer M. Selected recent advances in understanding the role of human mast cells in health and disease. J Allergy Clin Immunol 2022; 149:1833-1844. [PMID: 35276243 DOI: 10.1016/j.jaci.2022.01.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 11/17/2022]
Abstract
Mast cells are highly granular tissue-resident cells and key drivers of inflammation, particularly in allergies as well as in other inflammatory diseases. Most mast cell research was initially conducted in rodents but has increasingly shifted to the human system, with the advancement of research technologies and methodologies. Today we can analyze primary human cells including rare subpopulations, we can produce and maintain mast cells isolated from human tissues, and there are several human mast cell lines. These tools have substantially facilitated our understanding of their role and function in different organs in both health and disease. We can now define more clearly where human mast cells originate from, how they develop, which mediators they store, produce de novo, and release, how they are activated and by which receptors, and which neighbouring cells they interact with and by which mechanisms. Considerable progress has also been made regarding the potential contribution of mast cells to disease, which, in turn, has led to the development of novel approaches for preventing key pathogenic effects of mast cells, heralding the era of mast cell-targeted therapeutics. In this review, we present and discuss a selection of some of the most significant advancements and remaining gaps in our understanding of human mast cells during the last 25 years, with a focus on clinical relevance.
Collapse
Affiliation(s)
- Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Bernhard F Gibbs
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Carlo Pucillo
- Laboratory of Immunology, Department of Medicine, University of Udine, Udine, Italy
| | - Frank Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Frank Siebenhaar
- Institute for Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology, ITMP Allergology and Immunology, Berlin, Germany
| | - Joana Vitte
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France; IDESP, INSERM UA 11, Montpellier, France
| | | | - Moïse Michel
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France; Immunology Laboratory, CHU Nîmes, Nîmes, France
| | - Pier Giorgio Puzzovio
- Pharmacology and Experimental Therapeutics Unit, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marcus Maurer
- Institute for Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology, ITMP Allergology and Immunology, Berlin, Germany.
| |
Collapse
|
17
|
Yan ZB, Zhang JY, Lv YP, Tian WQ, Shan ZG, Mao FY, Liu YG, Chen WY, Wang P, Yang Y, Cheng P, Peng LS, Liao YL, Yue GY, Xu XL, Zhao YL, Lü MH, Zhuang Y. Helicobacter pylori-induced REDD1 modulates Th17 cell responses that contribute to gastritis. Clin Sci (Lond) 2021; 135:2541-2558. [PMID: 34730176 DOI: 10.1042/cs20210753] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/21/2021] [Accepted: 11/03/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Regulated in development and DNA damage responses-1 (REDD1) is a conserved and ubiquitous protein, which is induced in response to multiple stimuli. However, the regulation, function and clinical relevance of REDD1 in Helicobacter pylori-associated gastritis are presently unknown. APPROACH Immunohistochemistry, real-time PCR and Western blot analyses were performed to examine the levels of REDD1 in gastric samples from H. pylori-infected patients and mice. Gastric tissues from Redd1-/- and wildtype (WT, control) mice were examined for inflammation. Gastric epithelial cells (GECs), monocytes and T cells were isolated, stimulated and/or cultured for REDD1 regulation and functional assays. RESULTS REDD1 was increased in gastric mucosa of H. pylori-infected patients and mice. H. pylori induced GECs to express REDD1 via the phosphorylated cytotoxin associated gene A (cagA) that activated MAPKp38 pathway to mediate NF-κB directly binding to REDD1 promoter. Human gastric REDD1 increased with the severity of gastritis, and mouse REDD1 from non-marrow chimera-derived cells promoted gastric inflammation that was characterized by the influx of MHCII+ monocytes. Importantly, gastric inflammation, MHCII+ monocyte infiltration, IL-23 and IL-17A were attenuated in Redd1-/- mice. Mechanistically, REDD1 in GECs regulated CXCL1 production, which attracted MHCII+ monocytes migration by CXCL1-CXCR2 axis. Then H. pylori induced MHCII+ monocytes to secrete IL-23, which favored IL-17A-producing CD4+ cell (Th17 cell) polarization, thereby contributing to the development of H. pylori-associated gastritis. CONCLUSIONS The present study identifies a novel regulatory network involving REDD1, which collectively exert a pro-inflammatory effect within gastric microenvironment. Efforts to inhibit this REDD1-dependent pathway may prove valuable strategies in treating of H. pylori-associated gastritis.
Collapse
Affiliation(s)
- Zong-Bao Yan
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, 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 and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Yi-Pin Lv
- Department of Digestive Diseases, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Wen-Qing Tian
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Guo Shan
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, 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 and Laboratory Medicine, 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 and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Wan-Yan Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Pan Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Yun Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Liu-Sheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Ya-Ling Liao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Geng-Yu Yue
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiao-Lin Xu
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yong-Liang Zhao
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | - Yuan Zhuang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
- Department of Gastroenterology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Department of Gastroenterology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Jiangsu, China
| |
Collapse
|
18
|
miR-483-3p Promotes IL-33 Production from Fibroblast-Like Synoviocytes by Regulating ERK Signaling in Rheumatoid Arthritis. Inflammation 2021; 44:2302-2308. [PMID: 34160728 DOI: 10.1007/s10753-021-01503-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Abstract
Our previous studies have identified miR-483-3p to be highly expressed in synoviocytes from patients with rheumatoid arhtirits (RA); however, its effects on inflammation of RA fibroblast-like synoviocytes (FLSs) have remained unclear. The expression of miR-483-3p and cytokines in RA FLSs was detected using quantitative real-time polymerase chain reaction. Enzyme-linked immunosorbent was conducted to determine interleukin (IL)-33 production from RA FLSs. Western blotting was employed to quantify the levels of p-ERK and total ERK. Overexpressed miR-483-3p significantly increased the mRNA and protein expression of IL-33, but not of IL-27 or IL-34, in RA FLSs, whereas miR-483-3p suppression showed the opposite effects. Furthermore, miR-483-3p upregulation activated the ERK signaling pathway. The ERK signaling inhibitor PD98059 partly reversed the elevation of IL-33 levels mediated by miR-483-3p overexpression. Our results reveal that miR-483-3p promotes IL-33 expression by regulating the ERK signaling pathway in RA FLSs. Thus, miR-483-3p may be a potential effective target for RA treatment.
Collapse
|
19
|
Cheng L, Luo QQ, Chen SL. The role of intestinal mast cell infiltration in irritable bowel syndrome. J Dig Dis 2021; 22:143-151. [PMID: 33511763 DOI: 10.1111/1751-2980.12971] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 01/17/2021] [Accepted: 01/25/2021] [Indexed: 12/11/2022]
Abstract
As an essential part of the immune system, mast cells (MCs) play an important role in the pathogenesis of irritable bowel syndrome (IBS). Accumulating evidence has identified altered MC count and density in intestinal mucosa of patients with IBS; however, conflicting findings yield inconsistent conclusions. Currently, most studies have suggested intestinal MC infiltration in IBS patients. Considering the pivotal role of MCs in IBS, it is necessary to achieve a better understanding about the pathological changes in the intestine. The risk factors for IBS, including dietary habits, psychological factors, infection, and dysbiosis, are implicated to induce intestinal MC infiltration. Mechanistically, food may trigger immune-related allergic reactions and affect the intestinal microbiota activity. Some exogenous pathogens and altered profile of commensal bacteria promote intestinal MC recruitment through promoted release of chemokines from epithelial cells or direct activation of the immune system. In addition, psychological factors may affect the microenvironment where MCs live. MCs have been proven to interact with the enteric neurons and other immunocytes, evidenced by the close proximity of MCs to neurons and regional altered immune system components. A variety of mediators released by the enteric neurons, immunocytes, and MCs per se, such as neurotrophins, neuropeptides, cytokines, and chemokines, may have stimulant effects on MCs by modulating the survival, proliferation, and recruitment process of MCs in the intestine. In this review, the associations between IBS and intestinal MC density and the underlying mechanisms are discussed.
Collapse
Affiliation(s)
- Li Cheng
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Qing Qing Luo
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Sheng Liang Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
20
|
Moreno-Ochoa M, Valencia M, Morales-Figueroa G, Moya-Camarena S. Association of cagA+ Helicobacter pylori strains with high urease activity and dyspepsia in Mexican adults. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO (ENGLISH EDITION) 2020. [DOI: 10.1016/j.rgmxen.2020.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
21
|
Doulberis M, Kountouras J, Rogler G. Reconsidering the "protective" hypothesis of Helicobacter pylori infection in eosinophilic esophagitis. Ann N Y Acad Sci 2020; 1481:59-71. [PMID: 32770542 DOI: 10.1111/nyas.14449] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/12/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Since its discovery, Helicobacter pylori (H. pylori) has attracted attention in the biomedical world with its numerous pathophysiologic implications, both gastrointestinal and systemic. Beyond its well-established carcinogenic properties, emerging evidence also supports "harmful" proinflammatory and neurodegenerative roles of H. pylori. On the other hand, H. pylori infection has been proposed to be "protective" against several diseases, such as asthma and gastroesophageal reflux disease. Eosinophilic esophagitis (EoE) is a relatively new, allergen/immune-mediated disease, which has also been linked to these considerations. Main arguments are a postulated shift of immune responses by H. pylori from T helper 2 (TH 2) to TH 1 polarization, as well as a potential decline of the H. pylori burden with the dramatic parallel rise of ΕοΕ: a series of observational studies reported an inverse association. In this review, we counter these arguments by providing further epidemiological data, which point out that this generalization might be rather incomplete. We also discuss the limitations of the existing studies evaluating a possible association. Furthermore, we provide current evidence on common pathogenetic components, which share both entities. In summary, the claim that H. pylori is protective against EoE is rather incomplete, and further mechanistic studies are necessary to elucidate a possible association.
Collapse
Affiliation(s)
- Michael Doulberis
- Department of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland.,Second Medical Clinic, Faculty of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Jannis Kountouras
- Second Medical Clinic, Faculty of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Macedonia, Greece
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Liu YG, Teng YS, Shan ZG, Cheng P, Hao CJ, Lv YP, Mao FY, Yang SM, Chen W, Zhao YL, You N, Zou QM, Zhuang Y. Arrestin domain containing 3 promotes Helicobacter pylori-associated gastritis by regulating protease-activated receptor 1. JCI Insight 2020; 5:135849. [PMID: 32634127 PMCID: PMC7455081 DOI: 10.1172/jci.insight.135849] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 07/01/2020] [Indexed: 12/20/2022] Open
Abstract
Arrestin domain containing 3 (ARRDC3) represents a newly discovered α-arrestin involved in obesity, inflammation, and cancer. Here, we demonstrate a proinflammation role of ARRDC3 in Helicobacter pylori–associated gastritis. Increased ARRDC3 was detected in gastric mucosa of patients and mice infected with H. pylori. ARRDC3 in gastric epithelial cells (GECs) was induced by H. pylori, regulated by ERK and PI3K-AKT pathways in a cagA-dependent manner. Human gastric ARRDC3 correlated with the severity of gastritis, and mouse ARRDC3 from non-BM–derived cells promoted gastric inflammation. This inflammation was characterized by the CXCR2-dependent influx of CD45+CD11b+Ly6C–Ly6G+ neutrophils, whose migration was induced via the ARRDC3-dependent production of CXCL2 by GECs. Importantly, gastric inflammation was attenuated in Arrdc3–/– mice but increased in protease-activated receptor 1–/– (Par1–/–) mice. Mechanistically, ARRDC3 in GECs directly interacted with PAR1 and negatively regulated PAR1 via ARRDC3-mediated lysosomal degradation, which abrogated the suppression of CXCL2 production and following neutrophil chemotaxis by PAR1, thereby contributing to the development of H. pylori–associated gastritis. This study identifies a regulatory network involving H. pylori, GECs, ARRDC3, PAR1, and neutrophils, which collectively exert a proinflammatory effect within the gastric microenvironment. Efforts to inhibit this ARRDC3-dependent pathway may provide valuable strategies in treating of H. pylori–associated gastritis. A regulatory network including H. pylori, GECs, ARRDC3, PAR1, and neutrophils collectively exerts a pro-inflammatory effect within the gastric microenvironment.
Collapse
Affiliation(s)
- Yu-Gang Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China.,Department of Clinical Laboratory, the General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yong-Sheng Teng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Zhi-Guo Shan
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, and
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, 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 and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Yi-Pin Lv
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, 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 and Laboratory Medicine, 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 of Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Yong-Liang Zhao
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, and
| | - Nan You
- Department of Hepatobiliary Surgery, XinQiao Hospital, 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 and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Yuan Zhuang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
23
|
Eissmann MF, Buchert M, Ernst M. IL33 and Mast Cells-The Key Regulators of Immune Responses in Gastrointestinal Cancers? Front Immunol 2020; 11:1389. [PMID: 32719677 PMCID: PMC7350537 DOI: 10.3389/fimmu.2020.01389] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
The Interleukin (IL-)1 family IL33 is best known for eliciting type 2 immune responses by stimulating mast cells (MCs), regulatory T-cells (Tregs), innate lymphoid cells (ILCs) and other immune cells. MCs and IL33 provide critical control of immunological and epithelial homeostasis in the gastrointestinal (GI) tract. Meanwhile, the role of MCs in solid malignancies appears tissue-specific with both pro and anti-tumorigenic activities. Likewise, IL33 signaling significantly shapes immune responses in the tumor microenvironment, but these effects remain often dichotomous when assessed in experimental models of cancer. Thus, the balance between tumor suppressing and tumor promoting activities of IL33 are highly context dependent, and most likely dictated by the mixture of cell types responding to IL33. Adding to this complexity is the promiscuous nature by which MCs respond to cytokines other than IL33 and release chemotactic factors that recruit immune cells into the tumor microenvironment. In this review, we integrate the outcomes of recent studies on the role of MCs and IL33 in cancer with our own observations in the GI tract. We propose a working model where the most abundant IL33 responsive immune cell type is likely to dictate an overall tumor-supporting or tumor suppressing outcome in vivo. We discuss how these opposing responses affect the therapeutic potential of targeting MC and IL33, and highlight the caveats and challenges facing our ability to effectively harness MCs and IL33 biology for anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| |
Collapse
|
24
|
Satoh-Takayama N, Kato T, Motomura Y, Kageyama T, Taguchi-Atarashi N, Kinoshita-Daitoku R, Kuroda E, Di Santo JP, Mimuro H, Moro K, Ohno H. Bacteria-Induced Group 2 Innate Lymphoid Cells in the Stomach Provide Immune Protection through Induction of IgA. Immunity 2020; 52:635-649.e4. [PMID: 32240600 DOI: 10.1016/j.immuni.2020.03.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/15/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022]
Abstract
The intestinal microbiota shapes and directs immune development locally and systemically, but little is known about whether commensal microbes in the stomach can impact their immunological microenvironment. Here, we report that group 2 innate lymphoid cells (ILC2s) were the predominant ILC subset in the stomach and show that their homeostasis and effector functions were regulated by local commensal communities. Microbes elicited interleukin-7 (IL-7) and IL-33 production in the stomach, which in turn triggered the propagation and activation of ILC2. Stomach ILC2s were also rapidly induced following infection with Helicobacter pylori. ILC2-derived IL-5 resulted in the production of IgA, which coated stomach bacteria in both specific pathogen-free (SPF) and H. pylori-infected mice. Our study thus identifies ILC2-dependent IgA response that is regulated by the commensal microbiota, which is implicated in stomach protection by eliminating IgA-coated bacteria including pathogenic H. pylori.
Collapse
Affiliation(s)
- Naoko Satoh-Takayama
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, RIKEN Yokohama, Kanagawa 230-0045, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University Yokohama, Kanagawa 230-0045, Japan.
| | - Tamotsu Kato
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, RIKEN Yokohama, Kanagawa 230-0045, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University Yokohama, Kanagawa 230-0045, Japan
| | - Yasutaka Motomura
- Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University Suita, Osaka 565-0871, Japan; Laboratory for Innate Immune Systems, Center for Integrative Medical Sciences, RIKEN Yokohama, Kanagawa 230-0045, Japan
| | - Tomoko Kageyama
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, RIKEN Yokohama, Kanagawa 230-0045, Japan
| | - Naoko Taguchi-Atarashi
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, RIKEN Yokohama, Kanagawa 230-0045, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University Yokohama, Kanagawa 230-0045, Japan
| | - Ryo Kinoshita-Daitoku
- Department of Infection Microbiology, Research Institute for Microbial Diseases, Osaka University Suita, Osaka 565-0871, Japan
| | - Eisuke Kuroda
- Department of Infection Microbiology, Research Institute for Microbial Diseases, Osaka University Suita, Osaka 565-0871, Japan
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris 75015, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1223 Paris 75013, France
| | - Hitomi Mimuro
- Department of Infection Microbiology, Research Institute for Microbial Diseases, Osaka University Suita, Osaka 565-0871, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University Suita, Osaka 565-0871, Japan; Laboratory for Innate Immune Systems, Center for Integrative Medical Sciences, RIKEN Yokohama, Kanagawa 230-0045, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, RIKEN Yokohama, Kanagawa 230-0045, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University Yokohama, Kanagawa 230-0045, Japan; Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology Ebina, Kanagawa 243-0435, Japan.
| |
Collapse
|
25
|
Moreno-Ochoa MF, Valencia ME, Morales-Figueroa GG, Moya-Camarena SY. Association of cagA+ Helicobacter pylori strains with high urease activity and dyspepsia in Mexican adults. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2020; 85:404-409. [PMID: 32224002 DOI: 10.1016/j.rgmx.2019.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/23/2019] [Accepted: 09/09/2019] [Indexed: 11/26/2022]
Abstract
INTRODUCTION AND AIMS Helicobacter pylori (H. pylori) is associated with a higher risk of peptic ulcer and gastric cancer. The sole presence of the bacterium is not a determinant of clinical outcome, but rather the interaction of strain type and host factors determines the risk of disease. Our aim was to study the association between bacterial load, strain type, and gastric symptoms in H. pylori-positive subjects. MATERIALS AND METHODS In a community survey, a diagnostic 13C-urea breath test for H. pylori was performed on 302 volunteers that were not taking antibiotics, antacids, or proton pump inhibitors one month prior to the test. The breath test produced 25 H. pylori-positive subjects, between 25-74 years of age, who then took a gastric symptoms survey and were tested for the presence of the cagA genotype in gastric juice, using the Entero-test®. Bacterial load was determined as a measure of urease activity, utilizing the delta over baseline value, obtained in the 13C-urea breath test. RESULTS A total of 48% of the H. pylori-positive subjects were cagA+. A positive association was found between cagA status and high gastric urease activity (P<.0001) and the latter was significantly associated with the presence of symptoms (P<.0001). CONCLUSION Gastric urease activity was strongly associated with dyspeptic symptoms and cagA+ H. pylori. Elevated 13C-delta over baseline values could be used as indicators of a higher risk for gastric disease.
Collapse
Affiliation(s)
- M F Moreno-Ochoa
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Sonora, México
| | - M E Valencia
- Departamento de Nutrición Pública y Salud, Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Sonora, México
| | - G G Morales-Figueroa
- Departamento de Nutrición Pública y Salud, Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Sonora, México
| | - S Y Moya-Camarena
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Sonora, México.
| |
Collapse
|
26
|
Helicobacter pylori Induces IL-33 Production and Recruits ST-2 to Lipid Rafts to Exacerbate Inflammation. Cells 2019; 8:cells8101290. [PMID: 31640262 PMCID: PMC6830106 DOI: 10.3390/cells8101290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 12/30/2022] Open
Abstract
Helicobacter pylori colonizes human gastric epithelial cells and contributes to the development of several gastrointestinal disorders. Interleukin (IL)-33 is involved in various immune responses, with reported proinflammatory and anti-inflammatory effects, which may be associated with colitis and colitis-associated cancer. IL-33 induces the inflammatory cascade through its receptor, suppression of tumorigenicity-2 (ST-2). Binding of IL-33 to membrane-bound ST-2 (mST-2) recruits the IL-1 receptor accessory protein (IL-1RAcP) and activates intracellular signaling pathways. However, whether IL-33/ST-2 is triggered by H. pylori infection and whether this interaction occurs in lipid rafts remain unclear. Our study showed that both IL-33 and ST-2 expression levels were significantly elevated in H. pylori-infected cells. Confocal microscopy showed that ST-2 mobilized into the membrane lipid rafts during infection. Depletion of membrane cholesterol dampened H. pylori-induced IL-33 and IL-8 production. Furthermore, in vivo studies revealed IL-33/ST-2 upregulation, and severe leukocyte infiltration was observed in gastric tissues infected with H. pylori. Together, these results demonstrate that ST-2 recruitment into the lipid rafts serves as a platform for IL-33-dependent H. pylori infection, which aggravates inflammation in the stomach.
Collapse
|
27
|
Bassagh A, Jafarzadeh A, Kazemipour N, Nemati M, Aminizadeh N, Larussa T, Ghazizadeh M, Abasi MH, Mirkamandar E. Decreased circulating interleukin-33 concentration in Helicobacter pylori-infected patients with peptic ulcer: Evaluation of its association with a cytokine gene polymorphism, gender of patients and bacterial virulence factor CagA. Microb Pathog 2019; 136:103708. [PMID: 31491552 DOI: 10.1016/j.micpath.2019.103708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 01/10/2023]
Abstract
IL-33 has powerful immunoregulatory activities such as reinforcement of Th2 cell responses. The aim was to assess the circulating IL-33 levels and IL-33 rs1929992 polymorphism in H. pylori-infected peptic ulcer (PU) patients and asymptomatic (AS) subjects. Blood samples were obtained from 100 PU patients, 100 AS subjects and 100 uninfected individuals. Circulating IL-33 levels were detected by ELISA. After DNA extraction, the IL-33 rs1929992 polymorphism was determined using PCR-RFLP method. Serum IL-33 quantities were significantly lower in PU patients compared with AS and uninfected groups. IL-33 levels were higher in AS subjects compared with uninfected group. In PU, AS and uninfected groups, IL-33 levels were significantly higher in women than men. In PU and AS groups, the CagA+H. pylori-infected subjects exhibit higher IL-33 levels compared with carriers of CagA-H. pylori strains. In PU patients, the frequency of genotype GG and allele G at IL-33 rs1929992 was significantly higher compared with all healthy subjects (AS + uninfected groups). The presence of genotypes GG and AG, and allele G in rs1929992 conferred greater risk for PU. In whole H. pylori-infected population (PU + AS groups), IL-33 levels in individuals with genotype AA or allele A at rs1929992 were higher than subjects with GG genotype or allele G. The reduced IL-33 production could contribute to the PU development during H. pylori infection. The IL-33 levels may be affected by individual gender, rs1929992 polymorphism, and the CagA status of bacteria. The rs1929992-related GG genotype and G allele may be associated with PU development.
Collapse
Affiliation(s)
- Arezoo Bassagh
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran; Department of Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Nadia Kazemipour
- Department of Microbiology, Islamic Azad University of Kerman Branch, Kerman, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Najmeh Aminizadeh
- Department of Anatomy, Islamic Azad University of Kerman Branch, Kerman, Iran
| | - Tiziana Larussa
- Department of Health Sciences, University of Catanzaro "Magna Graecia", 88100, Catanzaro, Italy
| | | | | | - Ehsan Mirkamandar
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
28
|
Gang Liu Y, Teng YS, Cheng P, Kong H, Lv PY, Mao FY, Wu XL, Hao CJ, Chen W, Yang SM, Zhang JY, Peng LS, Wang TT, Han B, Ma Q, Zou QM, Zhuang AY. Abrogation of cathepsin C by
Helicobacter pylori
impairs neutrophil activation to promote gastric infection. FASEB J 2018; 33:5018-5033. [DOI: 10.1096/fj.201802016rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yu Gang Liu
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Yong Sheng Teng
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Ping Cheng
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Hui Kong
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Pin Yi Lv
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Fang Yuan Mao
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Xiao Long Wu
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Chuan Jie Hao
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Weisan Chen
- La Trobe Institute of Molecular ScienceLa Trobe University Bundoora Victoria Australia
| | - Shi Ming Yang
- Department of GastroenterologyXinQiao HospitalThird Military Medical University Chongqing China
| | - Jin Yu Zhang
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Liu Sheng Peng
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Ting Ting Wang
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - Bin Han
- Department of PharmacyAffiliated Hospital of North Sichuan Medical College Nanchong China
| | - Qiang Ma
- Department of Clinical LaboratoryAffiliated Hospital of North Sichuan Medical College Nanchong China
| | - Quan Ming Zou
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| | - And Yuan Zhuang
- Department of Microbiology and Biochemical PharmacyNational Engineering Research Centre of Immunological ProductsCollege of Pharmacy Chongqing China
| |
Collapse
|