1
|
Shen G, Wang Q, Li Z, Xie J, Han X, Wei Z, Zhang P, Zhao S, Wang X, Huang X, Xu M. Bridging Chronic Inflammation and Digestive Cancer: The Critical Role of Innate Lymphoid Cells in Tumor Microenvironments. Int J Biol Sci 2024; 20:4799-4818. [PMID: 39309440 PMCID: PMC11414386 DOI: 10.7150/ijbs.96338] [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: 03/16/2024] [Accepted: 08/09/2024] [Indexed: 09/25/2024] Open
Abstract
The incidence and mortality of digestive system-related cancers have always been high and attributed to the heterogeneity and complexity of the immune microenvironment of the digestive system. Furthermore, several studies have shown that chronic inflammation in the digestive system is responsible for cancer incidence; therefore, controlling inflammation is a potential strategy to stop the development of cancer. Innate Lymphoid Cells (ILC) represent a heterogeneous group of lymphocytes that exist in contrast to T cells. They function by interacting with cytokines and immune cells in an antigen-independent manner. In the digestive system cancer, from the inflammatory phase to the development, migration, and metastasis of tumors, ILC have been found to interact with the immune microenvironment and either control or promote these processes. The conventional treatments for digestive tumors have limited efficacy, therefore, ILC-associated immunotherapies are promising strategies. This study reviews the characterization of different ILC subpopulations, how they interact with and influence the immune microenvironment as well as chronic inflammation, and their promotional or inhibitory role in four common digestive system tumors, including pancreatic, colorectal, gastric, and hepatocellular cancers. In particular, the review emphasizes the role of ILC in associating chronic inflammation with cancer and the potential for enhanced immunotherapy with cytokine therapy and adoptive immune cell therapy.
Collapse
Affiliation(s)
- Guanliang Shen
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xinda Han
- Xinglin College, Nantong University, Nantong, Jiangsu, China
| | - Zehao Wei
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Songyun Zhao
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Xiumei Wang
- Affiliated Cancer Hospital of Inner Mongolia Medical University, 010020, Inner Mongolia, China
| | | | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
- Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
2
|
Gonciarz W, Płoszaj P, Chmiela M. Mycobacterium bovis BCG reverses deleterious effects of H. pylori components towards gastric barrier cells in vitro. Biomed Pharmacother 2024; 178:117193. [PMID: 39067167 DOI: 10.1016/j.biopha.2024.117193] [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: 05/29/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Mycobacterium bovis (M. bovis) Bacillus Calmette-Guerin (BCG) strain used in immunotherapy of bladder cancer (onco-BCG) due to its acid tolerance can be a candidate for prevention or reversion of deleterious effects towards gastric cell barrier initiated by gastric pathogen Helicobacter pylori (Hp) with high resistance to commonly used antibiotics. Colonization of gastric mucosa by Hp promotes oxidative stress, apoptosis resulting in the gastric barrier damage. The aim of this study was to examine the ability of onco-BCG bacilli to control the Hp driven gastric damage using the model of Cavia porcellus primary gastric epithelial cells or fibroblasts in vitro. These cells were treated with Hp surface antigens (glycine acid extract-GE or lipopolysaccharide-LPS) alone or with onco-BCG bacilli and evaluated for cell apoptosis and proliferation in conjunction with the level of soluble lipid peroxidation marker (s4HNE). The cell migration was determined by "wound healing assay", while cytokine response of cells, including interleukin (IL)-33, IL-1β, IL-8 and tumor necrosis factor alpha (TNF-α), by the ELISA. The apoptosis of cells pulsed in vitro with Hp surface components present in GE or with LPS was reduced after exposure of cells to mycobacteria. Similarly, the cell regeneration which was diminished by Hp LPS has been improved in response to mycobacteria. This study reveals that vaccine mycobacteria may reduce gastric barrier damage induced by Hp infection.
Collapse
Affiliation(s)
- Weronika Gonciarz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
| | - Patrycja Płoszaj
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Magdalena Chmiela
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
3
|
Wang X, Zhao G, Shao S, Yao Y. Helicobacter pylori triggers inflammation and oncogenic transformation by perturbing the immune microenvironment. Biochim Biophys Acta Rev Cancer 2024; 1879:189139. [PMID: 38897421 DOI: 10.1016/j.bbcan.2024.189139] [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: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024]
Abstract
The immune microenvironment plays a critical regulatory role in the pathogenesis of Helicobacter pylori (H. pylori). Understanding the mechanisms that drive the transition from chronic inflammation to cancer may provide new insights for early detection of gastric cancer. Although chronic inflammation is frequent in precancerous gastric conditions, the monitoring function of the inflammatory microenvironment in the progression from H. pylori-induced chronic inflammation to gastric cancer remains unclear. This literature review summarizes significant findings on how H. pylori triggers inflammatory responses and facilitates cancer development through the immune microenvironment. Furthermore, the implications for future research and clinical applications are also addressed. The review is divided into four main sections: inflammatory response and immune evasion mechanisms induced by H. pylori, immune dysregulation associated with gastric cancer, therapeutic implications, and future perspectives on H. pylori-induced gastric carcinogenesis with a focus on the immune microenvironment.
Collapse
Affiliation(s)
- Xiuping Wang
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, Jiangsu, China
| | - Guang Zhao
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, Jiangsu, China; Department of Emergency Medicine, Kunshan Hospital Affiliated to Jiangsu University, Kunshan 215300, Jiangsu, China
| | - Shihe Shao
- School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| | - Yongliang Yao
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, Jiangsu, China.
| |
Collapse
|
4
|
Santacroce L, Topi S, Bottalico L, Charitos IA, Jirillo E. Current Knowledge about Gastric Microbiota with Special Emphasis on Helicobacter pylori-Related Gastric Conditions. Curr Issues Mol Biol 2024; 46:4991-5009. [PMID: 38785567 PMCID: PMC11119845 DOI: 10.3390/cimb46050299] [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: 03/31/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
The gastric milieu, because of its very low acidic pH, is very harsh for bacterial growth. The discovery of Helicobacter pylori (H.p.) has opened a new avenue for studies on the gastric microbiota, thus indicating that the stomach is not a sterile environment. Nowadays, new technologies of bacterial identification have demonstrated the existence of other microorganisms in the gastric habitat, which play an important role in health and disease. This bacterium possesses an arsenal of compounds which enable its survival but, at the same time, damage the gastric mucosa. Toxins, such as cytotoxin-associated gene A, vacuolar cytotoxin A, lipopolysaccharides, and adhesins, determine an inflammatory status of the gastric mucosa which may become chronic, ultimately leading to a gastric carcinoma. In the initial stage, H.p. persistence alters the gastric microbiota with a condition of dysbiosis, predisposing to inflammation. Probiotics and prebiotics exhibit beneficial effects on H.p. infection, and, among them, anti-inflammatory, antioxidant, and antibacterial activities are the major ones. Moreover, the association of probiotics with prebiotics (synbiotics) to conventional anti-H.p. therapy contributes to a more efficacious eradication of the bacterium. Also, polyphenols, largely present in the vegetal kingdom, have been demonstrated to alleviate H.p.-dependent pathologies, even including the inhibition of tumorigenesis. The gastric microbiota composition in health and disease is described. Then, cellular and molecular mechanisms of H.p.-mediated damage are clarified. Finally, the use of probiotics, prebiotics, and polyphenols in experimental models and in patients infected with H.p. is discussed.
Collapse
Affiliation(s)
- Luigi Santacroce
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| | - Skender Topi
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania; (S.T.)
| | - Lucrezia Bottalico
- Department of Clinical Disciplines, University ‘Alexander Xhuvani’ of Elbasan, 3001 Elbasan, Albania; (S.T.)
| | - Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, Institute of Bari, 70124 Bari, Italy;
| | - Emilio Jirillo
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy;
| |
Collapse
|
5
|
Tong QY, Pang MJ, Hu XH, Huang XZ, Sun JX, Wang XY, Burclaff J, Mills JC, Wang ZN, Miao ZF. Gastric intestinal metaplasia: progress and remaining challenges. J Gastroenterol 2024; 59:285-301. [PMID: 38242996 DOI: 10.1007/s00535-023-02073-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/26/2023] [Indexed: 01/21/2024]
Abstract
Most gastric cancers arise in the setting of chronic inflammation which alters gland organization, such that acid-pumping parietal cells are lost, and remaining cells undergo metaplastic change in differentiation patterns. From a basic science perspective, recent progress has been made in understanding how atrophy and initial pyloric metaplasia occur. However, pathologists and cancer biologists have long been focused on the development of intestinal metaplasia patterns in this setting. Arguably, much less progress has been made in understanding the mechanisms that lead to the intestinalization seen in chronic atrophic gastritis and pyloric metaplasia. One plausible explanation for this disparity lies in the notable absence of reliable and reproducible small animal models within the field, which would facilitate the investigation of the mechanisms underlying the development of gastric intestinal metaplasia (GIM). This review offers an in-depth exploration of the current state of research in GIM, shedding light on its pivotal role in tumorigenesis. We delve into the histological subtypes of GIM and explore their respective associations with tumor formation. We present the current repertoire of biomarkers utilized to delineate the origins and progression of GIM and provide a comprehensive survey of the available, albeit limited, mouse lines employed for modeling GIM and engage in a discussion regarding potential cell lineages that serve as the origins of GIM. Finally, we expound upon the myriad signaling pathways recognized for their activity in GIM and posit on their potential overlap and interactions that contribute to the ultimate manifestation of the disease phenotype. Through our exhaustive review of the progression from gastric disease to GIM, we aim to establish the groundwork for future research endeavors dedicated to elucidating the etiology of GIM and developing strategies for its prevention and treatment, considering its potential precancerous nature.
Collapse
Affiliation(s)
- Qi-Yue Tong
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Min-Jiao Pang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Xiao-Hai Hu
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Xuan-Zhang Huang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Jing-Xu Sun
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Xin-Yu Wang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Joseph Burclaff
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Department of Medicine, Departments of Pathology and Immunology, Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
| | - Zhi-Feng Miao
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
6
|
Privitera G, Williams JJ, De Salvo C. The Importance of Th2 Immune Responses in Mediating the Progression of Gastritis-Associated Metaplasia to Gastric Cancer. Cancers (Basel) 2024; 16:522. [PMID: 38339273 PMCID: PMC10854712 DOI: 10.3390/cancers16030522] [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: 12/31/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Gastric cancer is one of the leading causes of cancer deaths worldwide, with chronic gastritis representing the main predisposing factor initiating the cascade of events leading to metaplasia and eventually progressing to cancer. A widely accepted classification distinguishes between autoimmune and environmental atrophic gastritis, mediated, respectively, by T cells promoting the destruction of the oxyntic mucosa, and chronic H. pylori infection, which has also been identified as the major risk factor for gastric cancer. The original dogma posits Th1 immunity as a main causal factor for developing gastritis and metaplasia. Recently, however, it has become evident that Th2 immune responses play a major role in the events causing chronic inflammation leading to tumorigenesis, and in this context, many different cell types and cytokines are involved. In particular, the activity of cytokines, such as IL-33 and IL-13, and cell types, such as mast cells, M2 macrophages and eosinophils, are intertwined in the process, promoting chronic gastritis-dependent and more diffuse metaplasia. Herein, we provide an overview of the critical events driving the pathology of this disease, focusing on the most recent findings regarding the importance of Th2 immunity in gastritis and gastric metaplasia.
Collapse
Affiliation(s)
- Giuseppe Privitera
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (G.P.); (J.J.W.)
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Joseph J. Williams
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (G.P.); (J.J.W.)
| | - Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (G.P.); (J.J.W.)
| |
Collapse
|
7
|
Xie D, Wu C, Wang D, Nisma Lena BA, Liu N, Ye G, Sun M. Wei-fu-chun tablet halted gastric intestinal metaplasia and dysplasia associated with inflammation by regulating the NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117020. [PMID: 37567428 DOI: 10.1016/j.jep.2023.117020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chi006Eese herbal medicine Weifuchun Tablets (WFC) approved by the State Food and Drug Administration in 1982 has been widely used in treating a variety of chronic stomach disorders including Chronic atrophic gastritis (CAG) and Gastric precancerous lesions in China clinically. This study aimed to investigate the efficacy and potential mechanism of WFC in treating Gastric intestinal metaplasia (GIM) and Gastric dysplasia (GDys). MATERIALS AND METHODS Rat GIM and GDys established by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) combined with hot paste, ethanol injury, and intermittent fasting were intervened by WFC. Body weight, histopathology, pH of gastric acid, pepsin activity, intestinal metaplasia index and inflammation were detected. Rat bone marrow derived macrophages (BMDMs) pretreated with WFC were stimulated by LPS. Inflammatory factors and the nuclear factor-kappa B (NF-κB) pathway were assessed. GES-1 cells pretreated by WFC were stimulated by MNNG and TNF-α, intestinal metaplasia index, the NF-κB pathway and interaction between P65 and CDX2 were detected. RESULTS WFC improved rat body weight, histopathology, pH value of gastric acid, activity of gastric pepsin, intestinal metaplasia (CDX2), inflammation (IL-1β, IL-6 and TNF-α), macrophage aggregation (CD68) in gastric mucosa in rat GIM and GDys. WFC inhibited inflammation (IL-1β and TNF-α) by inactivating the NF-κB pathway. WFC reduced the expression of CDX2 by inhibiting the binding of CDX2 promoter TSS upstream region with p65. CONCLUSION WFC blocked GIM and GDys associated with inflammation by regulating the NF-κB pathway.
Collapse
Affiliation(s)
- Dong Xie
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao Wu
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Wang
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bahaji Azami Nisma Lena
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ningning Liu
- Department of Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guan Ye
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, China.
| | - Mingyu Sun
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
8
|
Peng R, Xu C, Zhang L, Liu X, Peng D, Chen X, Liu D, Li R. M2 macrophages participate in ILC2 activation induced by Helicobacter pylori infection. Gut Microbes 2024; 16:2347025. [PMID: 38693666 PMCID: PMC11067991 DOI: 10.1080/19490976.2024.2347025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/19/2024] [Indexed: 05/03/2024] Open
Abstract
Helicobacter pylori (H. pylori) causes a diversity of gastric diseases. The host immune response evoked by H. pylori infection is complicated and can influence the development and progression of diseases. We have reported that the Group 2 innate lymphocytes (ILC2) were promoted and took part in building type-2 immunity in H. pylori infection-related gastric diseases. Therefore, in the present study, we aim to clarify how H. pylori infection induces the activation of ILC2. It was found that macrophages were necessary for activating ILC2 in H. pylori infection. Mechanistically, H. pylori infection up-regulated the expression of indoleamine 2,3-dioxygenase (IDO) in macrophages to induce M2 polarization, and the latter secreted the alarmin cytokine Thymic Stromal Lymphopoietin (TSLP) to arouse ILC2.
Collapse
Affiliation(s)
- Ruyi Peng
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan Province, China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha, Hunan Province, China
| | - Canxia Xu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan Province, China
| | - Linfang Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaoming Liu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, Hunan Province, China
| | - Dongzi Peng
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan Province, China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha, Hunan Province, China
| | - Xingcen Chen
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan Province, China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha, Hunan Province, China
| | - Deliang Liu
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan Province, China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha, Hunan Province, China
| | - Rong Li
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Research Center of Digestive Disease, Central South University, Changsha, Hunan Province, China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha, Hunan Province, China
| |
Collapse
|
9
|
Zhang S, Shen Y, Liu H, Zhu D, Fang J, Pan H, Liu W. Inflammatory microenvironment in gastric premalignant lesions: implication and application. Front Immunol 2023; 14:1297101. [PMID: 38035066 PMCID: PMC10684945 DOI: 10.3389/fimmu.2023.1297101] [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: 09/19/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Gastric precancerous lesions (GPL) are a major health concern worldwide due to their potential to progress to gastric cancer (GC). Understanding the mechanism underlying the transformation from GPL to GC can provide a fresh insight for the early detection of GC. Although chronic inflammation is prevalent in the GPL, how the inflammatory microenvironment monitored the progression of GPL-to-GC are still elusive. Inflammation has been recognized as a key player in the progression of GPL. This review aims to provide an overview of the inflammatory microenvironment in GPL and its implications for disease progression and potential therapeutic applications. We discuss the involvement of inflammation in the progression of GPL, highlighting Helicobacter pylori (H. pylori) as a mediator for inflammatory microenvironment and a key driver to GC progression. We explore the role of immune cells in mediating the progression of GPL, and focus on the regulation of inflammatory molecules in this disease. Furthermore, we discuss the potential of targeting inflammatory pathways for GPL. There are currently no specific drugs for GPL treatment, but traditional Chinese Medicine (TCM) and natural antioxidants, known as antioxidant and anti-inflammatory properties, exhibit promising effects in suppressing or reversing the progression of GPL. Finally, the challenges and future perspectives in the field are proposed. Overall, this review highlights the central role of the inflammatory microenvironment in the progression of GPL, paving the way for innovative therapeutic approaches in the future.
Collapse
Affiliation(s)
- Shengxiong Zhang
- Rehabilitation Department, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, China
- Department of Spleen and Stomach, GuangZhou Tianhe District Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Shen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Di Zhu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiansong Fang
- Science and Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huafeng Pan
- Science and Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
10
|
Cicek B, Danısman B, Yildirim S, Yuce N, Nikitovic D, Bolat I, Kuzucu M, Ceyran E, Bardas E, Golokhvast KS, Tsatsakis A, Taghizadehghalehjoughi A. Flavonoid-Rich Sambucus nigra Berry Extract Enhances Nrf2/HO-1 Signaling Pathway Activation and Exerts Antiulcerative Effects In Vivo. Int J Mol Sci 2023; 24:15486. [PMID: 37895164 PMCID: PMC10607857 DOI: 10.3390/ijms242015486] [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: 08/24/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Sambucus nigra (SN) berry extract is characterized by high antioxidant and anti-inflammatory activity. The current study aimed to investigate the effect of SN berry extract against indomethacin (IND)-induced gastric ulcer in rats and the mechanism involved. SN berry extract alleviated IND-induced gastric ulcers, as shown by assessing pathological manifestations in the gastric mucosa. These protective effects are attributed to attenuated oxidative damage to the gastric mucosa, correlated to increased activity of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), enhanced glutathione (GSH) levels, total antioxidant capacity (TAC), and upregulation of the Nrf2/HO-1 cascade. Moreover, oxidative stress markers, including malondialdehyde (MDA) and total oxidant status (TOS), were downregulated in SN-extract-treated animals. Furthermore, SN berry extract suppressed gastric mucosal inflammation by downregulating interleukin (IL)-33, IL-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) levels, and attenuating myeloperoxidase (MPO) activity. The protective effects of SN berry extract were similar to those exerted by esomeprazole (ESO), an acid-secretion-suppressive drug. In conclusion, SN berry extract has antiulcerative effects, alleviating oxidative stress and inflammation.
Collapse
Affiliation(s)
- Betul Cicek
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey; (B.C.); (E.B.)
| | - Betul Danısman
- Department of Biophysics, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey;
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary, Atatürk University, 25240 Erzurum, Turkey; (S.Y.); (I.B.)
| | - Neslihan Yuce
- Department of Medical Biochemistry, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey;
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Ismail Bolat
- Department of Pathology, Faculty of Veterinary, Atatürk University, 25240 Erzurum, Turkey; (S.Y.); (I.B.)
| | - Mehmet Kuzucu
- Department of Biology, Faculty of Arts and Sciences, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey;
| | - Ertuğrul Ceyran
- Central Research and Application Laboratory, Agri Ibrahim Cecen University, 41000 Agri, Turkey;
| | - Ebru Bardas
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey; (B.C.); (E.B.)
| | - Kirill S. Golokhvast
- Siberian Federal Scientific Centre of Agrobiotechnology RAS, 2B Centralnaya Street, 630501 Krasnoobsk, Russia;
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Ali Taghizadehghalehjoughi
- Department of Medical Pharmacology, Faculty of Medicine, Bilecik Şeyh Edebali University, 11230 Bilecik, Turkey;
| |
Collapse
|
11
|
Chatterjee A, Azevedo-Martins JM, Stachler MD. Interleukin-33 as a Potential Therapeutic Target in Gastric Cancer Patients: Current Insights. Onco Targets Ther 2023; 16:675-687. [PMID: 37583706 PMCID: PMC10424681 DOI: 10.2147/ott.s389120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/06/2023] [Indexed: 08/17/2023] Open
Abstract
Gastric cancer is a significant global health problem as it is the fifth most prevalent cancer worldwide and the fourth leading cause of cancer-related mortality. While cytotoxic chemotherapy remains the primary treatment for advanced GC, response rates are limited. Recent progresses, focused on molecular signalling within gastric cancer, have ignited new hope for potential therapeutic targets that may improve survival and/or reduce the toxic effects of traditional therapies. Carcinomas are generally initiated when critical regulatory genes get mutated, but the progression to malignancy is usually supported by the non-neoplastic cells that create a conducive environment for transformation and progression to occur. Interleukin 33 (IL-33) functions as a dual activity cytokine as it is also a nuclear factor. IL-33 is usually present in the nuclei of the cells. Upon tissue damage, it is released into the extracellular space and binds to its receptor, suppression of tumorigenicity 2 (ST2) L, which is expressed on the membranes of the target cells. IL-33 signalling activates the T Helper 2 (Th2) immune response among other responses. Although the studies on the role of IL-33 in gastric cancer are still in the early stages, they have revealed potentially important (though sometimes conflicting) functions or roles in cancer development and progression. The pro-tumorigenic roles include induction and the recruitment of tumor-associated immune cells, promoting metaplasia progression, and inducing stem cell like and EMT properties in gastric cancer cells. Therapeutic interventions to disrupt these functions may provide a unique strategy for gastric cancer prevention and treatment. This review aims to provide a summary of the role of IL-33 in GC, state its multiple functions in relation to GC, and show potential avenues for promising therapeutic investigation.
Collapse
Affiliation(s)
- Annesha Chatterjee
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| | | | - Matthew D Stachler
- University of California San Francisco, Department of Pathology, San Francisco, CA, USA
| |
Collapse
|
12
|
Wernly S, Paar V, Völkerer A, Semmler G, Datz C, Lichtenauer M, Wernly B. sST2 Levels Show No Association with Helicobacter pylori Infection in Asymptomatic Patients: Implications for Biomarker Research. Dig Dis Sci 2023:10.1007/s10620-023-08005-0. [PMID: 37338618 PMCID: PMC10352442 DOI: 10.1007/s10620-023-08005-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023]
Abstract
INTRODUCTION Helicobacter pylori (H. pylori) is a prevalent stomach bacterium that can cause a range of clinical outcomes, including gastric cancer. In recent years, soluble suppression of tumorigenicity-2 (sST2) has gained attention as a biomarker associated with various diseases, such as gastric cancer. The purpose of this study was to explore the possible connection between H. pylori infection and sST2 levels in patients who do not exhibit symptoms. METHODS A total of 694 patients from the Salzburg Colon Cancer Prevention Initiative (Sakkopi) were included in the study. The prevalence of H. pylori infection was determined by histology, and sST2 levels were measured in serum samples. Clinical and laboratory parameters, such as age, sex, BMI, smoking status, hypertension, and metabolic syndrome, were also collected. RESULTS The median sST2 concentration was similar between patients with (9.62; 7.18-13.44 ng/mL; p = 0.66) and without (9.67; 7.08-13.06 ng/mL) H. pylori. Logistic regression analysis did not show any association (OR 1.00; 95%CI 0.97-1.04; p = 0.93) between sST2 levels and H. pylori infection, which remained so (aOR 0.99; 95%CI 0.95-1.03; p = 0.60) after adjustment for age, sex, educational status, and metabolic syndrome. In addition, sensitivity analyses stratified by age, sex, BMI, smoking status, educational status, and the concomitant diagnosis of metabolic syndrome could not show any association between sST2 levels and H. pylori infection. CONCLUSION The results indicate that sST2 may not serve as a valuable biomarker in the diagnosis and treatment of H. pylori infection. Our findings are of relevance for further research investigating sST2, as we could not find an influence of asymptomatic H. pylori infection on sST2 concentration. WHAT IS ALREADY KNOWN?: Soluble suppression of tumorigenicity-2 (sST2) has gained attention as a biomarker associated with various diseases, such as gastric cancer. WHAT IS NEW IN THIS STUDY?: The median sST2 concentration was similar between patients with (9.62; 7.18-13.44 ng/mL; p = 0.66) and without (9.67; 7.08-13.06 ng/mL) H. pylori. WHAT ARE THE FUTURE CLINICAL AND RESEARCH IMPLICATIONS OF THE STUDY FINDINGS?: The results indicate that sST2 may not serve as a valuable biomarker in the diagnosis and treatment of H. pylori infection.
Collapse
Affiliation(s)
- Sarah Wernly
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Vera Paar
- Clinic II for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Andreas Völkerer
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christian Datz
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Clinic II for Internal Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Wernly
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, Salzburg, Austria.
- Institute of General Practice, Family Medicine and Preventive Medicine, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
13
|
Pisani LF, Teani I, Vecchi M, Pastorelli L. Interleukin-33: Friend or Foe in Gastrointestinal Tract Cancers? Cells 2023; 12:1481. [PMID: 37296602 PMCID: PMC10252908 DOI: 10.3390/cells12111481] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Accumulating evidence suggests that Interleukin-33 (IL-33), a member of the IL-1 family, has crucial roles in tissue homeostasis and repair, type 2 immunity, inflammation, and viral infection. IL-33 is a novel contributing factor in tumorigenesis and plays a critical role in regulating angiogenesis and cancer progression in a variety of human cancers. The partially unraveled role of IL-33/ST2 signaling in gastrointestinal tract cancers is being investigated through the analysis of patients' samples and by studies in murine and rat models. In this review, we discuss the basic biology and mechanisms of release of the IL-33 protein and its involvement in gastrointestinal cancer onset and progression.
Collapse
Affiliation(s)
- Laura Francesca Pisani
- Gastroenterology and Endoscopy Unit, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| | - Isabella Teani
- Department of Medicine, University of Verona, 37129 Verona, Italy;
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Luca Pastorelli
- Department of Health Sciences, University of Milan, 20122 Milan, Italy
- Gastroenterology and Liver Unit, ASST Santi Paolo e Carlo, 20142 Milan, Italy
| |
Collapse
|
14
|
Jiao Y, Yan Z, Yang A. The Roles of Innate Lymphoid Cells in the Gastric Mucosal Immunology and Oncogenesis of Gastric Cancer. Int J Mol Sci 2023; 24:ijms24076652. [PMID: 37047625 PMCID: PMC10095467 DOI: 10.3390/ijms24076652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a group of innate immune cells that have garnered considerable attention due to their critical roles in regulating immunity and tissue homeostasis. They are particularly abundant in the gastrointestinal tract, where they have been shown to interact with commensal bacteria, pathogens, and other components of the local microenvironment to influence host immune responses to infection and oncogenesis. Their tissue-residency properties enable gastric ILCs a localized and rapid response to alert and stress, which indicates their key potential in regulating immunosurveillance. In this review, we discuss the current understanding of the role of ILCs in the gastric mucosa, with a focus on their interactions with the gastric microbiota and Helicobacter pylori and their contributions to tissue homeostasis and inflammation. We also highlight recent findings on the involvement of ILCs in the pathogenesis of gastric cancer and the implications of targeting ILCs as a therapeutic approach. Overall, this review provides an overview of the diverse functions of ILCs in gastric mucosa and highlights their potential as targets for future therapies for gastric cancer.
Collapse
Affiliation(s)
- Yuhao Jiao
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zhiyu Yan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- 4 + 4 M.D. Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
15
|
Widmayer P, Pregitzer P, Breer H. Short-term high fat feeding induces inflammatory responses of tuft cells and mucosal barrier cells in the murine stomach. Histol Histopathol 2023; 38:273-286. [PMID: 35904321 DOI: 10.14670/hh-18-503] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Feeding mice with a high fat diet (HFD) induces inflammation and results in changes of gene expression and cellular composition in various tissues throughout the body, including the gastrointestinal tract. In the stomach, tuft cells expressing the receptor GPR120 are capable of sensing saturated long chain fatty acids (LCFAs) and thus may be involved in initiating mechanisms of mucosal inflammation. In this study, we assessed which cell types may additionally be affected by high fat feeding and which candidate molecular mediators might contribute to mucosa-protective immune responses. A high fat dietary intervention for 3 weeks caused an expansion of tuft cells that was accompanied by a higher frequency of mucosal mast cells and surface mucous cells which are a known source of the insult-associated cytokine interleukin 33 (IL-33). Our data demonstrate that both brush and mucosal mast cells comprise the enzyme ALOX5 and its activating protein FLAP and thus have the capacity for synthesizing leukotriene (LT). In HFD mice, several tuft cells showed a perinuclear colocalization of ALOX5 with FLAP which is indicative of an active LT synthesis. Monitoring changes in the expression of genes encoding elements of LT synthesis and signaling revealed that transcript levels of the leukotriene C4 synthase, LTC4S, catalyzing the first step in the biosynthesis of cysteinyl (cys) LTs, and the cysLT receptors, cysLTR2 and cysLTR3, were upregulated in mice on HFD. These mice also showed an increased expression level of IL-33 receptors, the membrane-bound ST2L and soluble isoform sST2, as well as the mast cell-specific protease MCPT1. Based on these findings it is conceivable that upon sensing saturated LCFAs tuft cells may elicit inflammatory responses which result in the production of cysLTs and activation of surface mucous cells as well as mucosal mast cells regulating gastric mucosal function and integrity.
Collapse
Affiliation(s)
- Patricia Widmayer
- Institute of Biology, Department of Physiology (190v), University of Hohenheim, Stuttgart, Germany.
| | - Pablo Pregitzer
- Institute of Biology, Department of Physiology (190v), University of Hohenheim, Stuttgart, Germany
| | - Heinz Breer
- Institute of Biology, Department of Physiology (190v), University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
16
|
Kuwabara R, Qin T, Alberto Llacua L, Hu S, Boekschoten MV, de Haan BJ, Smink AM, de Vos P. Extracellular matrix inclusion in immunoisolating alginate-based microcapsules promotes longevity, reduces fibrosis, and supports function of islet allografts in vivo. Acta Biomater 2023; 158:151-162. [PMID: 36610609 DOI: 10.1016/j.actbio.2022.12.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023]
Abstract
Immunoisolation of pancreatic-islets in alginate-microcapsules is applied to treat diabetes. However, long-term islet function is limited, which might be due to damaged and lack of contact with pancreatic extracellular matrix (ECM) components. Herein we investigated the impact of collagen IV combined with laminin sequences, either RGD, LRE, or PDSGR, on graft-survival of microencapsulated bioluminescent islets in vivo. Collagen IV with RGD had the most pronounced effect. It enhanced after 8-week implantation in immune-incompetent mice the bioluminescence of allogeneic islets by 3.2-fold, oxygen consumption rate by 14.3-fold and glucose-induced insulin release by 9.6-fold. Transcriptomics demonstrated that ECM enhanced canonical pathways involving insulin-secretion and that it suppressed pathways related to inflammation and hypoxic stress. Also, 5.8-fold fewer capsules were affected by fibrosis. In a subsequent longevity study in immune-competent mice, microencapsulated allografts containing collagen IV and RGD had a 2.4-fold higher functionality in the first week after implantation and remained at least 2.1-fold higher during the study. Islets in microcapsules containing collagen IV and RGD survived 211 ± 24.1 days while controls survived 125 ± 19.7 days. Our findings provide in vivo evidence for the efficacy of supplementing immunoisolating devices with specific ECM components to enhance functionality and longevity of islet-grafts in vivo. STATEMENT OF SIGNIFICANCE: Limitations in duration of survival of immunoisolated pancreatic islet grafts is a major obstacle for application of the technology to treat diabetes. Accumulating evidence supports that incorporation of extracellular matrix (ECM) molecules in the capsules enhances longevity of pancreatic islets. After selection of the most efficacious laminin sequence in vitro, we show in vivo that inclusion of collagen IV and RGD in alginate-based microcapsules enhances survival, insulin secretion function, and mitochondrial function. It also suppresses fibrosis by lowering proinflammatory cytokines secretion. Moreover, transcriptomic analysis shows that ECM-inclusion promotes insulin-secretion related pathways and attenuates inflammation and hypoxic stress related pathways in islets. We show that inclusion of ECM in immunoisolating devices is a promising strategy to promote long-term survival of islet-grafts.
Collapse
Affiliation(s)
- Rei Kuwabara
- Section Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, and University Medical Center Groningen, Hanzeplein 1, EA 11, Groningen 9713 GZ, the Netherlands; Department of Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Tian Qin
- Section Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, and University Medical Center Groningen, Hanzeplein 1, EA 11, Groningen 9713 GZ, the Netherlands.
| | - L Alberto Llacua
- Section Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, and University Medical Center Groningen, Hanzeplein 1, EA 11, Groningen 9713 GZ, the Netherlands
| | - Shuxian Hu
- Section Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, and University Medical Center Groningen, Hanzeplein 1, EA 11, Groningen 9713 GZ, the Netherlands
| | - Mark V Boekschoten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, Wageningen 6708 WE, the Netherlands
| | - Bart J de Haan
- Section Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, and University Medical Center Groningen, Hanzeplein 1, EA 11, Groningen 9713 GZ, the Netherlands
| | - Alexandra M Smink
- Section Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, and University Medical Center Groningen, Hanzeplein 1, EA 11, Groningen 9713 GZ, the Netherlands
| | - Paul de Vos
- Section Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, and University Medical Center Groningen, Hanzeplein 1, EA 11, Groningen 9713 GZ, the Netherlands
| |
Collapse
|
17
|
Liu L, Wang Y, Zhao Y, Zhang W, Liu J, Wang F, Wang P, Tang X. Global knowledge mapping and emerging trends in research between spasmolytic polypeptide-expressing metaplasia and gastric carcinogenesis: A bibliometric analysis from 2002 to 2022. Front Cell Infect Microbiol 2023; 12:1108378. [PMID: 36776551 PMCID: PMC9912936 DOI: 10.3389/fcimb.2022.1108378] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/28/2022] [Indexed: 01/30/2023] Open
Abstract
Background Spasmolytic polypeptide expression metaplasia (SPEM) occurs in the corpus of the stomach and is closely related to inflammations caused by H. pylori infection. Recently, SPEM was suggested as one of the dubious precancerous lesions of gastric cancer (GC). Thus, further research on SPEM cell transdifferentiation and its underlying mechanisms could facilitate the development of new molecular targets improving the therapeutics of GC. Using bibliometrics, we analyzed publications, summarized the research hotspots and provided references for scientific researchers engaged in related research fields. Methods We searched the Web of Science Core Collection (WoSCC) for publications related to SPEM-GC from 2002 to 2022. The VOSviewer, SCImago, CiteSpace and R software were used to visualize and analyze the data. Gene targets identified in the keyword list were analyzed for functional enrichment using the KEGG and GO databases. Results Of the 292 articles identified in the initial search, we observed a stable trend in SPEM-GC research but rapid growth in the number of citations. The United States was the leader in terms of quality publications and international cooperation among them. The total number of articles published by Chinese scholars was second to the United States. Additionally, despite its low centrality and average citation frequency, China has become one of the world's most dynamic countries in academics. In terms of productivity, Vanderbilt University was identified as the most productive institution. Further, we also observed that Gastroenterology was the highest co-cited journal, and Goldenring Jr. was the most prolific author with the largest centrality. Conclusion SPEM could serve as an initial step in diagnosing gastric precancerous lesions. Current hotspots and frontiers of research include SPEM cell lineage differentiation, interaction with H. pylori, disturbances of the mucosal microenvironment, biomarkers, clinical diagnosis and outcomes of SPEM, as well as the development of proliferative SPEM animal models. However, further research and collaboration are still required. The findings presented in this study can be used as reference for the research status of SPEM-GC and determine new directions for future studies.
Collapse
Affiliation(s)
- Lin Liu
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yukun Zhao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Zhang
- Department of Pathology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiong Liu
- Department of Pathology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengyun Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Xudong Tang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Xudong Tang,
| |
Collapse
|
18
|
Abstract
When discovered in the early 2000s, interleukin-33 (IL-33) was characterized as a potent driver of type 2 immunity and implicated in parasite clearance, as well as asthma, allergy, and lung fibrosis. Yet research in other models has since revealed that IL-33 is a highly pleiotropic molecule with diverse functions. These activities are supported by elusive release mechanisms and diverse expression of the IL-33 receptor, STimulation 2 (ST2), on both immune and stromal cells. Interestingly, IL-33 also supports type 1 immune responses during viral and tumor immunity and after allogeneic hematopoietic stem cell transplantation. Yet the IL-33-ST2 axis is also critical to the establishment of systemic homeostasis and tissue repair and regeneration. Despite these recent findings, the mechanisms by which IL-33 governs the balance between immunity and homeostasis or can support both effective repair and pathogenic fibrosis are poorly understood. As such, ongoing research is trying to understand the potential reparative and regulatory versus pro-inflammatory and pro-fibrotic roles for IL-33 in transplantation. This review provides an overview of the emerging regenerative role of IL-33 in organ homeostasis and tissue repair as it relates to transplantation immunology. It also outlines the known impacts of IL-33 in commonly transplanted solid organs and covers the envisioned roles for IL-33 in ischemia-reperfusion injury, rejection, and tolerance. Finally, we give a comprehensive summary of its effects on different cell populations involved in these processes, including ST2 + regulatory T cells, innate lymphoid cell type 2, as well as significant myeloid cell populations.
Collapse
|
19
|
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
|
20
|
Liu J, Liu L, Su Y, Wang Y, Zhu Y, Sun X, Guo Y, Shan J. IL-33 Participates in the Development of Esophageal Adenocarcinoma. Pathol Oncol Res 2022; 28:1610474. [PMID: 36110250 PMCID: PMC9469785 DOI: 10.3389/pore.2022.1610474] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022]
Abstract
Background: The progression from chronic gastroesophageal reflux disease (GERD) to Barrett esophagus (BE) and esophageal adenocarcinoma (EAC) is an inflammatory-driven neoplastic change. Interleukin-33 (IL-33) has identified as a crucial factor in several inflammatory disorders and malignancies.Methods: The high-density tissue microarray of the human EAC was analyzed with IL-33 immunohistochemistry staining (IHC). By anastomosing the jejunum with the esophagus, the rat model of EAC with mixed gastroduodenal reflux was established. The expression of IL-33 was determined using quantitative real-time polymerase chain reaction (RT-qPCR), western blot (WB), IHC and enzyme-linked immunosorbent assay (ELISA). Esophageal adenocarcinoma cells (OE19 and OE33) and human esophageal epithelial cells (HEECs) were used.Results: In the cytoplasm of human EAC tissue, IL-33 expression was substantially greater than in adjacent normal tissue. In rat model, the expression of IL-33 in the EAC group was considerably greater than in the control group, and this expression increased with the upgrade of pathological stage. In in vitro experiment, the mRNA and protein levels of IL-33 were considerably greater in OE19 and OE33 than in HEECs. The stimulation of IL-33 enhanced the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of OE19 and OE33, but soluble ST2 (sST2) inhibited these effects. IL-33 stimulated the release of IL-6 by OE19 and OE33 cells.Conclusion: This study demonstrated the overexpression of IL-33 in the transition from GERD to EAC and that IL-33 promoted carcinogenesis in EAC cells through ST2. IL-33 might be a possible preventive target for EAC.
Collapse
Affiliation(s)
- Jia Liu
- School of Medicine, Southwest Jiaotong University, Chengdu, China
- The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Lei Liu
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Yang Su
- School of Medicine, Southwest Jiaotong University, Chengdu, China
- The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Yi Wang
- North Sichuan Medical College, Nanchong, China
| | - Yuchun Zhu
- North Sichuan Medical College, Nanchong, China
| | - Xiaobin Sun
- Department of Gastroenterology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Yuanbiao Guo
- Medical Research Center, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Jing Shan
- Department of Gastroenterology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
- *Correspondence: Jing Shan,
| |
Collapse
|
21
|
Lenti MV, Facciotti F, Miceli E, Vanoli A, Fornasa G, Lahner E, Spadoni I, Giuffrida P, Arpa G, Pasini A, Rovedatti L, Caprioli F, Travelli C, Lattanzi G, Conti L, Klersy C, Vecchi M, Paulli M, Annibale B, Corazza GR, Rescigno M, Di Sabatino A. Mucosal Overexpression of Thymic Stromal Lymphopoietin and Proinflammatory Cytokines in Patients With Autoimmune Atrophic Gastritis. Clin Transl Gastroenterol 2022; 13:e00510. [PMID: 35905420 PMCID: PMC10476748 DOI: 10.14309/ctg.0000000000000510] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/06/2022] [Indexed: 09/06/2023] Open
Abstract
INTRODUCTION The immune mechanisms underlying human autoimmune atrophic gastritis (AAG) are poorly understood. We sought to assess immune mucosal alterations in patients with AAG. METHODS In 2017-2021, we collected gastric corpus biopsies from 24 patients with AAG (median age 62 years, interquartile range 56-67, 14 women), 26 age-matched and sex-matched healthy controls (HCs), and 14 patients with Helicobacter pylori infection (HP). We investigated the lamina propria mononuclear cell (LPMC) populations and the mucosal expression of thymic stromal lymphopoietin (TSLP) and nicotinamide phosphoribosyltransferase (NAMPT). Ex vivo cytokine production by organ culture biopsies, under different stimuli (short TSLP and zinc-l-carnosine), and the gastric vascular barrier through plasmalemma vesicle-associated protein-1 (PV1) were also assessed. RESULTS In the subset of CD19+ LPMC, CD38+ cells (plasma cells) were significantly higher in AAG compared with HC. Ex vivo production of tumor necrosis factor (TNF)-α, interleukin (IL)-15, and transforming growth factor β1 was significantly higher in AAG compared with HC. At immunofluorescence, both IL-7R and TSLP were more expressed in AAG compared with HC and HP, and short TSLP transcripts were significantly increased in AAG compared with HC. In the supernatants of AAG corpus mucosa, short TSLP significantly reduced TNF-α, while zinc-l-carnosine significantly reduced interferon-γ, TNF-α, IL-21, IL-6, and IL-15. NAMPT transcripts were significantly increased in AAG compared with HC. PV1 was almost absent in AAG, mildly expressed in HC, and overexpressed in HP. DISCUSSION Plasma cells, proinflammatory cytokines, and altered gastric vascular barrier may play a major role in AAG. TSLP and NAMPT may represent potential therapeutic targets, while zinc-l-carnosine may dampen mucosal inflammation.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Emanuela Miceli
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Alessandro Vanoli
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Giulia Fornasa
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University La Sapienza, Rome, Italy
| | - Ilaria Spadoni
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Paolo Giuffrida
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Giovanni Arpa
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Alessandra Pasini
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Laura Rovedatti
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, IRCCS Ca' Granda Hospital Foundation, University of Milan, Milan, Italy
| | - Cristina Travelli
- Department of Pharmaceutical Sciences, University of Pavia, Pavia, Italy
| | - Georgia Lattanzi
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milan, Italy
| | - Laura Conti
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University La Sapienza, Rome, Italy
| | - Catherine Klersy
- Clinical Epidemiology & Biometry, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, IRCCS Ca' Granda Hospital Foundation, University of Milan, Milan, Italy
| | - Marco Paulli
- Unit of Anatomic Pathology, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, University La Sapienza, Rome, Italy
| | - Gino Roberto Corazza
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Antonio Di Sabatino
- First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, University of Pavia, Pavia, Italy
| |
Collapse
|
22
|
IL-33 promotes gastric tumour growth in concert with activation and recruitment of inflammatory myeloid cells. Oncotarget 2022; 13:785-799. [PMID: 35677533 PMCID: PMC9159270 DOI: 10.18632/oncotarget.28238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/07/2022] [Indexed: 01/01/2023] Open
Abstract
Interleukin-33 (IL-33) is an IL-1 family cytokine known to promote T-helper (Th) type 2 immune responses that are often deregulated in gastric cancer (GC). IL-33 is overexpressed in human gastric tumours suggesting a role in driving GC progression although a causal link has not been proven. Here, we investigated the impact of IL-33 genetic deficiency in the well-characterized gp130F/F mouse model of GC. Expression of IL-33 (and it’s cognate receptor, ST2) was increased in human and mouse GC progression. IL-33 deficient gp130F/F/Il33−/− mice had reduced gastric tumour growth and reduced recruitment of pro-tumorigenic myeloid cells including key mast cell subsets and type-2 (M2) macrophages. Cell sorting of gastric tumours revealed that IL-33 chiefly localized to gastric (tumour) epithelial cells and was absent from tumour-infiltrating immune cells (except modest IL-33 enrichment within CD11b+ CX3CR1+CD64+MHCII+ macrophages). By contrast, ST2 was absent from gastric epithelial cells and localized exclusively within the (non-macrophage) immune cell fraction together with mast cell markers, Mcpt1 and Mcpt2. Collectively, we show that IL-33 is required for gastric tumour growth and provide evidence of a likely mechanism by which gastric epithelial-derived IL-33 drives mobilization of tumour-promoting inflammatory myeloid cells.
Collapse
|
23
|
Nagao M, Fukuda A, Omatsu M, Namikawa M, Sono M, Fukunaga Y, Masuda T, Araki O, Yoshikawa T, Ogawa S, Masuo K, Goto N, Hiramatsu Y, Muta Y, Tsuda M, Maruno T, Nakanishi Y, Taketo MM, Ferrer J, Tsuruyama T, Nakanuma Y, Taura K, Uemoto S, Seno H. Concurrent Activation of Kras and Canonical Wnt Signaling Induces Premalignant Lesions That Progress to Extrahepatic Biliary Cancer in Mice. Cancer Res 2022; 82:1803-1817. [PMID: 35247892 DOI: 10.1158/0008-5472.can-21-2176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/31/2021] [Accepted: 02/25/2022] [Indexed: 11/16/2022]
Abstract
Biliary cancer has long been known to carry a poor prognosis, yet the molecular pathogenesis of carcinoma of the extrahepatic biliary system and its precursor lesions remains elusive. Here we investigated the role of Kras and canonical Wnt pathways in the tumorigenesis of the extrahepatic bile duct (EHBD) and gall bladder (GB). In mice, concurrent activation of Kras and Wnt pathways induced biliary neoplasms that resembled human intracholecystic papillary-tubular neoplasm (ICPN) and biliary intraepithelial neoplasia (BilIN), putative precursors to invasive biliary cancer. At a low frequency, these lesions progressed to adenocarcinoma in a xenograft model, establishing them as precancerous lesions. Global gene expression analysis revealed increased expression of genes associated with c-Myc and TGFβ pathways in mutant biliary spheroids. Silencing or pharmacologic inhibition of c-Myc suppressed proliferation of mutant biliary spheroids, whereas silencing of Smad4/Tgfbr2 or pharmacologic inhibition of TGFβ signaling increased proliferation of mutant biliary spheroids and cancer formation in vivo. Human ICPNs displayed activated Kras and Wnt signals and c-Myc and TGFβ pathways. Thus, these data provide direct evidence that concurrent activation of the Kras and canonical Wnt pathways results in formation of ICPN and BilIN, which could develop into biliary cancer. SIGNIFICANCE This work shows how dysregulation of canonical cell growth pathways drives precursors to biliary cancers and identifies several molecular vulnerabilities as potential therapeutic targets in these precursors to prevent oncogenic progression.
Collapse
Affiliation(s)
- Munemasa Nagao
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Mayuki Omatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Mio Namikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Makoto Sono
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Yuichi Fukunaga
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan.,Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Tomonori Masuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Osamu Araki
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Takaaki Yoshikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Satoshi Ogawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Kenji Masuo
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Norihiro Goto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Yukiko Hiramatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Yu Muta
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Motoyuki Tsuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Makoto Mark Taketo
- Kitano Hospital, The Tazuke Kofukai Medical Research Institute, Kita-ku, Osaka, Japan.,iACT, Kyoto University Hospital, Sakyo-ku, Kyoto, Japan
| | - Jorge Ferrer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Spain.,Genetics and Genomics Section, Department of Metabolism, Digestion and Reproduction, National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, United Kingdom
| | - Tatsuaki Tsuruyama
- Department of Drug Discovery Medicine, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Yasuni Nakanuma
- Department of Diagnostic Pathology, Fukui Prefecture Saiseikai Hospital, Fukui, Japan
| | - Kojiro Taura
- Division of Hepatobiliary Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Shinji Uemoto
- Division of Hepatobiliary Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
24
|
Sonic Hedgehog acts as a macrophage chemoattractant during regeneration of the gastric epithelium. NPJ Regen Med 2022; 7:3. [PMID: 35022438 PMCID: PMC8755719 DOI: 10.1038/s41536-021-00196-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 11/25/2021] [Indexed: 11/19/2022] Open
Abstract
Sonic Hedgehog (Shh), secreted from gastric parietal cells, contributes to the regeneration of the epithelium. The recruitment of macrophages plays a central role in the regenerative process. The mechanism that regulates macrophage recruitment in response to gastric injury is largely unknown. Here we tested the hypothesis that Shh stimulates macrophage chemotaxis to the injured epithelium and contributes to gastric regeneration. A mouse model expressing a myeloid cell-specific deletion of Smoothened (LysMcre/+;Smof/f) was generated using transgenic mice bearing loxP sites flanking the Smo gene (Smo loxP) and mice expressing a Cre recombinase transgene from the Lysozyme M locus (LysMCre). Acetic acid injury was induced in the stomachs of both control and LysMcre/+;Smof/f (SmoKO) mice and gastric epithelial regeneration and macrophage recruitment analyzed over a period of 7 days post-injury. Bone marrow-derived macrophages (BM-Mø) were collected from control and SmoKO mice. Human-derived gastric organoid/macrophage co-cultures were established, and macrophage chemotaxis measured. Compared to control mice, SmoKO animals exhibited inhibition of ulcer repair and normal epithelial regeneration, which correlated with decreased macrophage infiltration at the site of injury. Bone marrow chimera experiments using SmoKO donor cells showed that control chimera mice transplanted with SmoKO bone marrow donor cells exhibited a loss of ulcer repair, and transplantation of control bone marrow donor cells to SmoKO mice rescued epithelial cell regeneration. Histamine-stimulated Shh secretion in human organoid/macrophage co-cultures resulted in macrophage migration toward the gastric epithelium, a response that was blocked with Smo inhibitor Vismodegib. Shh-induced macrophage migration was mediated by AKT signaling. In conclusion, Shh signaling acts as a macrophage chemoattractant via a Smo-dependent mechanism during gastric epithelial regeneration in response to injury.
Collapse
|
25
|
Zhang M, Duffen JL, Nocka KH, Kasaian MT. IL-13 Controls IL-33 Activity through Modulation of ST2. THE JOURNAL OF IMMUNOLOGY 2021; 207:3070-3080. [PMID: 34789557 DOI: 10.4049/jimmunol.2100655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022]
Abstract
IL-33 is a multifunctional cytokine that mediates local inflammation upon tissue damage. IL-33 is known to act on multiple cell types including group 2 innate lymphoid cells (ILC2s), Th2 cells, and mast cells to drive production of Th2 cytokines including IL-5 and IL-13. IL-33 signaling activity through transmembrane ST2L can be inhibited by soluble ST2 (sST2), which acts as a decoy receptor. Previous findings suggested that modulation of IL-13 levels in mice lacking decoy IL-13Rα2, or mice lacking IL-13, impacted responsiveness to IL-33. In this study, we used Il13 -/- mice to investigate whether IL-13 regulates IL-33 activity by modulating the transmembrane and soluble forms of ST2. In Il13 -/- mice, the effects of IL-33 administration were exacerbated relative to wild type (WT). Il13 -/- mice administered IL-33 i.p. had heightened splenomegaly, more immune cells in the peritoneum including an expanded ST2L+ ILC2 population, increased eosinophilia in the spleen and peritoneum, and reduced sST2 in the circulation and peritoneum. In the spleen, lung, and liver of mice given IL-33, gene expression of both isoforms of ST2 was increased in Il13 -/- mice relative to WT. We confirmed fibroblasts to be an IL-13-responsive cell type that can regulate IL-33 activity through production of sST2. This study elucidates the important regulatory activity that IL-13 exerts on IL-33 through induction of IL-33 decoy receptor sST2 and through modulation of ST2L+ ILC2s.
Collapse
Affiliation(s)
- Melvin Zhang
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Jennifer L Duffen
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Karl H Nocka
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Marion T Kasaian
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| |
Collapse
|
26
|
Jeong H, Lee B, Kim KH, Cho SY, Cho Y, Park J, Lee Y, Oh Y, Hwang BR, Jang AR, Park JH, Park JH, Jeong SH, Lee D, Lee YC, Lim KM, Goldenring JR, Nam KT. WFDC2 Promotes Spasmolytic Polypeptide-Expressing Metaplasia Through the Up-Regulation of IL33 in Response to Injury. Gastroenterology 2021; 161:953-967.e15. [PMID: 34116028 PMCID: PMC8380710 DOI: 10.1053/j.gastro.2021.05.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS WAP 4-disulfide core domain protein 2 (WFDC2), also known as human epididymis protein 4, is a small secretory protein that is highly expressed in fibrosis and human cancers, particularly in the ovaries, lungs, and stomach. However, the role of WFDC2 in carcinogenesis is not fully understood. The present study aimed to investigate the role of WFDC2 in gastric carcinogenesis with the use of preneoplastic metaplasia models. METHODS Three spasmolytic polypeptide-expressing metaplasia (SPEM) models were established in both wild-type and Wfdc2-knockout mice with DMP-777, L635, and high-dose tamoxifen, respectively. To reveal the functional role of WFDC2, we performed transcriptomic analysis with DMP-777-treated gastric corpus specimens. RESULTS Wfdc2-knockout mice exhibited remarkable resistance against oxyntic atrophy, SPEM emergence, and accumulation of M2-type macrophages in all 3 SPEM models. Transcriptomic analysis revealed that Wfdc2-knockout prevented the up-regulation of interleukin-33 (IL33) expression in the injured mucosal region of SPEM models. Notably, supplementation of recombinant WFDC2 induced IL33 production and M2 macrophage polarization, and ultimately promoted SPEM development. Moreover, long-term treatment with recombinant WFDC2 was able to induce SPEM development. CONCLUSIONS WFDC2 expressed in response to gastric injury promotes SPEM through the up-regulation of IL33 expression. These findings provide novel insights into the role of WFDC2 in gastric carcinogenesis.
Collapse
Affiliation(s)
- Haengdueng Jeong
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Buhyun Lee
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Kwang H Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | | | - Yejin Cho
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jeongeun Park
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Yura Lee
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yeseul Oh
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Bo Ram Hwang
- Department of Internal Medicine, Graduate School, Yonsei University College of Medicine, Seoul, Korea
| | - Ah-Ra Jang
- Laboratory of Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Jong-Hwan Park
- Laboratory of Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Ji-Ho Park
- Department of Surgery, Gyeongsang National University Hospital, Jinju, Korea
| | - Sang-Ho Jeong
- Department of Surgery, Gyeongsang National University Hospital, Jinju, Korea
| | - Daekee Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Yong Chan Lee
- Department of Internal Medicine, Graduate School, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul, Korea.
| | - James R Goldenring
- Epithelial Biology Center and Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville VA Medical Center, Nashville, Tennessee.
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
27
|
Interference of LPS H. pylori with IL-33-Driven Regeneration of Caviae porcellus Primary Gastric Epithelial Cells and Fibroblasts. Cells 2021; 10:cells10061385. [PMID: 34199843 PMCID: PMC8227243 DOI: 10.3390/cells10061385] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Lipopolysaccharide (LPS) of Helicobacter pylori (Hp) bacteria causes disintegration of gastric tissue cells in vitro. It has been suggested that interleukin (IL)-33 is involved in healing gastric injury. Aim: To elucidate whether Hp LPS affects regeneration of gastric barrier initiated by IL-33. Methods: Primary gastric epithelial cells or fibroblasts from Caviae porcellus were transfected with siRNA IL-33. Such cells, not exposed or treated with LPS Hp, were sub-cultured in the medium with or without exogenous IL-33. Then cell migration was assessed in conjunction with oxidative stress and apoptosis, activation of extracellular signal-regulated kinase (Erk), production of collagen I and soluble ST2 (IL-33 decoy). Results: Control cells not treated with LPS Hp migrated in the presence of IL-33. The pro-regenerative activity of IL-33 was related to stimulation of cells to collagen I production. Wound healing by cells exposed to LPS Hp was inhibited even in the presence of IL-33. This could be due to increased oxidative stress and apoptosis in conjunction with Erk activation, sST2 elevation and modulation of collagen I production. Conclusions: The recovery of gastric barrier cells during Hp infection potentially can be affected due to downregulation of pro-regenerative activity of IL-33 by LPS Hp.
Collapse
|
28
|
Pisani LF, Tontini GE, Gentile C, Marinoni B, Teani I, Nandi N, Creo P, Asti E, Bonavina L, Vecchi M, Pastorelli L. Proinflammatory Interleukin-33 Induces Dichotomic Effects on Cell Proliferation in Normal Gastric Epithelium and Gastric Cancer. Int J Mol Sci 2021; 22:ijms22115792. [PMID: 34071419 PMCID: PMC8197967 DOI: 10.3390/ijms22115792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/08/2021] [Accepted: 05/25/2021] [Indexed: 01/02/2023] Open
Abstract
Interleukin (IL)-33 is a member of the interleukin (IL)-1 family of cytokines linked to the development of inflammatory conditions and cancer in the gastrointestinal tract. This study is designed to investigate whether IL-33 has a direct effect on human gastric epithelial cells (GES-1), the human gastric adenocarcinoma cell line (AGS), and the gastric carcinoma cell line (NCI-N87) by assessing its role in the regulation of cell proliferation, migration, cell cycle, and apoptosis. Cell cycle regulation was also determined in ex vivo gastric cancer samples obtained during endoscopy and surgical procedures. Cell lines and tissue samples underwent stimulation with rhIL-33. Proliferation was assessed by XTT and CFSE assays, migration by wound healing assay, and apoptosis by caspase 3/7 activity assay and annexin V assay. Cell cycle was analyzed by means of propidium iodine assay, and gene expression regulation was assessed by RT-PCR profiling. We found that IL-33 has an antiproliferative and proapoptotic effect on cancer cell lines, and it can stimulate proliferation and reduce apoptosis in normal epithelial cell lines. These effects were also confirmed by the analysis of cell cycle gene expression, which showed a reduced expression of pro-proliferative genes in cancer cells, particularly in genes involved in G0/G1 and G2/M checkpoints. These results were confirmed by gene expression analysis on bioptic and surgical specimens. The aforementioned results indicate that IL-33 may be involved in cell proliferation in an environment- and cell-type-dependent manner.
Collapse
Affiliation(s)
- Laura Francesca Pisani
- Gastroenterology ans Endoscopy Unit, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (L.F.P.); (P.C.)
| | - Gian Eugenio Tontini
- Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milano, Italy; (G.E.T.); (C.G.); (B.M.); (I.T.); (N.N.); (E.A.); (L.B.)
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Carmine Gentile
- Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milano, Italy; (G.E.T.); (C.G.); (B.M.); (I.T.); (N.N.); (E.A.); (L.B.)
| | - Beatrice Marinoni
- Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milano, Italy; (G.E.T.); (C.G.); (B.M.); (I.T.); (N.N.); (E.A.); (L.B.)
| | - Isabella Teani
- Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milano, Italy; (G.E.T.); (C.G.); (B.M.); (I.T.); (N.N.); (E.A.); (L.B.)
| | - Nicoletta Nandi
- Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milano, Italy; (G.E.T.); (C.G.); (B.M.); (I.T.); (N.N.); (E.A.); (L.B.)
| | - Pasquale Creo
- Gastroenterology ans Endoscopy Unit, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (L.F.P.); (P.C.)
| | - Emanuele Asti
- Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milano, Italy; (G.E.T.); (C.G.); (B.M.); (I.T.); (N.N.); (E.A.); (L.B.)
- Division of General Surgery, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| | - Luigi Bonavina
- Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milano, Italy; (G.E.T.); (C.G.); (B.M.); (I.T.); (N.N.); (E.A.); (L.B.)
- Division of General Surgery, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
- Gastroenterology and Liver Unit, ASST Santi Paolo e Carlo, Ospedale San Paolo, 20100 Milano, Italy
| | - Luca Pastorelli
- Department of Health Sciences, Università degli Studi di Milano, 20133 Milano, Italy
- Department of Pathophysiology and Trasplantation, Università degli Studi di Milano, 20133 Milano, Italy
- Correspondence: ; Tel.: +39-0252774683
| |
Collapse
|
29
|
Hoffmann W. Trefoil Factor Family (TFF) Peptides and Their Links to Inflammation: A Re-evaluation and New Medical Perspectives. Int J Mol Sci 2021; 22:ijms22094909. [PMID: 34066339 PMCID: PMC8125380 DOI: 10.3390/ijms22094909] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Trefoil factor family peptides (TFF1, TFF2, TFF3), together with mucins, are typical exocrine products of mucous epithelia. Here, they act as a gastric tumor suppressor (TFF1) or they play different roles in mucosal innate immune defense (TFF2, TFF3). Minute amounts are also secreted as endocrine, e.g., by the immune and central nervous systems. As a hallmark, TFF peptides have different lectin activities, best characterized for TFF2, but also TFF1. Pathologically, ectopic expression occurs during inflammation and in various tumors. In this review, the role of TFF peptides during inflammation is discussed on two levels. On the one hand, the expression of TFF1-3 is regulated by inflammatory signals in different ways (upstream links). On the other hand, TFF peptides influence inflammatory processes (downstream links). The latter are recognized best in various Tff-deficient mice, which have completely different phenotypes. In particular, TFF2 is secreted by myeloid cells (e.g., macrophages) and lymphocytes (e.g., memory T cells), where it modulates immune reactions triggering inflammation. As a new concept, in addition to lectin-triggered activation, a hypothetical lectin-triggered inhibition of glycosylated transmembrane receptors by TFF peptides is discussed. Thus, TFFs are promising players in the field of glycoimmunology, such as galectins and C-type lectins.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
30
|
Kawai K, Uchiyama M, Hester J, Issa F. IL-33 drives the production of mouse regulatory T cells with enhanced in vivo suppressive activity in skin transplantation. Am J Transplant 2021; 21:978-992. [PMID: 33314772 PMCID: PMC7613121 DOI: 10.1111/ajt.16266] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 01/25/2023]
Abstract
Regulatory T cells (Tregs) are crucial mediators of immune homeostasis with the ability to modulate allogeneic response and control transplant rejection. Although Treg-based cell therapies have shown immense promise, methods to optimize current strategies are critical for successful implementation within the clinic. IL-33 is a cytokine with pleiotropic properties and effects on Treg function and development. In this study, we explored the unique properties of Treg populations activated through the IL-33/ST2 pathway, aiming to exploit their tolerogenic properties for cell therapy. We show that treatment with exogenous IL-33 results in a generalized downregulation of genes critical to T cell biology together with an upregulation of Treg-associated genes. Tregs that develop in response to IL-33 upregulate critical Treg-associated markers, yet without developing enhanced in vitro suppressive capacity. Conversely, these Tregs display potent regulatory activity in vivo, promoting long-term skin allograft survival in a stringent transplantation model. Detailed transcriptomic and immunophenotypic analyses of IL-33-expanded Tregs reveal an enhancement in graft-homing chemokine receptors, which may be partly responsible for their superior in vivo activity that is not reflected in vitro. IL-33 treatment is therefore an attractive adjunctive strategy for patients receiving Treg cell therapeutics.
Collapse
Affiliation(s)
- Kento Kawai
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Masateru Uchiyama
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK,Department of Surgery, Teikyo University, Tokyo, Japan
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Mahapatro M, Erkert L, Becker C. Cytokine-Mediated Crosstalk between Immune Cells and Epithelial Cells in the Gut. Cells 2021; 10:cells10010111. [PMID: 33435303 PMCID: PMC7827439 DOI: 10.3390/cells10010111] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/28/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Cytokines are small proteins that are secreted by a vast majority of cell types in the gut. They not only establish cell-to-cell interactions and facilitate cellular signaling, but also regulate both innate and adaptive immune responses, thereby playing a central role in genetic, inflammatory, and infectious diseases of the gut. Both, immune cells and gut epithelial cells, play important roles in intestinal disease development. The epithelium is located in between the mucosal immune system and the gut microbiome. It not only establishes an efficient barrier against gut microbes, but it also signals information from the gut lumen and its composition to the immune cell compartment. Communication across the epithelial cell layer also occurs in the other direction. Intestinal epithelial cells respond to immune cell cytokines and their response influences and shapes the microbial community within the gut lumen. Thus, the epithelium should be seen as a translator or a moderator between the microbiota and the mucosal immune system. Proper communication across the epithelium seems to be a key to gut homeostasis. Indeed, current genome-wide association studies for intestinal disorders have identified several disease susceptibility loci, which map cytokine signatures and their related signaling genes. A thorough understanding of this tightly regulated cytokine signaling network is crucial. The main objective of this review was to shed light on how cytokines can orchestrate epithelial functions such as proliferation, cell death, permeability, microbe interaction, and barrier maintenance, thereby safeguarding host health. In addition, cytokine-mediated therapy for inflammation and cancer are discussed.
Collapse
|
32
|
De Salvo C, Pastorelli L, Petersen CP, Buttò LF, Buela KA, Omenetti S, Locovei SA, Ray S, Friedman HR, Duijser J, Xin W, Osme A, Cominelli F, Mahabeleshwar GH, Mills JC, Goldenring JR, Pizarro TT. Interleukin 33 Triggers Early Eosinophil-Dependent Events Leading to Metaplasia in a Chronic Model of Gastritis-Prone Mice. Gastroenterology 2021; 160:302-316.e7. [PMID: 33010253 PMCID: PMC7755675 DOI: 10.1053/j.gastro.2020.09.040] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Interleukin (IL)33/IL1F11 is an important mediator for the development of type 2 T-helper cell (Th2)-driven inflammatory disorders and has also been implicated in the pathogenesis of gastrointestinal (GI)-related cancers, including gastric carcinoma. We therefore sought to mechanistically determine IL33's potential role as a critical factor linking chronic inflammation and gastric carcinogenesis using gastritis-prone SAMP1/YitFc (SAMP) mice. METHODS SAMP and (parental control) AKR mice were assessed for baseline gastritis and progression to metaplasia. Expression/localization of IL33 and its receptor, ST2/IL1R4, were characterized in corpus tissues, and activation and neutralization studies were both performed targeting the IL33/ST2 axis. Dissection of immune pathways leading to metaplasia was evaluated, including eosinophil depletion studies using anti-IL5/anti-CCR3 treatment. RESULTS Progressive gastritis and, ultimately, intestinalized spasmolytic polypeptide-expressing metaplasia (SPEM) was detected in SAMP stomachs, which was absent in AKR but could be moderately induced with exogenous, recombinant IL33. Robust peripheral (bone marrow) expansion of eosinophils and local recruitment of both eosinophils and IL33-expressing M2 macrophages into corpus tissues were evident in SAMP. Interestingly, IL33 blockade did not affect bone marrow-derived expansion and local infiltration of eosinophils, but markedly decreased M2 macrophages and SPEM features, while eosinophil depletion caused a significant reduction in both local IL33-producing M2 macrophages and SPEM in SAMP. CONCLUSIONS IL33 promotes metaplasia and the sequelae of eosinophil-dependent downstream infiltration of IL33-producing M2 macrophages leading to intestinalized SPEM in SAMP, suggesting that IL33 represents a critical link between chronic gastritis and intestinalizing metaplasia that may serve as a potential therapeutic target for preneoplastic conditions of the GI tract.
Collapse
Affiliation(s)
| | - Luca Pastorelli
- Department of Pathology; Department of IRCCS Policlinico San Donato, Gastroenterology & Gastrointestinal Endoscopy Unit, San Donato Milanese, 20097 and Department of Biomedical Sciences, University of Milan, Milan, 20122, Italy
| | - Christine P. Petersen
- Department of Department of Surgery and the Epithelial Biology Center, Vanderbilt University, Nashville, TN, 37235, USA
| | - Ludovica F. Buttò
- Department of Medicine/Division of Gastroenterology & Liver Disease, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | | | | | - Silviu A. Locovei
- Department of Pathology; Department of Medicine/Division of Gastroenterology & Liver Disease, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | | | | | | | | | | | - Fabio Cominelli
- Department of Medicine/Division of Gastroenterology & Liver Disease, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | | | - Jason C. Mills
- Department of Medicine, Gastroenterology Division, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - James R. Goldenring
- Department of Department of Surgery and the Epithelial Biology Center, Vanderbilt University, Nashville, TN, 37235, USA
| | | |
Collapse
|
33
|
Meyer AR, Engevik AC, Madorsky T, Belmont E, Stier MT, Norlander AE, Pilkinton MA, McDonnell WJ, Weis JA, Jang B, Mallal SA, Peebles RS, Goldenring JR. Group 2 Innate Lymphoid Cells Coordinate Damage Response in the Stomach. Gastroenterology 2020; 159:2077-2091.e8. [PMID: 32891625 PMCID: PMC7726005 DOI: 10.1053/j.gastro.2020.08.051] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Severe injury to the lining of the stomach leads to changes in the epithelium (reprogramming) that protect and promote repair of the tissue, including development of spasmolytic polypeptide-expressing metaplasia (SPEM) and tuft and foveolar cell hyperplasia. Acute gastric damage elicits a type-2 inflammatory response that includes production of type-2 cytokines and infiltration by eosinophils and alternatively activated macrophages. Stomachs of mice that lack interleukin 33 (IL33) or interleukin 13 (IL13) did not undergo epithelial reprogramming after drug-induced injury. We investigated the role of group 2 innate lymphoid cells (ILC2s) in gastric epithelial repair. METHODS Acute gastric injury was induced in C57BL/6J mice (wild-type and RAG1 knockout) by administration of L635. We isolated ILC2s by flow cytometry from stomachs of mice that were and were not given L635 and performed single-cell RNA sequencing. ILC2s were depleted from wild-type and RAG1-knockout mice by administration of anti-CD90.2. We assessed gastric cell lineages, markers of metaplasia, inflammation, and proliferation. Gastric tissue microarrays from patients with gastric adenocarcinoma were analyzed by immunostaining. RESULTS There was a significant increase in the number of GATA3-positive ILC2s in stomach tissues from wild-type mice after L635-induced damage, but not in stomach tissues from IL33-knockout mice. We characterized a marker signature of gastric mucosal ILC2s and identified a transcription profile of metaplasia-associated ILC2s, which included changes in expression of Il5, Il13, Csf2, Pd1, and Ramp3; these changes were validated by quantitative polymerase chain reaction and immunocytochemistry. Depletion of ILC2s from mice blocked development of metaplasia after L635-induced injury in wild-type and RAG1-knockout mice and prevented foveolar and tuft cell hyperplasia and infiltration or activation of macrophages after injury. Numbers of ILC2s were increased in stomach tissues from patients with SPEM compared with patients with normal corpus mucosa. CONCLUSIONS In analyses of stomach tissues from mice with gastric tissue damage and patients with SPEM, we found evidence of type 2 inflammation and increased numbers of ILC2s. Our results suggest that ILC2s coordinate the metaplastic response to severe gastric injury.
Collapse
Affiliation(s)
- Anne R Meyer
- Department of Cell and Developmental Biology, Nashville, Tennessee; Epithelial Biology Center, Nashville, Tennessee
| | - Amy C Engevik
- Epithelial Biology Center, Nashville, Tennessee; Section of Surgical Sciences, Nashville, Tennessee
| | | | | | - Matthew T Stier
- Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee
| | - Allison E Norlander
- Division of Allergy, Pulmonary and Critical Care Medicine, Nashville, Tennessee
| | - Mark A Pilkinton
- Division of Infectious Disease, Nashville, Tennessee; Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Wyatt J McDonnell
- Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee
| | - Jared A Weis
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Bogun Jang
- Department of Pathology, Jeju National University School of Medicine, Jeju, Korea
| | - Simon A Mallal
- Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee; Division of Infectious Disease, Nashville, Tennessee; Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - R Stokes Peebles
- Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee; Division of Allergy, Pulmonary and Critical Care Medicine, Nashville, Tennessee; Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; Nashville VA Medical Center, Nashville, Tennessee
| | - James R Goldenring
- Department of Cell and Developmental Biology, Nashville, Tennessee; Epithelial Biology Center, Nashville, Tennessee; Section of Surgical Sciences, Nashville, Tennessee; Nashville VA Medical Center, Nashville, Tennessee.
| |
Collapse
|
34
|
Teal E, Dua-Awereh M, Hirshorn ST, Zavros Y. Role of metaplasia during gastric regeneration. Am J Physiol Cell Physiol 2020; 319:C947-C954. [PMID: 32755448 DOI: 10.1152/ajpcell.00415.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Spasmolytic polypeptide/trefoil factor 2 (TFF2)-expressing metaplasia (SPEM) is a mucous-secreting reparative lineage that emerges at the ulcer margin in response to gastric injury. Under conditions of chronic inflammation with parietal cell loss, SPEM has been found to emerge and evolve into neoplasia. Cluster-of-differentiation gene 44 (CD44) is known to coordinate normal and metaplastic epithelial cell proliferation. In particular, CD44 variant isoform 9 (CD44v9) associates with the cystine-glutamate transporter xCT, stabilizes the protein, and provides defense against reactive oxygen species (ROS). xCT stabilization by CD44v9 leads to defense against ROS by cystine uptake, glutathione (GSH) synthesis, and maintenance of the redox balance within the intracellular environment. Furthermore, p38 signaling is a known downstream ROS target, leading to diminished cell proliferation and migration, two vital processes of gastric epithelial repair. CD44v9 emerges during repair of the gastric epithelium after injury, where it is coexpressed with other markers of SPEM. The regulatory mechanisms for the emergence of CD44v9 and the role of CD44v9 during the process of gastric epithelial regeneration are largely unknown. Inflammation and M2 macrophage infiltration have recently been demonstrated to play key roles in the induction of SPEM after injury. The following review proposes new insights into the functional role of metaplasia in the process of gastric regeneration in response to ulceration. Our insights are extrapolated from documented studies reporting oxyntic atrophy and SPEM development and our current unpublished findings using the acetic acid-induced gastric injury model.
Collapse
Affiliation(s)
- Emma Teal
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Martha Dua-Awereh
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Sabrina T Hirshorn
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Yana Zavros
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
35
|
Oya Y, Hayakawa Y, Koike K. Tumor microenvironment in gastric cancers. Cancer Sci 2020; 111:2696-2707. [PMID: 32519436 PMCID: PMC7419059 DOI: 10.1111/cas.14521] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment favors the growth and expansion of cancer cells. Many cell types are involved in the tumor microenvironment such as inflammatory cells, fibroblasts, nerves, and vascular endothelial cells. These stromal cells contribute to tumor growth by releasing various molecules to either directly activate the growth signaling in cancer cells or remodel surrounding areas. This review introduces recent advances in findings on the interactions within the tumor microenvironment such as in cancer-associated fibroblasts (CAFs), immune cells, and endothelial cells, in particular those established in mouse gastric cancer models. In mice, myofibroblasts in the gastric stroma secrete R-spondin and support normal gastric stem cells. Most CAFs promote tumor growth in a paracrine manner, but CAF population appears to be heterogeneous in terms of their function and origin, and include both tumor-promoting and tumor-restraining populations. Among immune cell populations, tumor-associated macrophages, including M1 and M2 macrophages, and myeloid-derived suppressor cells (MDSCs), are reported to directly or indirectly promote gastric tumorigenesis by secreting soluble factors or modulating immune responses. Endothelial cells or blood vessels not only fuel tumors with nutrients, but also interact with cancer stem cells and immune cells by secreting chemokines or cytokines, and act as a cancer niche. Understanding these interactions within the tumor microenvironment would contribute to unraveling new therapeutic targets.
Collapse
Affiliation(s)
- Yukiko Oya
- Department of GastroenterologyGraduate school of Medicinethe University of TokyoTokyoJapan
| | - Yoku Hayakawa
- Department of GastroenterologyGraduate school of Medicinethe University of TokyoTokyoJapan
| | - Kazuhiko Koike
- Department of GastroenterologyGraduate school of Medicinethe University of TokyoTokyoJapan
| |
Collapse
|
36
|
Engevik AC, Kaji I, Goldenring JR. The Physiology of the Gastric Parietal Cell. Physiol Rev 2020; 100:573-602. [PMID: 31670611 PMCID: PMC7327232 DOI: 10.1152/physrev.00016.2019] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 10/10/2019] [Accepted: 10/13/2019] [Indexed: 12/11/2022] Open
Abstract
Parietal cells are responsible for gastric acid secretion, which aids in the digestion of food, absorption of minerals, and control of harmful bacteria. However, a fine balance of activators and inhibitors of parietal cell-mediated acid secretion is required to ensure proper digestion of food, while preventing damage to the gastric and duodenal mucosa. As a result, parietal cell secretion is highly regulated through numerous mechanisms including the vagus nerve, gastrin, histamine, ghrelin, somatostatin, glucagon-like peptide 1, and other agonists and antagonists. The tight regulation of parietal cells ensures the proper secretion of HCl. The H+-K+-ATPase enzyme expressed in parietal cells regulates the exchange of cytoplasmic H+ for extracellular K+. The H+ secreted into the gastric lumen by the H+-K+-ATPase combines with luminal Cl- to form gastric acid, HCl. Inhibition of the H+-K+-ATPase is the most efficacious method of preventing harmful gastric acid secretion. Proton pump inhibitors and potassium competitive acid blockers are widely used therapeutically to inhibit acid secretion. Stimulated delivery of the H+-K+-ATPase to the parietal cell apical surface requires the fusion of intracellular tubulovesicles with the overlying secretory canaliculus, a process that represents the most prominent example of apical membrane recycling. In addition to their unique ability to secrete gastric acid, parietal cells also play an important role in gastric mucosal homeostasis through the secretion of multiple growth factor molecules. The gastric parietal cell therefore plays multiple roles in gastric secretion and protection as well as coordination of physiological repair.
Collapse
Affiliation(s)
- Amy C Engevik
- Departments of Surgery and of Cell and Developmental Biology and the Epithelial Biology Center, Vanderbilt University School of Medicine, Vanderbilt University Medical Center and the Nashville VA Medical Center, Nashville, Tennessee
| | - Izumi Kaji
- Departments of Surgery and of Cell and Developmental Biology and the Epithelial Biology Center, Vanderbilt University School of Medicine, Vanderbilt University Medical Center and the Nashville VA Medical Center, Nashville, Tennessee
| | - James R Goldenring
- Departments of Surgery and of Cell and Developmental Biology and the Epithelial Biology Center, Vanderbilt University School of Medicine, Vanderbilt University Medical Center and the Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|
37
|
Proregenerative Activity of IL-33 in Gastric Tissue Cells Undergoing Helicobacter Pylori-Induced Apoptosis. Int J Mol Sci 2020; 21:ijms21051801. [PMID: 32151084 PMCID: PMC7084496 DOI: 10.3390/ijms21051801] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Interleukin (IL)-33 is a proinflammatory mediator that alerts the host immune system to disorders in tissue homeostasis. Aim. To understand the role of IL-33 in modulating gastric tissue cell growth affected by Helicobacter pylori (H. pylori). Methods. IL-33 production in guinea pigs (Caviae porcellus) experimentally infected with H. pylori was evaluated by ELISA or immunohistochemical staining. The proregenerative activity of IL-33 was evaluated using gastric epithelial cells and fibroblasts that were naive or transfected with IL-33 siRNA exposed to H. pylori glycine acid extract antigenic complex (GE), as well as by measuring cell migration, proliferation, metabolic activity and apoptosis. Animals infected by H. pylori responded with increased production of IL-33. Also, cells treated in vitro with GE released more IL-33 than cells that were unstimulated. Silencing IL-33 in cells resulted in downregulation of metabolic activity, adhesion, migration and proliferation, especially after treatment with H. pylori GE, as well as upregulation of cells apoptosis associated with caspase 3 increase and Bcl-xL decrease, suggesting proregenerative activity of IL-33. Interestingly, upregulation of cell proliferation by IL-33 was Erk independent. Our results indicate that IL-33 may protect gastric tissue from loss of homeostasis caused by deleterious effects of H. pylori components and the inflammatory response developed during infection.
Collapse
|
38
|
Wu MC, Leong PY, Chiou JY, Chen HH, Huang JY, Wei JCC. Increased Risk of Systemic Lupus Erythematosus in Patients With Helicobacter pylori Infection: A Nationwide Population-Based Cohort Study. Front Med (Lausanne) 2020; 6:330. [PMID: 32064263 PMCID: PMC7000519 DOI: 10.3389/fmed.2019.00330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori (HP) infection is associated with systemic lupus erythematosus (SLE), but the related results have been controversial. Therefore, this study investigated the association between HP infection and SLE by using a nationwide longitudinal population-based cohort. We identified 41,651 patients with HP infection and 83,302 matched controls between 2000 and 2013 from the Longitudinal Health Insurance Research Database of the National Taiwan Insurance Research Database. Age, gender, comorbidities, and medical visits were matched at a 1:2 ratio by using propensity score analysis. The adjusted hazard ratio (aHR) of SLE was calculated by multiple Cox regression. Furthermore, sensitivity test and stratified analysis were performed. The SLE incidence rate was 1.17 [95% confidence interval (CI): 0.89–1.54] per 100,000 person-months in the HP cohort, and the hazard ratio was 1.63 (95% CI: 1.12–2.37) in comparison with the propensity score-matched control cohort. After multivariate adjustment, patients with HP infection had a significantly high overall aHR (1.58; 95% CI: 1.08–2.30) of SLE. Stratified analysis revealed the aHR of 8.23 (95% CI: 1.77–38.32) in patients <30 years old, and the p for interaction between age and HP infection was 0.039. For age–sex subgroup analysis, the highest aHR was 12.74 (95% CI: 1.55–104.59) in young (aged <30 years) female patients with HP infection. HP infection is associated with a 1.63-fold increased SLE risk, particularly with female patients aged <30 years. Future research is required to elucidate the underlying mechanism of this association.
Collapse
Affiliation(s)
- Meng-Che Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Gastroenterology, Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Pui-Ying Leong
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jeng-Yuan Chiou
- School of Health Policy and Management, Chung Shan Medical University, Taichung, Taiwan
| | - Huang-Hsi Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jing-Yang Huang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
39
|
Lin Q, Han J, Sun Q, Wen L, Wang S. Functional variant of IL33 is associated with survival of osteosarcoma patients. J Bone Oncol 2019; 20:100270. [PMID: 31890491 PMCID: PMC6931128 DOI: 10.1016/j.jbo.2019.100270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/17/2019] [Accepted: 11/25/2019] [Indexed: 11/25/2022] Open
Abstract
Objectives Previous genome-wide association study showed that GLDC/IL33 loci were associated with overall survival in patients with osteosarcoma (OS). We performed a replication study to explore whether variants of GLDC/IL33 are associated with the survival of OS patients and to further verify their functional role in the gene expression. Methods A total of 216 patients with OS were enrolled. The overall survival time was calculated from the date of diagnosis till the date of last follow-up or mortality. Two SNPs were genotyped, including rs55933544 and rs74438701. OS specimens were obtained from 72 patients during surgery. The gene expression level of IL33 and GLDC was evaluated by qPCR. Patients were classified into two groups according to the 5-year overall survival (death/survival). The chi-square test was used to analyze difference of genotype frequency. The Student t-test was used to compare the gene expression level between different genotypes. Cumulative survival time was calculated by the Kaplan–Meier method and analyzed by the log-rank test. Results Genotype TT of rs55933544 was significantly associated with the event of death (0.176 vs. 0.061, p < 0.001). Patients with no risk allele T of rs55933544 showed a 5-year overall survival of 81.4% (110/141), which was significantly higher than an overall survival of 55.0% (29/54) for patients with one risk allele and 44.8% (12/21) for patients with two risk alleles (p < 0.01). Genotype TT of rs55933544 were indicative of remarkably lower expression of IL33 than genotype CC (0.00041 ± 0.00025 vs. 0.00065 ± 0.00031, p = 0.04). Patients with low IL33 expression presented remarkably worse survival as compared with the patients with high IL33 expression (p < .01) Conclusions Variant rs55933544 was associated with the survival time of OS patients. IL33 may contribute to a poor prognosis of OS. Further investigation into the biological mechanisms by which IL33 influences the overall survival can shed light on the improvement of clinical outcome for OS patients.
Collapse
Affiliation(s)
- Qingxi Lin
- Department of Orthopedic Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing 210008, China
| | - Jingjing Han
- Department of Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Sun
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing 210008, China
| | - Li Wen
- Department of Orthopedic Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing 210008, China
| | - Shoufeng Wang
- Department of Orthopedic Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing 210008, China
| |
Collapse
|
40
|
Zhang JZ, Wu ZH, Cheng Q. Screening and identification of key biomarkers in nasopharyngeal carcinoma: Evidence from bioinformatic analysis. Medicine (Baltimore) 2019; 98:e17997. [PMID: 31770211 PMCID: PMC6890310 DOI: 10.1097/md.0000000000017997] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As for the lack of simple and effective diagnostic methods at the early of the nasopharyngeal carcinoma (NPC), the mortality rate of NPC still remains high. Therefore, it is meaningful to explore the precise molecular mechanisms involved in the proliferation, carcinogenesis, and recurrence of NPC and thus find an effective diagnostic way and make a better therapeutic strategy.Three gene expression data sets (GSE64634, GSE53819, and GSE12452) were downloaded from Gene Expression Omnibus (GEO) and analyzed using the online tool GEO2R to identify differentially expressed genes (DEGs). Gene ontology functional analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of the DEGs were performed in Database for Annotation, Visualization and Integrated Discovery. The Search Tool for the Retrieval of Interacting Genes database was used to evaluate the interactions of DEGs and to construct a protein-protein interaction network using Cytoscape software. Hub genes were validated with the cBioPortal database.The overlap among the 3 data sets contained 306 genes were identified to be differentially expressed between NPC and non-NPC samples. A total of 13 genes (DNAAF1, PARPBP, TTC18, GSTA3, RCN1, MUC5AC, POU2AF1, FAM83B, SLC22A16, SPEF2, ERICH3, CCDC81, and IL33) were identified as hub genes with degrees ≥10.The present study was attempted to identify and functionally analyze the DEGs that may be involved in the carcinogenesis or progression of NPC by using comprehensive bioinformatics analyses and unveiled a series of hub genes and pathways. A total of 306 DEGs and 13 hub genes were identified and may be regarded as diagnostic biomarkers for NPC. However, more experimental studies are needed to carried out elucidate the biologic function of these genes results for NPC.
Collapse
Affiliation(s)
- Ji-Zhou Zhang
- Graduate School, Nanjing University of Chinese Medicine, Nanjing
- Oncology Department, Wenzhou Traditional Chinese Medicine affiliated to Zhejiang Chinese Medicine University, Wenzhou
| | - Zeng-Hong Wu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Cheng
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
41
|
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: 2.6] [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
|
42
|
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: 0.4] [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
|
43
|
Lee HY, Park EA, Lee KJ, Lee KH, Park SJ. Increased Innate Lymphoid Cell 3 and IL-17 Production in Mouse Lamina Propria Stimulated with Giardia lamblia. THE KOREAN JOURNAL OF PARASITOLOGY 2019; 57:225-232. [PMID: 31284344 PMCID: PMC6616171 DOI: 10.3347/kjp.2019.57.3.225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022]
Abstract
Innate lymphoid cells (ILCs) are key players during an immune response at the mucosal surfaces, such as lung, skin, and gastrointestinal tract. Giardia lamblia is an extracellular protozoan pathogen that inhabits the human small intestine. In this study, ILCs prepared from the lamina propria of mouse small intestine were incubated with G. lamblia trophozoites. Transcriptional changes in G. lamblia-exposed ILCs resulted in identification of activation of several immune pathways. Secretion of interleukin (IL)-17A, IL-17F, IL-1β, and interferon-γ was increased, whereas levels of IL-13, IL-5, and IL22, was maintained or reduced upon exposure to G. lamblia. Goup 3 ILC (ILC3) was found to be dominant amongst the ILCs, and increased significantly upon co-cultivation with G. lamblia trophozoites. Oral inoculation of G. lamblia trophozoites into mice resulted in their presence in the small intestine, of which, the highest number of parasites was detected at the 5 days-post infection. Increased ILC3 was observed amongst the ILC population at the 5 days-post infection. These findings indicate that ILC3 from the lamina propria secretes IL-17 in response to G. lamblia, leading to the intestinal pathology observed in giardiasis.
Collapse
Affiliation(s)
- Hye-Yeon Lee
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Eun-Ah Park
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Kyung-Jo Lee
- Department of Life Science, Sogang University, Seoul 04107, Korea
| | - Kyu-Ho Lee
- Department of Life Science, Sogang University, Seoul 04107, Korea
| | - Soon-Jung Park
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
44
|
Abstract
Transcription factors (TFs) are proteins that control the transcription of genetic information from DNA to mRNA by binding to specific DNA sequences either on their own or with other proteins as a complex. TFs thus support or suppress the recruitment of the corresponding RNA polymerase. In general, TFs are classified by structure or function. The TF, Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), is expressed in all cell types and tissues. NF-κB signaling and crosstalk are involved in several steps of carcinogenesis including in sequences involving pathogenic stimulus, chronic inflammation, fibrosis, establishment of its remodeling to the precancerous niche (PCN) and transition of a normal cell to a cancer cell. Triggered by various inflammatory cytokines, NF-κB is activated along with other TFs with subsequent stimulation of cell proliferation and inhibition of apoptosis. The involvement of NF-κB in carcinogenesis provides an opportunity to develop anti-NF-κB therapies. The complexity of these interactions requires that we elucidate those aspects of NF-κB interactions that play a role in carcinogenesis, the sequence of events leading to cancer.
Collapse
|
45
|
Osaki LH, Bockerstett KA, Wong CF, Ford EL, Madison BB, DiPaolo RJ, Mills JC. Interferon-γ directly induces gastric epithelial cell death and is required for progression to metaplasia. J Pathol 2019; 247:513-523. [PMID: 30511397 DOI: 10.1002/path.5214] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/14/2018] [Accepted: 11/29/2018] [Indexed: 12/26/2022]
Abstract
Chronic inflammation of the gastric mucosa, often caused by autoimmune gastritis and/or infection with Helicobacter pylori, can lead to atrophy of acid-secreting parietal cells with metaplasia of remaining cells. The histological pattern marks a critical step in the progression from chronic gastritis to gastric cancer, yet underlying mechanism(s) of inflammation-induced cell death of gastric epithelial cells are poorly understood. We investigated direct effects of a type 1 cytokine associated with autoimmunity and infection, interferon-γ (IFN-γ), on gastric epithelial cells. IFN-γ was applied to three-dimensional organoid cultures of gastric epithelial cells derived from gastric corpus gland (gastroids) of control and IFN-γ receptor-deficient mice. Gastroids were also treated with supernatants from activated immune cells isolated from a mouse model of autoimmune-mediated atrophic gastritis (TxA23) with and without IFN-γ expression. Finally, histopathological analysis of atrophy and metaplasia severity was performed in TxA23 mice and compared to TxA23 × Ifng-/- mice. Gastric epithelial cells in gastroid cultures expressed IFN-γ receptor in the basolateral membrane, and gastroids died when treated with IFN-γ in an IFN-γ receptor-dependent manner. Supernatants from immune cells containing high levels of IFN-γ were highly toxic to gastroids, and toxicity was tempered when IFN-γ was either neutralized using a monoclonal antibody or when supernatants from Ifng-/- mouse immune cells were used. Finally, TxA23 × Ifng-/- mice showed near-complete abrogation of pre-cancerous histopathological atrophy and metaplasia versus IFN-γ-sufficient controls. We identify IFN-γ as a critical promoter of parietal cell atrophy with metaplasia during the progression of gastritis to gastric atrophy and metaplasia. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Luciana H Osaki
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Kevin A Bockerstett
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Chun F Wong
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Eric L Ford
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Blair B Madison
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Richard J DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Jason C Mills
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA.,Developmental Biology, Pathology & Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
46
|
Genetic Polymorphisms in Inflammatory and Other Regulators in Gastric Cancer: Risks and Clinical Consequences. Curr Top Microbiol Immunol 2019; 421:53-76. [PMID: 31123885 DOI: 10.1007/978-3-030-15138-6_3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori infection is associated with the development of a chronic inflammatory response, which may induce peptic ulcers, gastric cancer (GC), and mucosa-associated lymphoid tissue (MALT) lymphoma. Chronic H. pylori infection promotes the genetic instability of gastric epithelial cells and interferes with the DNA repair systems in host cells. Colonization of the stomach with H. pylori is an important cause of non-cardia GC and gastric MALT lymphoma. The reduction of GC development in patients who underwent anti-H. pylori eradication schemes has also been well described. Individual susceptibility to GC development depends on the host's genetic predisposition, H. pylori virulence factors, environmental conditions, and geographical determinants. Biological determinants are urgently sought to predict the clinical course of infection in individuals with confirmed H. pylori infection. Possible candidates for such biomarkers include genetic aberrations such as single-nucleotide polymorphisms (SNPs) found in various cytokines/growth factors (e.g., IL-1β, IL-2, IL-6, IL-8, IL-10, IL-13, IL-17A/B, IFN-γ, TNF, TGF-β) and their receptors (IL-RN, TGFR), innate immunity receptors (TLR2, TLR4, CD14, NOD1, NOD2), enzymes involved in signal transduction cascades (PLCE1, PKLR, PRKAA1) as well as glycoproteins (MUC1, PSCA), and DNA repair enzymes (ERCC2, XRCC1, XRCC3). Bacterial determinants related to GC development include infection with CagA-positive (particularly with a high number of EPIYA-C phosphorylation motifs) and VacA-positive isolates (in particular s1/m1 allele strains). The combined genotyping of bacterial and host determinants suggests that the accumulation of polymorphisms favoring host and bacterial features increases the risk for precancerous and cancerous lesions in patients.
Collapse
|
47
|
Hagen SJ, Ang LH, Zheng Y, Karahan SN, Wu J, Wang YE, Caron T, Gad A, Muthupalani S, Fox JG. Loss of Tight Junction Protein Claudin 18 Promotes Progressive Neoplasia Development in Mouse Stomach. Gastroenterology 2018; 155:1852-1867. [PMID: 30195448 PMCID: PMC6613545 DOI: 10.1053/j.gastro.2018.08.041] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 08/12/2018] [Accepted: 08/24/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Loss of claudin 18 (CLDN18), a membrane-spanning tight junction protein, occurs during early stages of development of gastric cancer and associates with shorter survival times of patients. We investigated whether loss of CLDN18 occurs in mice that develop intraepithelial neoplasia with invasive glands due to infection with Helicobacter pylori, and whether loss is sufficient to promote the development of similar lesions in mice with or without H pylori infection. METHODS We performed immunohistochemical analyses in levels of CLDN18 in archived tissues from B6:129 mice infected with H pylori for 6 to 15 months. We analyzed gastric tissues from B6:129S5-Cldn18tm1Lex/Mmucd mice, in which the CLDN18 gene was disrupted in gastric tissues (CLDN18-knockout mice), or from control mice with a full-length CLDN18 gene (CLDN18+/+; B6:129S5/SvEvBrd) or heterozygous disruption of CLDN18 (CLDN18+/-; B6:129S5/SvEvBrd) that were infected with H pylori SS1 or PMSS1 at 6 weeks of age and tissues collected for analysis at 20 and 30 weeks after infection. Tissues from CLDN18-knockout mice and control mice with full-length CLDN18 gene expression were also analyzed without infection at 7 weeks and 2 years after birth. Tissues from control and CLDN18-knockout mice were analyzed by electron microscopy, stained by conventional methods and analyzed for histopathology, prepared by laser capture microdissection and analyzed by RNAseq, and immunostained for lineage markers, proliferation markers, and stem cell markers and analyzed by super-resolution or conventional confocal microscopy. RESULTS CLDN18 had a basolateral rather than apical tight junction localization in gastric epithelial cells. B6:129 mice infected with H pylori, which developed intraepithelial neoplasia with invasive glands, had increasing levels of CLDN18 loss over time compared with uninfected mice. In B6:129 mice infected with H pylori compared with uninfected mice, CLDN18 was first lost from most gastric glands followed by disrupted and reduced expression in the gastric neck and in surface cells. Gastric tissues from CLDN18-knockout mice had low levels of inflammation but increased cell proliferation, expressed markers of intestinalized proliferative spasmolytic polypeptide-expressing metaplasia, and had defects in signal transduction pathways including p53 and STAT signaling by 7 weeks after birth compared with full-length CLDN18 gene control mice. By 20 to 30 weeks after birth, gastric tissues from uninfected CLDN18-knockout mice developed intraepithelial neoplasia that invaded the submucosa; by 2 years, gastric tissues contained large and focally dysplastic polypoid tumors with invasive glands that invaded the serosa. CONCLUSIONS H pylori infection of B6:129 mice reduced the expression of CLDN18 early in gastric cancer progression, similar to previous observations from human gastric tissues. CLDN18 regulates cell lineage differentiation and cellular signaling in mouse stomach; CLDN18-knockout mice develop intraepithelial neoplasia and then large and focally dysplastic polypoid tumors in the absence of H pylori infection.
Collapse
Affiliation(s)
- Susan J. Hagen
- Department of Surgery/Division of General Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Lay-Hong Ang
- Department of Surgery/Division of General Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA,Harvard Medical School, Boston, MA 02115, USA
| | - Yi Zheng
- Department of Surgery/Division of General Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA,Harvard Medical School, Boston, MA 02115, USA,Present address: Perkin-Elmer Corporation, Hopkinton, MA 01748, USA
| | - Salih N. Karahan
- Department of Surgery/Division of General Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA,Dr. Karahan was a visiting medical student from the Koç University School of Medicine, Bakirkoy, Istanbul,TURKEY
| | - Jessica Wu
- Department of Surgery/Division of General Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA,Present address: Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yaoyu E. Wang
- Harvard Medical School, Boston, MA 02115, USA,Center for Cancer Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02130 USA
| | - Tyler Caron
- Department of Surgery/Division of General Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA,Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA,Present address: Broad Institute, Cambridge, MA 02142, USA
| | - Aniket Gad
- Department of Surgery/Division of General Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Sureshkumar Muthupalani
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
48
|
Thaper D, Prabha V. Molecular mimicry: An explanation for autoimmune diseases and infertility. Scand J Immunol 2018; 88:e12697. [PMID: 29935034 DOI: 10.1111/sji.12697] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/20/2018] [Indexed: 01/01/2023]
Abstract
Microorganisms execute an enthralling range of adjustments to survive in the host. Among the various strategies employed by microorganisms to surmount the host immune response, the phenomenon of molecular mimicry empowers the microorganisms to manoeuvre host physiology and cellular functions for their own advantage by mimicking the host proteins and initiating autoimmunity. This phenomena, by and large, has been studied in context of autoimmune diseases; however, its implications have also been reported in infertility. Hence, in this article, we provide a review of the various instances of molecular mimicry initiated by bacteria, parasites and viruses in the world of autoimmune diseases and infertility.
Collapse
Affiliation(s)
- Deepali Thaper
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Vijay Prabha
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
49
|
Liu MJ, Jiang K, Zhang J, Zhou L, Zhao JW, Wang BM. RIP3 mediates IL-33 production in gastric epithelial cells with intestinal metaplasia. Shijie Huaren Xiaohua Zazhi 2018; 26:964-971. [DOI: 10.11569/wcjd.v26.i16.964] [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] [Indexed: 02/06/2023] Open
Abstract
AIM To explore the relationship between receptor-interacting protein kinase 3 (RIP3) signaling pathway and gastric intestinal metaplasia (GIM), and the regulatory effect of this signaling pathway on inflammatory cytokines.
METHODS Gastric tissues from healthy controls, patients with chronic non-atrophic gastritis, patients with GIM, and patients with dysplasia were collected to detect the expression of RIP3 in GIM by immunohistochemistry and RT-PCR. Human gastric epithelial cell line GES-1 was stimulated with sodium deoxycholate (DCA) to observe the relationship between CDX2, a key gene involved in intestinal metaplasia, and RIP3 signaling pathway. The regulation of inflammatory cytokines by RIP3 was also assessed.
RESULTS Compared with the control and chronic non-atrophic gastritis groups, the expression of RIP3 mRNA in the gastric mucosa of GIM patients and dysplasia patients was up-regulated, and the expression of RIP3 protein in the gastric epithelium of GIM patients and dysplasia patients was also up-regulated. In GES-1 cells stimulated with DCA, the expression of CDX2 protein and the RIP3 signaling pathway-associated proteins was increased in a concentration-dependent manner, accompanied by up-regulation of IL-33 expression. Necrostatin-1 (Nec-1), a specific inhibitor of the RIP3 signaling pathway, had no effect on CDX2 expression, but significantly down-regulated the expression of RIP3 and IL-33.
CONCLUSION RIP3 has no effect on the occurrence of GIM, but it may affect GIM progression by regulating the expression of IL-33 in gastric epithelial cells with intestinal metaplasia, suggesting that it may be a potential therapeutic target for preventing GIM progression.
Collapse
Affiliation(s)
- Meng-Jing Liu
- Department of Gastroenterology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Kui Jiang
- Department of Gastroenterology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Jun Zhang
- Department of Gastroenterology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Lu Zhou
- Department of Gastroenterology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Jing-Wen Zhao
- Department of Gastroenterology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| | - Bang-Mao Wang
- Department of Gastroenterology, General Hospital of Tianjin Medical University, Tianjin 300052, China
| |
Collapse
|
50
|
Petersen CP, Meyer AR, DeSalvo C, Choi E, Schlegel C, Petersen A, Engevik AC, Prasad N, Levy SE, Peebles RS, Pizarro TT, Goldenring JR. A signalling cascade of IL-33 to IL-13 regulates metaplasia in the mouse stomach. Gut 2018; 67:805-817. [PMID: 28196875 PMCID: PMC5681443 DOI: 10.1136/gutjnl-2016-312779] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/13/2017] [Accepted: 01/15/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Alternatively activated macrophages (M2) are associated with the progression of spasmolytic polypeptide-expressing metaplasia (SPEM) in the stomach. However, the precise mechanism(s) and critical mediators that induce SPEM are unknown. DESIGN To determine candidate genes important in these processes, macrophages from the stomach corpus of mice with SPEM (DMP-777-treated) or advanced SPEM (L635-treated) were isolated and RNA sequenced. Effects on metaplasia development after acute parietal cell loss induced by L635 were evaluated in interleukin (IL)-33, IL-33 receptor (ST2) and IL-13 knockout (KO) mice. RESULTS Profiling of metaplasia-associated macrophages in the stomach identified an M2a-polarised macrophage population. Expression of IL-33 was significantly upregulated in macrophages associated with advanced SPEM. L635 induced metaplasia in the stomachs of wild-type mice, but not in the stomachs of IL-33 and ST2 KO mice. While IL-5 and IL-9 were not required for metaplasia induction, IL-13 KO mice did not develop metaplasia in response to L635. Administration of IL-13 to ST2 KO mice re-established the induction of metaplasia following acute parietal cell loss. CONCLUSIONS Metaplasia induction and macrophage polarisation after parietal cell loss is coordinated through a cytokine signalling network of IL-33 and IL-13, linking a combined response to injury by both intrinsic mucosal mechanisms and infiltrating M2 macrophages.
Collapse
Affiliation(s)
- Christine P. Petersen
- Departments of Cell and Developmental Biology, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Anne R. Meyer
- Departments of Cell and Developmental Biology, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Carlo DeSalvo
- Department of Pathology, Case Western Reserve School of Medicine, Cleveland, OH
| | - Eunyoung Choi
- Department of Nashville VA Medical Center, Vanderbilt University, Nashville, TN,Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Cameron Schlegel
- Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Alec Petersen
- Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Amy C. Engevik
- Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| | - Nripesh Prasad
- Department of HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | - Shawn E. Levy
- Department of HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | | | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve School of Medicine, Cleveland, OH
| | - James R. Goldenring
- Department of Nashville VA Medical Center, Vanderbilt University, Nashville, TN,Departments of Cell and Developmental Biology, Vanderbilt University, Nashville, TN,Department of Surgery, Vanderbilt University, Nashville, TN,Department of Epithelial Biology Center, Vanderbilt University, Nashville, TN
| |
Collapse
|