1
|
Chen W, Wang YJ. Multifaceted roles of OCT4 in tumor microenvironment: biology and therapeutic implications. Oncogene 2025:10.1038/s41388-025-03408-x. [PMID: 40229384 DOI: 10.1038/s41388-025-03408-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
OCT4 (Octamer-binding transcription factor 4, encoded by the POU5F1 gene) is a master transcription factor for maintaining the self-renewal and pluripotency of pluripotent stem cells, as well as a pioneer factor regulating epigenetics-driven cell reprogramming and cell fate conversion. It is also detected in a variety of cancer tissues and particularly in a small subpopulation of cancer cells known as cancer stem cells (CSCs). Accumulating evidence has revealed that CSCs are a dynamic population, exhibiting shift between multipotency and differentiation states, or quiescence and proliferation states. Such cellular plasticity of CSCs is profoundly influenced by dynamic interplay between CSCs and the tumor microenvironment (TME). Here, we review recent evidence showing that OCT4 expressed in CSCs plays a multifaceted role in shaping the TME by interacting with the cellular TME components, including cancer-associated fibroblasts, tumor endothelial cells, tumor-infiltrating immune cells, as well as the non-cellular TME components, such as extracellular matrix (ECM), metabolites, soluble factors (e.g., growth factors, cytokines and chemokines), and intra-tumoral microbiota. Together, OCT4 regulates crucial processes encompassing ECM remodeling, epithelial-mesenchymal transition, metabolic reprogramming, angiogenesis, and immune responses. The complex and bidirectional interactions between OCT4-expressing CSCs and the TME create a supportive niche for tumor growth, invasion, and resistance to therapy. Better understanding OCT4's roles in such interactions can provide deeper insights into potential therapeutic strategies and targets for disrupting the supportive environment of tumors. The emerging therapies targeting OCT4 in CSCs might hold promise to resensitize therapeutic-resistant cancer cells, and to eradicate all cancer cells when combined with other therapies targeting the bulk of differentiated cancer cells as well as the TME.
Collapse
Affiliation(s)
- Wenjie Chen
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying-Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Yan Z, Liu Y, Yuan Y. The plasticity of epithelial cells and its potential in the induced differentiation of gastric cancer. Cell Death Discov 2024; 10:512. [PMID: 39719478 DOI: 10.1038/s41420-024-02275-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Cell plasticity refers to the deviation of cells from normal terminal differentiation states when faced with environmental and genetic toxic stresses, resulting in the phenomenon of transforming into other cell or tissue phenotypes. Unlocking phenotype plasticity has been defined as a hallmark of malignant tumors. The stomach is one of the organs in the body with the highest degree of self-renewal and exhibits significant cell plasticity. In this paper, based on the review of the characteristics of normal differentiation of gastric epithelial cells and their markers, the four main phenotypes of gastric epithelial cell remodeling and their relationship with gastric cancer (GC) are drawn. Furthermore, we summarize the regulatory factors and mechanisms that affect gastric epithelial cell plasticity and outline the current status of research and future prospection for the treatment targeting gastric epithelial cell plasticity. This study has important theoretical reference value for the in-depth exploration of epithelial cell plasticity and the tumor heterogeneity caused by it, as well as for the precise treatment of GC.
Collapse
Affiliation(s)
- Ziwei Yan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yingnan Liu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China.
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Wu Q, Li T, Cui Y, Jiang H, Fu Y, Jiang Q, Ding X. Unveiling clinicopathologic features and outcomes for endoscopic submucosal dissection of early gastric cancer at gastric angulus in China. BMC Cancer 2024; 24:924. [PMID: 39080615 PMCID: PMC11290107 DOI: 10.1186/s12885-024-12610-1] [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: 03/19/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND With advances in endoscopic submucosal dissection (ESD) technique, an increasing number of the Chinese population are being diagnosed with early gastric cancers (EGCs) at gastric angulus. However, the relationship between gastric angulus and EGCs remains obscure. OBJECTIVES We aimed to unveil the unreported location characteristics of gastric angulus in Chinese EGC patients and the correlation between the degree of submucosal fibrosis and ESD outcomes. METHODS We retrospectively reviewed the medical records of EGC patients treated with ESD from January 2010 to March 2023. We retrospectively investigated and analyzed 740 EGC patients using multiple analyses. RESULTS Following gastric antrum (53.1%), the gastric angulus (21.8%) emerged as the second-most prevalent site for EGCs. It had highest incidence of severe submucosal fibrosis and ulceration than the other parts. Multivariate analysis showed independent associations of submucosal fibrosis at the angulus with ulceration (OR: 3.714, 95% CI: 1.041-13.249), procedure duration (OR: 1.037, 95% CI: 1.014-1.061), and perforation complication (OR: 14.611, 95% CI: 1.626-131.277) (all P < 0.05). CONCLUSIONS The gastric angulus demonstrates the highest incidence of severe submucosal fibrosis and ulceration for EGCs identified by ESD. This condition is linked to unfavorable outcomes, typically increased perforation risks and prolonged operation duration. Therefore, meticulous dissection is crucial for patients with EGCs in the gastric angulus.
Collapse
Affiliation(s)
- Qiaoyan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
| | - Tongyu Li
- Department of Hematology, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
| | - Yangyang Cui
- Department of Histopathology, Ningbo Diagnostic Pathology Center, No. 685 North Huancheng Road, Ningbo, Zhejiang Province, 315021, China
| | - Haizhong Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
| | - Yangbo Fu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
| | - Qi Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China
| | - Xiaoyun Ding
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China.
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, No. 59, Liuting Street, Ningbo, Zhejiang Province, 315010, China.
| |
Collapse
|
4
|
Wang Y, Zhang Z, Chen Q, Chen T. Simultaneous application of oral and intravaginal probiotics for Helicobacter pylori and its antibiotic-therapy-induced vaginal dysbacteriosis. NPJ Biofilms Microbiomes 2024; 10:49. [PMID: 38902244 PMCID: PMC11190290 DOI: 10.1038/s41522-024-00521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Helicobacter pylori is a prevalent bacterial pathogen globally, implicated in various gastrointestinal disorders. Current recommended antibiotic therapies for H. pylori infection have been proven to be therapeutically insufficient, with low eradication rates and high recurrence rates. Emerging evidence suggests that antibiotic therapy for H. pylori can lead to gastrointestinal and subsequent vaginal dysbiosis, posing challenges for conventional antibiotic approaches. Thus, this article proposes a novel probiotic therapy involving simultaneous oral and intra-vaginal probiotic administration alongside antibiotics for H. pylori treatment, aiming to enhance eradication rates and mitigate dysbiosis. We begin by providing an overview of gastrointestinal and vaginal microbiota and their interconnectedness through the vagina-gut axis. We then review the efficacy of current antibiotic regimens for H. pylori and discuss how antibiotic treatment impacts the vaginal microenvironment. To explore the feasibility of this approach, we evaluate the effectiveness of oral and intra-vaginal probiotics in restoring normal microbiota in the gastrointestinal and vaginal tracts, respectively. Additionally, we analyze the direct mechanisms by which oral and intra-vaginal probiotics act on their respective tracts and discuss potential cross-tract mechanisms. Considering the potential synergistic therapeutic effects of probiotics in both the gastrointestinal and vaginal tracts, dual-channel probiotic therapy holds promise as a more effective approach for H. pylori eradication and dysbiosis mitigation, presenting a novel concept in the collaborative treatment of gastrointestinal and genital disorders.
Collapse
Affiliation(s)
- Yufan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- National Engineering Research Centre for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Zhenyu Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Qi Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- National Engineering Research Centre for Bioengineering Drugs and Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
5
|
Krzysiek-Maczka G, Brzozowski T, Ptak-Belowska A. Helicobacter pylori-activated fibroblasts as a silent partner in gastric cancer development. Cancer Metastasis Rev 2023; 42:1219-1256. [PMID: 37460910 PMCID: PMC10713772 DOI: 10.1007/s10555-023-10122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/20/2023] [Indexed: 12/18/2023]
Abstract
The discovery of Helicobacter pylori (Hp) infection of gastric mucosa leading to active chronic gastritis, gastroduodenal ulcers, and MALT lymphoma laid the groundwork for understanding of the general relationship between chronic infection, inflammation, and cancer. Nevertheless, this sequence of events is still far from full understanding with new players and mediators being constantly identified. Originally, the Hp virulence factors affecting mainly gastric epithelium were proposed to contribute considerably to gastric inflammation, ulceration, and cancer. Furthermore, it has been shown that Hp possesses the ability to penetrate the mucus layer and directly interact with stroma components including fibroblasts and myofibroblasts. These cells, which are the source of biophysical and biochemical signals providing the proper balance between cell proliferation and differentiation within gastric epithelial stem cell compartment, when exposed to Hp, can convert into cancer-associated fibroblast (CAF) phenotype. The crosstalk between fibroblasts and myofibroblasts with gastric epithelial cells including stem/progenitor cell niche involves several pathways mediated by non-coding RNAs, Wnt, BMP, TGF-β, and Notch signaling ligands. The current review concentrates on the consequences of Hp-induced increase in gastric fibroblast and myofibroblast number, and their activation towards CAFs with the emphasis to the altered communication between mesenchymal and epithelial cell compartment, which may lead to inflammation, epithelial stem cell overproliferation, disturbed differentiation, and gradual gastric cancer development. Thus, Hp-activated fibroblasts may constitute the target for anti-cancer treatment and, importantly, for the pharmacotherapies diminishing their activation particularly at the early stages of Hp infection.
Collapse
Affiliation(s)
- Gracjana Krzysiek-Maczka
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Tomasz Brzozowski
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Agata Ptak-Belowska
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland
| |
Collapse
|
6
|
Matsuoka T, Yashiro M. The Role of the Transforming Growth Factor-β Signaling Pathway in Gastrointestinal Cancers. Biomolecules 2023; 13:1551. [PMID: 37892233 PMCID: PMC10605301 DOI: 10.3390/biom13101551] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Transforming growth factor-β (TGF-β) has attracted attention as a tumor suppressor because of its potent growth-suppressive effect on epithelial cells. Dysregulation of the TGF-β signaling pathway is considered to be one of the key factors in carcinogenesis, and genetic alterations affecting TGF-β signaling are extraordinarily common in cancers of the gastrointestinal system, such as hereditary nonpolyposis colon cancer and pancreatic cancer. Accumulating evidence suggests that TGF-β is produced from various types of cells in the tumor microenvironment and mediates extracellular matrix deposition, tumor angiogenesis, the formation of CAFs, and suppression of the anti-tumor immune reaction. It is also being considered as a factor that promotes the malignant transformation of cancer, particularly the invasion and metastasis of cancer cells, including epithelial-mesenchymal transition. Therefore, elucidating the role of TGF-β signaling in carcinogenesis, cancer invasion, and metastasis will provide novel basic insight for diagnosis and prognosis and the development of new molecularly targeted therapies for gastrointestinal cancers. In this review, we outline an overview of the complex mechanisms and functions of TGF-β signaling. Furthermore, we discuss the therapeutic potentials of targeting the TGF-β signaling pathway for gastrointestinal cancer treatment and discuss the remaining challenges and future perspectives on targeting this pathway.
Collapse
Affiliation(s)
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan;
| |
Collapse
|
7
|
Liu S, Deng Z, Zhu J, Ma Z, Tuo B, Li T, Liu X. Gastric immune homeostasis imbalance: An important factor in the development of gastric mucosal diseases. Biomed Pharmacother 2023; 161:114338. [PMID: 36905807 DOI: 10.1016/j.biopha.2023.114338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 03/11/2023] Open
Abstract
The gastric mucosal immune system is a unique immune organ independent of systemic immunity that not only maintains nutrient absorption but also plays a role in resisting the external environment. Gastric mucosal immune disorder leads to a series of gastric mucosal diseases, including autoimmune gastritis (AIG)-related diseases, Helicobacter pylori (H. pylori)-induced diseases, and various types of gastric cancer (GC). Therefore, understanding the role of gastric mucosal immune homeostasis in gastric mucosal protection and the relationship between mucosal immunity and gastric mucosal diseases is very important. This review focuses on the protective effect of gastric mucosal immune homeostasis on the gastric mucosa, as well as multiple gastric mucosal diseases caused by gastric immune disorders. We hope to offer new prospects for the prevention and treatment of gastric mucosal diseases.
Collapse
Affiliation(s)
- Shuhui Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zilin Deng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
8
|
Opila J, Krzysiek-Maczka G. Direct tool for quantitative analysis of cell/object dynamic behavior - metastasis and far beyond. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 229:107245. [PMID: 36455469 DOI: 10.1016/j.cmpb.2022.107245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/17/2022] [Accepted: 11/13/2022] [Indexed: 06/17/2023]
Abstract
INTRODUCTION The dynamics and depth of invasion as well as the ability of cancer cells to penetrate the walls of lymphatic or blood vessels represent critical survival-influencing factors in cancer patients. Depending on the cell type and tissue environment, cancer cell invasion differ in terms of motility mechanism and migration modes. Thus, there is the need of effective models allowing not only for single cell invasion potential assessment but also for collective migration and expansive growth evaluation in 3D microenvironment e.g. basement membranes. To meet this task, the specimens should be compared and analyzed in terms of the dynamics of movement and the evolution of the shape. OBJECTIVES Our main objective was development of the mathematical method that enables fast and credible calculation of parameters of shape and position, namely standard deviations (σX, σY), centroid position (μX, μY) and correlation coefficient ρ, based only on the contour of the aggregate. METHODS In order to accomplish this goal we measured geometrical properties of aggregates of RGM1 cells seeded in 3D Geltrex basement membrane. Referential microscopic images were taken 24 and 48 h after seeding and cell group dynamics was registered over 8 h periods using time lapse microscopy. RESULTS Based on gathered data, we managed to develop and fully test universal numerical tool allowing for estimation of statistical parameters of cell groups and aggregates which then allows for the precise evaluation of their behavior within microenvironment with time. CONCLUSION We conclude, that our tool is suitable for any research on the metastatic potential and motility of cancer cells in a given microenvironment, regardless of the migration mechanism, which together with the advanced analysis like cell single-cell transcriptomic, proteomic, and chromatin accessibility data may allow to identify precise targets for anti-cancer therapies, to predict the degree of malignancy of neoplastic lesions as well as it can be useful during architecting therapeutic strategies. Moreover, the developed tool seems to be broadly applicable for assessment of behavioural dynamics of any population.
Collapse
Affiliation(s)
- Janusz Opila
- Department of Applied Computer Sciences, The Faculty of Management, AGH University of Science and Technology, Cracow 30-059, Poland.
| | - Gracjana Krzysiek-Maczka
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, Cracow 31-531, Poland.
| |
Collapse
|
9
|
Naumann M, Ferino L, Sharafutdinov I, Backert S. Gastric Epithelial Barrier Disruption, Inflammation and Oncogenic Signal Transduction by Helicobacter pylori. Curr Top Microbiol Immunol 2023; 444:207-238. [PMID: 38231220 DOI: 10.1007/978-3-031-47331-9_8] [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 exemplifies one of the most favourable bacterial pathogens worldwide. The bacterium colonizes the gastric mucosa in about half of the human population and constitutes a major risk factor for triggering gastric diseases such as stomach cancer. H. pylori infection represents a prime example of chronic inflammation and cancer-inducing bacterial pathogens. The microbe utilizes a remarkable set of virulence factors and strategies to control cellular checkpoints of inflammation and oncogenic signal transduction. This chapter emphasizes on the pathogenicity determinants of H. pylori such as the cytotoxin-associated genes pathogenicity island (cagPAI)-encoded type-IV secretion system (T4SS), effector protein CagA, lipopolysaccharide (LPS) metabolite ADP-glycero-β-D-manno-heptose (ADP-heptose), cytotoxin VacA, serine protease HtrA, and urease, and how they manipulate various key host cell signaling networks in the gastric epithelium. In particular, we highlight the H. pylori-induced disruption of cell-to-cell junctions, pro-inflammatory activities, as well as proliferative, pro-apoptotic and anti-apoptotic responses. Here we review these hijacked signal transduction events and their impact on gastric disease development.
Collapse
Affiliation(s)
- Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto Von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Lorena Ferino
- Institute of Experimental Internal Medicine, Medical Faculty, Otto Von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Irshad Sharafutdinov
- Dept. Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany
| | - Steffen Backert
- Dept. Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany.
| |
Collapse
|
10
|
Jantaree P, Yu Y, Chaithongyot S, Täger C, Sarabi MA, Meyer TF, Boccellato F, Maubach G, Naumann M. Human gastric fibroblasts ameliorate A20-dependent cell survival in co-cultured gastric epithelial cells infected by Helicobacter pylori. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119364. [PMID: 36162648 DOI: 10.1016/j.bbamcr.2022.119364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Crosstalk within the gastric epithelium, which is closely in contact with stromal fibroblasts in the gastric mucosa, has a pivotal impact in proliferation, differentiation and transformation of the gastric epithelium. The human pathogen Helicobacter pylori colonises the gastric epithelium and represents a risk factor for gastric pathophysiology. Infection of H. pylori induces the activation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which is involved in the pro-inflammatory response but also in cell survival. In co-cultures with human gastric fibroblasts (HGF), we found that apoptotic cell death is reduced in the polarised human gastric cancer cell line NCI-N87 or in gastric mucosoids during H. pylori infection. Interestingly, suppression of apoptotic cell death in NCI-N87 cells involved an enhanced A20 expression regulated by NF-κB activity in response to H. pylori infection. Moreover, A20 acts as an important negative regulator of caspase-8 activity, which was suppressed in NCI-N87 cells during co-culture with gastric fibroblasts. Our results provide evidence for NF-κB-dependent regulation of apoptotic cell death in cellular crosstalk and highlight the protective role of gastric fibroblasts in gastric epithelial cell death during H. pylori infection.
Collapse
Affiliation(s)
- Phatcharida Jantaree
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Yanfei Yu
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Supattra Chaithongyot
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Christian Täger
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Mohsen Abdi Sarabi
- Department of Internal Medicine, Division of Cardiology and Angiology, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Thomas F Meyer
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian Albrechts University and University Hospital Schleswig Holstein, 24105 Kiel, Germany
| | - Francesco Boccellato
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Gunter Maubach
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
11
|
Fang Z, Meng Q, Xu J, Wang W, Zhang B, Liu J, Liang C, Hua J, Zhao Y, Yu X, Shi S. Signaling pathways in cancer-associated fibroblasts: recent advances and future perspectives. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 43:3-41. [PMID: 36424360 PMCID: PMC9859735 DOI: 10.1002/cac2.12392] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/20/2022] [Accepted: 11/04/2022] [Indexed: 11/26/2022]
Abstract
As a critical component of the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) play important roles in cancer initiation and progression. Well-known signaling pathways, including the transforming growth factor-β (TGF-β), Hedgehog (Hh), Notch, Wnt, Hippo, nuclear factor kappa-B (NF-κB), Janus kinase (JAK)/signal transducer and activator of transcription (STAT), mitogen-activated protein kinase (MAPK), and phosphoinositide 3-kinase (PI3K)/AKT pathways, as well as transcription factors, including hypoxia-inducible factor (HIF), heat shock transcription factor 1 (HSF1), P53, Snail, and Twist, constitute complex regulatory networks in the TME to modulate the formation, activation, heterogeneity, metabolic characteristics and malignant phenotype of CAFs. Activated CAFs remodel the TME and influence the malignant biological processes of cancer cells by altering the transcriptional and secretory characteristics, and this modulation partially depends on the regulation of signaling cascades. The results of preclinical and clinical trials indicated that therapies targeting signaling pathways in CAFs demonstrated promising efficacy but were also accompanied by some failures (e.g., NCT01130142 and NCT01064622). Hence, a comprehensive understanding of the signaling cascades in CAFs might help us better understand the roles of CAFs and the TME in cancer progression and may facilitate the development of more efficient and safer stroma-targeted cancer therapies. Here, we review recent advances in studies of signaling pathways in CAFs and briefly discuss some future perspectives on CAF research.
Collapse
Affiliation(s)
- Zengli Fang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Qingcai Meng
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jin Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Wei Wang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Bo Zhang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jiang Liu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Chen Liang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jie Hua
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Yingjun Zhao
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Si Shi
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China,Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| |
Collapse
|
12
|
Navashenaq JG, Shabgah AG, Banach M, Jamialahmadi T, Penson PE, Johnston TP, Sahebkar A. The interaction of Helicobacter pylori with cancer immunomodulatory stromal cells: New insight into gastric cancer pathogenesis. Semin Cancer Biol 2022; 86:951-959. [PMID: 34600095 DOI: 10.1016/j.semcancer.2021.09.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/20/2021] [Accepted: 09/26/2021] [Indexed: 01/27/2023]
Abstract
Gastric cancer is the fourth most common cause of cancer-linked deaths in the world. Gastric tumor cells have biological characteristics such as rapid proliferation, high invasiveness, and drug resistance, which result in recurrence and poor survival. Helicobacter pylori (H. pylori) has been proposed as a first-class carcinogen for gastric cancer according to the 1994 world health organization (WHO) classification. One of the important mechanisms by which H. pylori affects the gastric environment and promotes carcinogenesis is triggering inflammation. H. pylori induces an inflammatory response and a plethora of different signal transduction processes, leading to gastric mucosal disturbance, chronic gastritis, and a multi-step complex pathway that initiates carcinogenesis. It seems undeniable that the interaction between various cell types, including immune cells, gastric epithelium, glands, and stem cells, is vital for the progression and development of carcinogenesis concerning H. pylori. The interactions of H. pylori with surrounding cells play a key role in cancer progression. In this review, we discuss the interplay between H. pylori and tumor-supportive cells, including mesenchymal stem cells (MSCs), cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and myeloid derived-suppressor cells (MDSCs) in gastric cancer. It is hoped that clarifying the specific mechanisms for 'cross-talk' between H. pylori and these cells will provide promising strategies for developing new treatments.
Collapse
Affiliation(s)
| | | | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK; Liverpool Centre for Cardiovascular Science, Liverpool, UK
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Deng R, Zheng H, Cai H, Li M, Shi Y, Ding S. Effects of helicobacter pylori on tumor microenvironment and immunotherapy responses. Front Immunol 2022; 13:923477. [PMID: 35967444 PMCID: PMC9371381 DOI: 10.3389/fimmu.2022.923477] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/04/2022] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori is closely associated with gastric cancer. During persistent infection, Helicobacter pylori can form a microenvironment in gastric mucosa which facilitates the survival and colony formation of Helicobacter pylori. Tumor stromal cells are involved in this process, including tumor-associated macrophages, mesenchymal stem cells, cancer-associated fibroblasts, and myeloid-derived suppressor cells, and so on. The immune checkpoints are also regulated by Helicobacter pylori infection. Helicobacter pylori virulence factors can also act as immunogens or adjuvants to elicit or enhance immune responses, indicating their potential applications in vaccine development and tumor immunotherapy. This review highlights the effects of Helicobacter pylori on the immune microenvironment and its potential roles in tumor immunotherapy responses.
Collapse
Affiliation(s)
- Ruiyi Deng
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University First Medical School, Beijing, China
| | - Huiling Zheng
- Peking University Third Hospital, Department of Gastroenterology, Beijing, China
| | - Hongzhen Cai
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University First Medical School, Beijing, China
| | - Man Li
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University Third Medical School, Beijing, China
| | - Yanyan Shi
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
| | - Shigang Ding
- Peking University Third Hospital, Department of Gastroenterology, Beijing, China
| |
Collapse
|
14
|
He J, Hu W, Ouyang Q, Zhang S, He L, Chen W, Li X, Hu C. Helicobacter pylori infection induces stem cell-like properties in Correa cascade of gastric cancer. Cancer Lett 2022; 542:215764. [PMID: 35654291 DOI: 10.1016/j.canlet.2022.215764] [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: 01/10/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 02/09/2023]
Abstract
Gastric cancer (GC) is the fourth leading cause of cancer-related death. Its poor prognosis is attributed to unclear pathogenesis. Currently, the most widely accepted model for elucidating the mechanism of GC is the Correa cascade, which covers several histological lesions of the gastric mucosa. GC stem cells (CSCs) are crucial for oncogenesis in the Correa cascade and GC progression. As Helicobacter pylori (H. pylori) is the etiological factor in the Correa cascade, growing evidence suggests that enhancement of gastric stem cell-like properties and increase in CSCs correlate with H. pylori infection. In this paper, we review recent studies that present pathogenic mechanisms by which H. pylori induces gastric stem cell-like properties and CSCs, which may supplement the existing Correa model of GC. First, the dysfunction of developmental signaling pathways associated with H. pylori infection leads to the enhancement of gastric stemness. Second, H. pylori infection promotes alteration of the gastric mucosal microenvironment. In addition, epithelial-mesenchymal transition (EMT) may contribute to H. pylori-induced gastric stemness. Taken together, understanding these pathogeneses will provide potential therapeutic targets for the treatment of CSCs and malignant GC in H. pylori induced-Correa cascade of GC.
Collapse
Affiliation(s)
- JunJian He
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - WeiChao Hu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - ShengWei Zhang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - LiJiao He
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - WeiYan Chen
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - XinZhe Li
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - ChangJiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
15
|
Krzysiek-Maczka G, Targosz A, Wrobel T, Paw M, Szczyrk U, Opila J, Strzalka M, Wierdak M, Major P, Brzozowski T, Czyz J, Ptak-Belowska A. Time-extended exposure of gastric epithelial cells to secretome of Helicobacter pylori-activated fibroblasts induces reprogramming of gastric epithelium towards pre-cancerogenic and pro-invasive phenotype. Am J Cancer Res 2022; 12:1337-1371. [PMID: 35411238 PMCID: PMC8984895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/28/2021] [Indexed: 06/14/2023] Open
Abstract
Despite of the improvement in gastric cancer (GC) therapies patients still suffer from cancer recurrence and metastasis. Recently, the high ratio of these events combined with increased chemoresistance has been related to the asymptomatic Helicobacter pylori (Hp) infections. The limited efficiency of GC treatment strategies is also increasingly attributed to the activity of tumor stroma with the key role of cancer-associated fibroblasts (CAFs). In order to investigate the influence of Hp infection within stromal gastric tissue on cancer initiation and progression, we have exposed normal gastric epithelial cells to long-term influence of Hp-activated gastric fibroblast secretome. We have referred obtained results to this secretome influence on cancer cell lines. The invasive properties of cells were checked by time-lapse video microscopy and basement membrane assays. The expression of invasion-related factors was checked by RT-PCR, Western Blot, immunofluorescence and Elisa. Hp-activated gastric fibroblast secretome induced EMT type 3-related shifts of RGM1 cell phenotype; in particular it augmented their motility, cytoskeletal plasticity and invasiveness. These effects were accompanied by Snail1/Twist activation, the up-regulation of cytokeratin19/FAP/TNC/Integrin-β1 and MMPs, and by the induction of cMethigh/pEGFRhigh phenotype. Mechanistic studies suggest that this microevolution next to TGFβ relies also on c-Met/EGFR signaling interplay and engages HGF-Integrin-Ras-dependent Twist activation leading to MMP and TNC upregulation with subsequent positive auto- and paracrine feedback loops intensifying this process. Similar shifts were detected in cancer cells exposed to this secretome. Collectively, we show that the secretome of Hp-infected fibroblasts induces reprogramming/microevolution of epithelial and cancer cells towards type 3 EMT-related invasive phenotype in a manner reciprocally reliant next to TGFβ on cMet/Integrin-β1/p-EGFR-dependent axis. Apparently, the phenotypical plasticity of Hp-activated fibroblast reprogrammed gastric epithelial cells determines their susceptibility to the pro-invasive signaling, which results in re-organization of gastric niches and provides the cues for GC promotion/progression.
Collapse
Affiliation(s)
- Gracjana Krzysiek-Maczka
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College31-531 Cracow, Poland
| | - Aneta Targosz
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College31-531 Cracow, Poland
| | - Tomasz Wrobel
- Department of Cell Biology, The Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University30-387 Cracow, Poland
| | - Milena Paw
- Department of Cell Biology, The Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University30-387 Cracow, Poland
| | - Urszula Szczyrk
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College31-531 Cracow, Poland
| | - Janusz Opila
- Department of Applied Computer Sciences, The Faculty of Management, AGH University of Science and Technology30-059 Cracow, Poland
| | - Malgorzata Strzalka
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College31-531 Cracow, Poland
| | - Mateusz Wierdak
- Clinic of General, Oncological and Metabolic Surgery, 2nd Department of General Surgery, The Faculty of Medicine, Jagiellonian University Medical College30-688 Cracow, Poland
| | - Piotr Major
- Clinic of General, Oncological and Metabolic Surgery, 2nd Department of General Surgery, The Faculty of Medicine, Jagiellonian University Medical College30-688 Cracow, Poland
| | - Tomasz Brzozowski
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College31-531 Cracow, Poland
| | - Jarosław Czyz
- Department of Cell Biology, The Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University30-387 Cracow, Poland
| | - Agata Ptak-Belowska
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College31-531 Cracow, Poland
| |
Collapse
|
16
|
Senchukova MA, Tomchuk O, Shurygina EI. Helicobacter pylori in gastric cancer: Features of infection and their correlations with long-term results of treatment. World J Gastroenterol 2021; 27:6290-6305. [PMID: 34712033 PMCID: PMC8515796 DOI: 10.3748/wjg.v27.i37.6290] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/21/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is a spiral-shaped bacterium responsible for the development of chronic gastritis, gastric ulcer, gastric cancer (GC), and MALT-lymphoma of the stomach. H. pylori can be present in the gastric mucosa (GM) in both spiral and coccoid forms. However, it is not known whether the severity of GM contamination by various vegetative forms of H. pylori is associated with clinical and morphological characteristics and long-term results of GC treatment. AIM To establish the features of H. pylori infection in patients with GC and their correlations with clinical and morphological characteristics of diseases and long-term results of treatment. METHODS Of 109 patients with GC were included in a prospective cohort study. H. pylori in the GM and tumor was determined by rapid urease test and by immunohistochemically using the antibody to H. pylori. The results obtained were compared with the clinical and morphological characteristics and prognosis of GC. Statistical analysis was performed using the Statistica 10.0 software. RESULTS H. pylori was detected in the adjacent to the tumor GM in 84.5% of cases, of which a high degree of contamination was noted in 50.4% of the samples. Coccoid forms of H. pylori were detected in 93.4% of infected patients, and only coccoid-in 68.9%. It was found that a high degree of GM contamination by the coccoid forms of H. pylori was observed significantly more often in diffuse type of GC (P = 0.024), in poorly differentiated GC (P = 0.011), in stage T3-4 (P = 0.04) and in N1 (P = 0.011). In cases of moderate and marked concentrations of H. pylori in GM, a decrease in 10-year relapse free and overall survival from 55.6% to 26.3% was observed (P = 0.02 and P = 0.07, respectively). The relationship between the severity of the GM contamination by the spiral-shaped forms of H. pylori and the clinical and morphological characteristics and prognosis of GC was not revealed. CONCLUSION The data obtained indicates that H. pylori may be associated not only with induction but also with the progression of GC.
Collapse
Affiliation(s)
- Marina A Senchukova
- Department of Oncology, Orenburg State Medical University, Orenburg 460000, Russia
| | - Olesya Tomchuk
- Department of Histology, Cytology, Embryology, Orenburg State Medical University, Orenburg 460000, Russia
| | - Elena I Shurygina
- Department of Pathology, Orenburg State Medical University, Orenburg 460000, Russia
| |
Collapse
|