1
|
Koley S, Mukherjee M. Comprehensive analysis of multiple cytokines to stratify uropathogenic Escherichia coli pathogenesis in mouse model of urinary tract infection. Cytokine 2024; 178:156577. [PMID: 38479049 DOI: 10.1016/j.cyto.2024.156577] [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: 09/30/2023] [Revised: 02/27/2024] [Accepted: 03/06/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE Urinary tract infection (UTI) is one of the most common human bacterial infections primarily caused by uropathogenic E. coli (UPEC). Empiric treatment in UTI cause emergence of multidrug resistance and limit treatment options. Understanding UTI at the molecular level with respect to the causative pathogen as well as subsequent host response pose an absolute necessity towards appropriate clinical management. This study aimed to investigate host cytokine response in mouse UTI model with respect to bacterial colonization and associated virulence gene expression upon infection. METHOD Mouse UTI model was established with two clinical UPEC isolates E. coli NP105 and E. coli P025. UPEC colonization in bladder and kidney was evaluated by bacterial culture (CFU/ml). Histopathology of the tissues were examined by hematoxylin and eosin staining. PCR and real time PCR were used to detect the incidence and expression of respective bacterial genes. Cytokine concentrations in tissues and sera were evaluated using ELISA. GraphPad prism version 8.0.2 was used for statistical interpretation. RESULT Highest bacterial colonization was observed on 7th and 9th day post infection (p.i). in bladder and kidney of mouse infected with E. coli P025 and E. coli NP105 respectively with a distinct difference in relative expression of fimH and papC adhesin genes in vivo. IL-1β level in tissues and sera of E. coli NP105 and E. coli P025 infected mouse was significantly different but the IL-17A, GCSF, TGF-β levels were comparable. CONCLUSION These findings show a role of IL1β to stratify pathogenicity of UPEC in mouse UTI model.
Collapse
Affiliation(s)
- Snehashis Koley
- Department of Biochemistry and Medical Biotechnology, School of Tropical Medicine, Kolkata, 700073
| | - Mandira Mukherjee
- Department of Biochemistry and Medical Biotechnology, School of Tropical Medicine, Kolkata, 700073.
| |
Collapse
|
2
|
Ke Y, Tan C, Zhen J, Dong W. Global status and trends of gastric cancer and gastric microbiota research: a bibliometric analysis. Front Microbiol 2024; 15:1341012. [PMID: 38655079 PMCID: PMC11037409 DOI: 10.3389/fmicb.2024.1341012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/12/2024] [Indexed: 04/26/2024] Open
Abstract
Background Numerous studies have cast light on the relationship between the gastric microbiota and gastric carcinogenesis. In this study, we conducted a bibliometric analysis of the relevant literature in the field of gastric cancer and the gastric microbiota and clarified its research status, hotspots, and development trends. Materials and methods Publications were retrieved from the Web of Science Core Collection on 18 July 2023. CiteSpace 6.2.R4, VOSviewer 1.6.19.0, and Biblioshiny were used for the co-occurrence and cooperation analyses of countries, institutions, authors, references, and keywords. A keyword cluster analysis and an emergence analysis were performed, and relevant knowledge maps were drawn. Results The number of published papers in this field totaled 215 and showed an increasing trend. The analysis of funding suggested that the input in this field is increasing steadily. China had the highest number of publications, while the United States had the highest betweenness centrality. Baylor College of Medicine published the most articles cumulatively. Both Ferreira RM and Cooker OO had the highest citation frequency. The journal Helicobacter showed the most interest in this field, while Gut provided a substantial research foundation. A total of 280 keywords were obtained using CiteSpace, which were primarily focused on the eradication and pathogenic mechanisms of Helicobacter pylori, as well as the application of the gastric microbiota in the evaluation and treatment of gastric cancer. The burst analysis suggested that in the future, research may focus on the application of gastric microorganisms, particularly Fusobacterium nucleatum, in the diagnosis and treatment of gastric cancer, along with their pathogenic mechanisms. Conclusion Current studies have been tracking the eradication of Helicobacter pylori and its pathogenic mechanisms, as well as changes in the gastric microbiota during gastric carcinogenesis. Future research may focus on the clinical application and pathogenesis of stomach microorganisms through bacteria such as Fusobacterium nucleatum.
Collapse
Affiliation(s)
- Yujia Ke
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junhai Zhen
- Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
3
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
4
|
Zhang X, He Y, Zhang X, Fu B, Song Z, Wang L, Fu R, Lu X, Xing J, Lv J, Guo M, Huo X, Liu X, Lu J, Du X, Ge Z, Chen Z, Li C. Sustained exposure to Helicobacter pylori induces immune tolerance by desensitizing TLR6. Gastric Cancer 2024; 27:324-342. [PMID: 38310631 PMCID: PMC10896808 DOI: 10.1007/s10120-023-01461-7] [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: 06/27/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024]
Abstract
Helicobacter pylori (H. pylori, Hp) has been designated a class I carcinogen and is closely associated with severe gastric diseases. During colonization in the gastric mucosa, H. pylori develops immune escape by inducing host immune tolerance. The gastric epithelium acts as the first line of defense against H. pylori, with Toll-like receptors (TLRs) in gastric epithelial cells being sensitive to H. pylori components and subsequently activating the innate immune system. However, the mechanism of immune tolerance induced by H. pylori through the TLR signalling pathway has not been fully elucidated. In this research, we detected the expression of TLRs and inflammatory cytokines in GES-1 cells upon sustained exposure to H. pylori or H. pylori lysate from 1 to 30 generations and in Mongolian gerbils infected with H. pylori for 5 to 90 weeks. We found that the levels of TLR6 and inflammatory cytokines first increased and then dropped during the course of H. pylori treatment in vitro and in vivo. The restoration of TLR6 potentiated the expression of IL-1β and IL-8 in GES-1 cells, which recruited neutrophils and reduced the colonization of H. pylori in the gastric mucosa of gerbils. Mechanistically, we found that persistent infection with H. pylori reduces the sensitivity of TLR6 to bacterial components and regulates the expression of inflammatory cytokines in GES-1 cells through TLR6/JNK signaling. The TLR6 agonist obviously alleviated inflammation in vitro and in vivo. Promising results suggest that TLR6 may be a potential candidate immunotherapy drug for H. pylori infection.
Collapse
Affiliation(s)
- Xiulin Zhang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, Beijing, People's Republic of China
| | - Yang He
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
- School of Nursing, Dalian Medical University, Dalian, People's Republic of China
| | - Xiaolu Zhang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Bo Fu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Zidai Song
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Liang Wang
- Peking University Ninth School of Clinical Medicine, Beijing, People's Republic of China
| | - Rui Fu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Xuancheng Lu
- Laboratory Animal Center, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Jin Xing
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Jianyi Lv
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Meng Guo
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Xueyun Huo
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Xin Liu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Jing Lu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Xiaoyan Du
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, USA
| | - Zhenwen Chen
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China
| | - Changlong Li
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Science, Beijing Key Laboratory of Cancer Invasion & Metastasis Research, Laboratory for Clinical Medicine, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
5
|
Reuter S, Raspe J, Taube C. Microbes little helpers and suppliers for therapeutic asthma approaches. Respir Res 2024; 25:29. [PMID: 38218816 PMCID: PMC10787474 DOI: 10.1186/s12931-023-02660-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024] Open
Abstract
Bronchial asthma is a prevalent and increasingly chronic inflammatory lung disease affecting over 300 million people globally. Initially considered an allergic disorder driven by mast cells and eosinophils, asthma is now recognized as a complex syndrome with various clinical phenotypes and immunological endotypes. These encompass type 2 inflammatory endotypes characterized by interleukin (IL)-4, IL-5, and IL-13 dominance, alongside others featuring mixed or non-eosinophilic inflammation. Therapeutic success varies significantly based on asthma phenotypes, with inhaled corticosteroids and beta-2 agonists effective for milder forms, but limited in severe cases. Novel antibody-based therapies have shown promise, primarily for severe allergic and type 2-high asthma. To address this gap, novel treatment strategies are essential for better control of asthma pathology, prevention, and exacerbation reduction. One promising approach involves stimulating endogenous anti-inflammatory responses through regulatory T cells (Tregs). Tregs play a vital role in maintaining immune homeostasis, preventing autoimmunity, and mitigating excessive inflammation after pathogenic encounters. Tregs have demonstrated their ability to control both type 2-high and type 2-low inflammation in murine models and dampen human cell-dependent allergic airway inflammation. Furthermore, microbes, typically associated with disease development, have shown immune-dampening properties that could be harnessed for therapeutic benefits. Both commensal microbiota and pathogenic microbes have demonstrated potential in bacterial-host interactions for therapeutic purposes. This review explores microbe-associated approaches as potential treatments for inflammatory diseases, shedding light on current and future therapeutics.
Collapse
Affiliation(s)
- Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany.
| | - Jonas Raspe
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
| |
Collapse
|
6
|
Zhang X, Zhang K, Yan L, Wang P, Zhao F, Hu S. The role of toll-like receptors in immune tolerance induced by Helicobacter pylori infection. Helicobacter 2023; 28:e13020. [PMID: 37691007 DOI: 10.1111/hel.13020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Helicobacter pylori (H. pylori) is a gram-negative, microaerobic bacterium that colonizes the gastric mucosa in about half of the world's population. H. pylori infection can lead to various diseases. Chronic infection by H. pylori exposes the gastric mucosa to bacterial components such as lipopolysaccharide (LPS), outer membrane vesicles (OMVs), and several toxic proteins. Infected with H. pylori activates the release of pro-inflammatory factors and triggers inflammatory responses that damage the gastric mucosa. As the only microorganism that permanently colonizes the human stomach, H. pylori can suppress host immunity to achieve long-term colonization. Toll-like receptors (TLRs) play a crucial role in T-cell activation, promoting innate immune responses and immune tolerance during H. pylori infection. Among the 10 TLRs found in humans, TLR2, TLR4, TLR5, and TLR9 have been thoroughly investigated in relation to H. pylori-linked immune regulation. In the present review, we provide a comprehensive analysis of the various mechanisms employed by different TLRs in the induction of immune tolerance upon H. pylori infection, which will contribute to the research of pathogenic mechanism of H. pylori.
Collapse
Affiliation(s)
- Xiulin Zhang
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Ke Zhang
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Linlin Yan
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Pengfei Wang
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Fan Zhao
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| | - Shoukui Hu
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, China
| |
Collapse
|
7
|
Liu Q, Chen C, He Y, Mai W, Ruan S, Ning Y, Li Y. Notch Signaling Regulates the Function and Phenotype of Dendritic Cells in Helicobacter pylori Infection. Microorganisms 2023; 11:2818. [PMID: 38004829 PMCID: PMC10673485 DOI: 10.3390/microorganisms11112818] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Notch signaling manipulates the function and phenotype of dendritic cells (DCs), as well as the interaction between DCs and CD4+ T cells. However, the role of Notch signaling in Helicobacter pylori (H. pylori) infection remains elusive. Murine bone marrow-derived dendritic cells (BMDCs) were pretreated in the absence or presence of Notch signaling inhibitor DAPT prior to H. pylori stimulation and the levels of Notch components, cytokines and surface markers as well as the differentiation of CD4+ T cells in co-culture were measured using quantitative real-time PCR (qRT-PCR), Western blot, enzyme-linked immunosorbent assay (ELISA) and flow cytometry. Compared with the control, the mRNA expression of all Notch receptors and Notch ligands Dll4 and Jagged1 was up-regulated in H. pylori-stimulated BMDCs. The blockade of Notch signaling by DAPT influenced the production of IL-1β and IL-10 in H. pylori-pulsed BMDCs, and reduced the expression of Notch1, Notch3, Notch4, Dll1, Dll3 and Jagged2. In addition, DAPT pretreatment decreased the expression of maturation markers CD80, CD83, CD86, and major histocompatibility complex class II (MHC-II) of BMDCs, and further skewed Th17/Treg balance toward Treg. Notch signaling regulates the function and phenotype of DCs, thus mediating the differentiation of CD4+ T cells during H. pylori infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunshan Ning
- School of Laboratory Medicine and Biotechnology, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China (W.M.)
| | - Yan Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China (W.M.)
| |
Collapse
|
8
|
Fuchs S, Gong R, Gerhard M, Mejías-Luque R. Immune Biology and Persistence of Helicobacter pylori in Gastric Diseases. Curr Top Microbiol Immunol 2023; 444:83-115. [PMID: 38231216 DOI: 10.1007/978-3-031-47331-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Helicobacter pylori is a prevalent pathogen, which affects more than 40% of the global population. It colonizes the human stomach and persists in its host for several decades or even a lifetime, if left untreated. The persistent infection has been linked to various gastric diseases, including gastritis, peptic ulcers, and an increased risk for gastric cancer. H. pylori infection triggers a strong immune response directed against the bacterium associated with the infiltration of innate phagocytotic immune cells and the induction of a Th1/Th17 response. Even though certain immune cells seem to be capable of controlling the infection, the host is unable to eliminate the bacteria as H. pylori has developed remarkable immune evasion strategies. The bacterium avoids its killing through innate recognition mechanisms and manipulates gastric epithelial cells and immune cells to support its persistence. This chapter focuses on the innate and adaptive immune response induced by H. pylori infection, and immune evasion strategies employed by the bacterium to enable persistent infection.
Collapse
Affiliation(s)
- Sonja Fuchs
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Ruolan Gong
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich (TUM), Trogerstraße 30, 81675, Munich, Germany.
| |
Collapse
|
9
|
Helicobacter pylori promotes gastric cancer progression through the tumor microenvironment. Appl Microbiol Biotechnol 2022; 106:4375-4385. [PMID: 35723694 DOI: 10.1007/s00253-022-12011-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 02/05/2023]
Abstract
Gastric cancer (GC) is a leading type of cancer. Although immunotherapy has yielded important recent progress in the treatment of GC, the prognosis remains poor due to drug resistance and frequent recurrence and metastasis. There are multiple known risk factors for GC, and infection with Helicobacter pylori is one of the most significant. The mechanisms underlying the associations of H. pylori and GC remain unclear, but it is well known that infection can alter the tumor microenvironment (TME). The TME and the tumor itself constitute a complete ecosystem, and the TME plays critical roles in tumor progression, metastasis, and drug resistance. H. pylori infection can act synergistically with the TME to cause DNA damage and abnormal expression of multiple genes and activation of signaling pathways. It also modulates the host immune system in ways that enhance the proliferation and metastasis of tumor cells, promote epithelial-mesenchymal transition, inhibit apoptosis, and provide energy support for tumor growth. This review elaborates myriad ways that H. pylori infections promote the occurrence and progression of GC by influencing the TME, providing new directions for immunotherapy treatments for this important disease. KEY POINTS: • H. pylori infections cause DNA damage and affect the repair of the TME to DNA damage. • H. pylori infections regulate oncogenes or activate the oncogenic signaling pathways. • H. pylori infections modulate the immune system within the TME.
Collapse
|
10
|
Tan J, Taitz J, Sun SM, Langford L, Ni D, Macia L. Your Regulatory T Cells Are What You Eat: How Diet and Gut Microbiota Affect Regulatory T Cell Development. Front Nutr 2022; 9:878382. [PMID: 35529463 PMCID: PMC9067578 DOI: 10.3389/fnut.2022.878382] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Modern industrial practices have transformed the human diet over the last century, increasing the consumption of processed foods. Dietary imbalance of macro- and micro-nutrients and excessive caloric intake represent significant risk factors for various inflammatory disorders. Increased ingestion of food additives, residual contaminants from agricultural practices, food processing, and packaging can also contribute deleteriously to disease development. One common hallmark of inflammatory disorders, such as autoimmunity and allergies, is the defect in anti-inflammatory regulatory T cell (Treg) development and/or function. Treg represent a highly heterogeneous population of immunosuppressive immune cells contributing to peripheral tolerance. Tregs either develop in the thymus from autoreactive thymocytes, or in the periphery, from naïve CD4+ T cells, in response to environmental antigens and cues. Accumulating evidence demonstrates that various dietary factors can directly regulate Treg development. These dietary factors can also indirectly modulate Treg differentiation by altering the gut microbiota composition and thus the production of bacterial metabolites. This review provides an overview of Treg ontogeny, both thymic and peripherally differentiated, and highlights how diet and gut microbiota can regulate Treg development and function.
Collapse
Affiliation(s)
- Jian Tan
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jemma Taitz
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Shir Ming Sun
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Lachlan Langford
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Sydney Cytometry, The University of Sydney and The Centenary Institute, Sydney, NSW, Australia
- *Correspondence: Laurence Macia
| |
Collapse
|
11
|
Abstract
Transforming Growth Factor-β is a potent regulator of the immune system, acting at every stage from thymic differentiation, population of the periphery, control of responsiveness, tissue repair and generation of memory. It is therefore a central player in the immune response to infectious pathogens, but its contribution is often clouded by multiple roles acting on different cells in time and space. Hence, context is all-important in understanding when TGF-β is beneficial or detrimental to the outcome of infection. In this review, a full range of infectious agents from viruses to helminth parasites are explored within this framework, drawing contrasts and general conclusions about the importance of TGF-β in these diseases.
Collapse
Affiliation(s)
- Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
12
|
Gough NR, Xiang X, Mishra L. TGF-β Signaling in Liver, Pancreas, and Gastrointestinal Diseases and Cancer. Gastroenterology 2021; 161:434-452.e15. [PMID: 33940008 PMCID: PMC8841117 DOI: 10.1053/j.gastro.2021.04.064] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/05/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023]
Abstract
Genetic alterations affecting transforming growth factor-β (TGF-β) signaling are exceptionally common in diseases and cancers of the gastrointestinal system. As a regulator of tissue renewal, TGF-β signaling and the downstream SMAD-dependent transcriptional events play complex roles in the transition from a noncancerous disease state to cancer in the gastrointestinal tract, liver, and pancreas. Furthermore, this pathway also regulates the stromal cells and the immune system, which may contribute to evasion of the tumors from immune-mediated elimination. Here, we review the involvement of the TGF-β pathway mediated by the transcriptional regulators SMADs in disease progression to cancer in the digestive system. The review integrates human genomic studies with animal models that provide clues toward understanding and managing the complexity of the pathway in disease and cancer.
Collapse
Affiliation(s)
- Nancy R. Gough
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, New York
| | - Xiyan Xiang
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, New York
| | - Lopa Mishra
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, New York; Center for Translational Medicine, Department of Surgery, The George Washington University, Washington, District of Columbia.
| |
Collapse
|
13
|
Dos Santos Viana I, Cordeiro Santos ML, Santos Marques H, Lima de Souza Gonçalves V, Bittencourt de Brito B, França da Silva FA, Oliveira E Silva N, Dantas Pinheiro F, Fernandes Teixeira A, Tanajura Costa D, Oliveira Souza B, Lima Souza C, Vasconcelos Oliveira M, Freire de Melo F. Vaccine development against Helicobacter pylori: from ideal antigens to the current landscape. Expert Rev Vaccines 2021; 20:989-999. [PMID: 34139141 DOI: 10.1080/14760584.2021.1945450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022]
Abstract
Introduction: The interest of the world scientific community for an effective vaccine against Helicobacter pylori infection arises from its high prevalence and association with many diseases. Moreover, with an immunological response that is not always effective for the eradication of the bacteria and an increasing antibiotic resistance in the treatment of this infection, the search for a vaccine and new therapeutic modalities to control this infection is urgent.Areas covered: We bring an overview of the infection worldwide, discussing its prevalence, increasing resistance to antibiotics used in its therapy, in addition to the response of the immune system to the infection registered so far. Moreover, we address the most used antigens and their respective immunological responses expected or registered up to now. Finally, we address the trials and their partial results in development for such vaccines.Expert opinion: Although several studies for the development of an effective vaccine against this pathogen are taking place, many are still in the preclinical phase or even without updated information. In this sense, taking into account the high prevalence and association with important comorbidities, the interest of the pharmaceutical industry in developing an effective vaccine against this pathogen is questioned.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Davi Tanajura Costa
- Instituto Multidisciplinar Em Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | - Briza Oliveira Souza
- Instituto Multidisciplinar Em Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | - Cláudio Lima Souza
- Instituto Multidisciplinar Em Saúde, Universidade Federal da Bahia, Bahia, Brazil
| | | | | |
Collapse
|
14
|
Liu Z, Li J, Hu X, Xu H. Helicobacter pylori-induced protein tyrosine phosphatase receptor type C as a prognostic biomarker for gastric cancer. J Gastrointest Oncol 2021; 12:1058-1073. [PMID: 34295557 DOI: 10.21037/jgo-21-305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/17/2021] [Indexed: 12/14/2022] Open
Abstract
Background Helicobacter pylori (H. pylori) infection is closely associated with the tumorigenesis of gastric cancer. The aim of the present study was to identify the key regulator in H. pylori-related gastric cancer and to study the expression level and clinical value of the indicated key regulator in gastric cancer. Methods The GSE6143 dataset was used to identify differentially expressed genes (DEGs) with limma R package, and enrichment analysis was done using the Metascape web-based portal. The protein-protein interaction analysis was done using Search Tool for the Retrieval of Interacting Genes/Proteins. Gastric adenocarcinoma AGS and BGC-823 cells were treated with H. pylori strain 26695 to construct the in vitro H. pylori infection model, and quantitative reverse transcription polymerase chain reaction was used to analyze the mRNA levels of indicated genes. The correlation analysis between two genes in gastric cancer was done by GEPIA. Furthermore, the PTPRC expression by pathological features analysis was conducted in UALCAN, an easy to use, interactive web-portal (http://ualcan.path.uab.edu). The survival analysis for gastric cancer, based on PTPRC expression levels, was done using the Kaplan-Meier plotter. Results DEGs in gastric mucosa with or without H. pylori infection were identified and enriched in immune-related pathways and cancer pathways. The protein-protein interaction analysis confirmed the enrichment analysis of gene ontology. H. pylori strain 26695 exposure also confirmed the alteration of gene expression levels in AGS and BGC-823 cells. PTPRC was co-expressed with CSF2RB and TNFRSF7, indicating a significant positive correlation in gastric cancer. PTPRC was overexpressed in gastric cancer, and the overexpression of PTPRC was positively correlated with the progression of gastric cancer. Furthermore, the high expression of PTPRC could act as a poor prognostic factor for gastric cancer patients, especially for those at advanced stage. Conclusions H. pylori-induced PTPRC is overexpressed in gastric cancer, and the overexpression of PTPRC is positively associated with the development of gastric cancer. The high expression of PTPRC could serve as poor prognostic biomarker for gastric cancer patients, especially for those at advanced stage. H. pylori-induced PTPRC is a prognostic biomarker for gastric cancer.
Collapse
Affiliation(s)
- Zichuan Liu
- Department of Internal Medicine, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Jianchang Li
- Department of Internal Medicine, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Xiaoshan Hu
- Department of Gastrointestinal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Houwei Xu
- Department of Gastrointestinal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|