1
|
Wang L, Wang MH, Yuan YH, Xu RZ, Bai L, Wang MZ. Identification and validation of extracellular matrix-related genes in the progression of gastric cancer with intestinal metaplasia. World J Gastrointest Oncol 2025; 17:105160. [DOI: 10.4251/wjgo.v17.i6.105160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/31/2025] [Accepted: 04/23/2025] [Indexed: 06/13/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a highly lethal malignancy with a high incidence and mortality rate globally. Its development follows the Correa model, with intestinal metaplasia (IM) being a critical precursor to GC. However, the mechanisms underlying IM progression to GC remain unclear. This study explored extracellular matrix (ECM)-related gene changes during IM progression to GC, aiming to identify biomarkers that could improve early diagnosis and treatment strategies for GC, ultimately enhancing patient outcomes.
AIM To analyze transcriptome sequencing data, molecular biomarkers that can predict GC risk and monitor IM progression can be identified, providing new insights and strategies for preventing IM-GC transformation.
METHODS Weighted gene co-expression network analysis served for confirming gene modules. Upregulated ECM-related genes were further tested using univariate Cox regression and least absolute shrinkage and selection operator analysis to select hub genes and construct a survival analysis model. The intestinal cell model was established by stimulating GES-1 cells with chenodeoxycholic acid.
RESULTS Weighted gene co-expression network analysis identified 1709 differentially expressed genes from the GSE191275 dataset, while The Cancer Genome Atlas stomach adenocarcinoma revealed 4633 differentially expressed genes. The intersection of these datasets identified 71 upregulated and 171 downregulated genes, which were enriched in ECM-related pathways. Univariate Cox regression analysis identified six genes with prognostic significance, and least absolute shrinkage and selection operator regression pinpointed secreted protein acidic and rich in cysteine (SPARC) and SERPINE1 as non-zero coefficient genes. A prognostic model integrating clinical tumor node metastasis staging, age, SERPINE1, and SPARC was constructed. Immunohistochemistry analysis confirmed an increasing expression of SPARC protein from normal gastric mucosa (-), to IM (+- to +), and to GC (+ to ++), with significant differences (P < 0.05). Western blot analysis demonstrated significantly higher SPARC expression in induced intestinal cells compared to GES-1. Furthermore, after SPARC knockdown in the human GC cell line HGC27, cell counting kit-8 and colony formation assays showed a reduction in cell proliferative ability, while the wound healing assay revealed impaired cell migration capacity.
CONCLUSION Comprehensive analysis suggested that a model incorporating clinical tumor node metastasis staging, age, and SPARC/SERPINE1 expression served as a prognostic predictor for GC. Moreover, elevated SPARC expression in IM and GC suggests its potential as a proper biomarker to detect GC in early stage and as a novel therapeutic target, guiding clinical applications.
Collapse
Affiliation(s)
- Lu Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Baotou Medical College, Baotou Medical College, Baotou 300000, Inner Mongolia Autonomous Region, China
| | - Meng-Han Wang
- Baotou Medical College, Baotou 300000, Inner Mongolia Autonomous Region, China
| | - Yao-Hong Yuan
- Baotou Medical College, Baotou 300000, Inner Mongolia Autonomous Region, China
| | - Rui-Ze Xu
- Baotou Medical College, Baotou 300000, Inner Mongolia Autonomous Region, China
| | - Lu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Baotou Medical College, Baotou Medical College, Baotou 300000, Inner Mongolia Autonomous Region, China
| | - Mi-Zhu Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Baotou Medical College, Baotou 300000, Inner Mongolia Autonomous Region, China
| |
Collapse
|
2
|
Pellegrino R, Gravina AG. Potential of traditional Chinese medicine in gastrointestinal disorders: Hericium erinaceus in chronic atrophic gastritis. World J Gastroenterol 2025; 31:106615. [DOI: 10.3748/wjg.v31.i20.106615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 05/28/2025] Open
Abstract
Traditional Chinese medicine (TCM) has been extensively explored with various naturally derived compounds as a potential therapeutic agent for chronic atrophic gastritis (CAG). In addition to the aspects discussed in the reviewed article, this invited commentary explores the initial available evidence on a fungus from TCM, Hericium erinaceus, in the context of CAG. Initial clinical data suggest the potential of this fungus in inducing clinical and histological improvements in patients with CAG, as well as a marked antimicrobial activity against Helicobacter pylori infection. Preclinical cellular evidence also indicates an antineoplastic role in gastric carcinogenesis, mediated by two components: Erinacine A and S. Further evidence is needed to propose this fungus as a potential complementary therapeutic approach for CAG.
Collapse
Affiliation(s)
- Raffaele Pellegrino
- Division of Hepatogastroenterology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Campania, Italy
| | - Antonietta Gerarda Gravina
- Division of Hepatogastroenterology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Campania, Italy
| |
Collapse
|
3
|
Papantoniou K, Aggeletopoulou I, Pastras P, Triantos C. The Role of Somatostatin in the Gastrointestinal Tract. BIOLOGY 2025; 14:558. [PMID: 40427747 PMCID: PMC12109247 DOI: 10.3390/biology14050558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
The gastrointestinal (GI) tract is responsible for food digestion and host protection from harmful stimuli; however, its function as an endocrine organ is also well documented. Somatostatin (SST) was first discovered in the hypothalamus, but the GI tract is its main producer and target organ. SST is a potent inhibitor of many GI functions, including peristalsis, hormone secretion, and gastric acid production, while its anti-inflammatory effects contribute to the integrity of the intestinal barrier. These data make SST and its analogs useful agents in clinical practice. As our understanding of SST metabolism and function evolves, their use in a wide variety of medical conditions can improve patient care.
Collapse
Affiliation(s)
| | | | | | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University of Patras, 26504 Patras, Greece; (K.P.); (I.A.); (P.P.)
| |
Collapse
|
4
|
Wu S, Luo Y, Wei F, Li Y, Fan J, Chen Y, Zhang W, Li X, Xu Y, Chen Z, Xia C, Hu M, Li P, Gu Q. Lactic acid bacteria target NF-κB signaling to alleviate gastric inflammation. Food Funct 2025; 16:3101-3119. [PMID: 40152095 DOI: 10.1039/d4fo06308b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Helicobacter pylori (H. pylori) infection and the resulting gastric inflammation are major contributors to gastric cancer development. Probiotics, particularly Lactobacillus, are promising for their anti-inflammatory potential, yet their exact mechanisms in inhibiting H. pylori-induced inflammation are unclear. In our previous study, Lactiplantibacillus plantarum ZJ316 (L. plantarum ZJ316) demonstrated strong anti-inflammatory effects against H. pylori infection in vivo, but its precise mechanisms were not fully understood. Here, we aimed to investigate how L. plantarum ZJ316 inhibits the inflammatory response to H. pylori infection. Our results demonstrated that L. plantarum ZJ316 effectively reduced the expression of pro-inflammatory cytokines in H. pylori-infected AGS cells. Mechanistically, L. plantarum ZJ316 inhibited the NF-κB signaling pathway by preventing the degradation of IκBα, suppressing p65 phosphorylation, and blocking the nuclear translocation of phosphorylated p65. Treatment with the NF-κB inhibitor BAY 11-7082 further decreased tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8), and interleukin-1β (IL-1β) levels, confirming the inhibitory effect of L. plantarum ZJ316 on the NF-κB pathway. In H. pylori-infected mice, oral administration of L. plantarum ZJ316 significantly alleviated inflammatory cell infiltration, reduced TNF-α and pepsinogen II (PGII) levels, and increased interleukin-10 (IL-10) levels in serum. A comparative metagenomic analysis of the gastric microbiota revealed a decrease in Prevotella and Desulfovibrio, alongside an increase in Ligilactobacillus and Akkermansia, supporting the protective effects of L. plantarum ZJ316 and correlating with their decreased inflammatory response. In summary, administration of L. plantarum ZJ316 demonstrated robust anti-inflammatory effects against H. pylori infection by suppressing NF-κB signaling and promoting favorable changes in the gastric microbiota composition. Therefore, L. plantarum ZJ316 holds promise as a novel functional food for protecting the body against H. pylori infection.
Collapse
Affiliation(s)
- Shiying Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yuenuo Luo
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Fangtong Wei
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Jiayi Fan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yongqiang Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Wenjie Zhang
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Xuelong Li
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Yang Xu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ziqi Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Chenlan Xia
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Mingyang Hu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
5
|
Tao J, Zhang Z, Meng L, Zhang L, Wang J, Li Z. Risk prediction model for precancerous gastric lesions based on magnifying endoscopy combined with narrow-band imaging features. Front Oncol 2025; 15:1554523. [PMID: 40255428 PMCID: PMC12006015 DOI: 10.3389/fonc.2025.1554523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/13/2025] [Indexed: 04/22/2025] Open
Abstract
Background This study aimed to construct and validate diagnostic models for the Operative Link on Gastritis Assessment (OLGA) and Operative Link on Gastric Intestinal Metaplasia Assessment (OLGIM) staging systems using three different methodologies based on magnifying endoscopy with narrow-band imaging (ME-NBI) features, to evaluate model performance, and to analyse risk factors for high-risk OLGA/OLGIM stages. Methods We enrolled 356 patients who underwent white-light endoscopy and ME-NBI at the Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, between January 2022 and September 2023. Clinical data were recorded. Chi-square or Fisher's exact tests were used to analyse differences in endoscopic features between OLGA/OLGIM stages. Variables showing statistical significance underwent collinearity diagnosis before model inclusion. We constructed predictive models using Bayesian stepwise discrimination, random forest, and XGBoost algorithms. Receiver operating characteristic (ROC) curves were plotted using Python 3.12.4. Model accuracy, area under the ROC curve (AUC), sensitivity, and specificity were calculated for comprehensive validation. Results All three models demonstrated excellent diagnostic performance, with random forest and XGBoost models showing marginally superior accuracy, AUC values, and sensitivity compared with the Bayesian stepwise discrimination model. For OLGA staging, the AUC values were 0.928, 0.958, and 0.966, with accuracies of 0.854, 0.902, and 0.918 for Bayesian, random forest, and XGBoost models, respectively. For OLGIM staging, the corresponding AUC values were 0.924, 0.975, and 0.979, with accuracies of 0.910, 0.938, and 0.927. Risk factors for high-risk OLGA included lesion location (subcardial and lower body greater curvature), intestinal metaplasia patches, lesion size, demarcation line (DL), and margin regularity of micro-capillary demarcation line (MCDL). Risk factors for high-risk OLGIM included Helicobacter pylori infection status, mucosal condition, lesion location (lesser curvature and lower body greater curvature), erosion, lesion size, DL, vessel and epithelial classification (VEC), white globe appearance (WGA), and MCDL margin regularity. Conclusions All three models demonstrated robust accuracy and predictive capability, confirming that conventional white-light endoscopy combined with ME-NBI features provides valuable diagnostic reference for clinical risk assessment of precancerous gastric lesions.
Collapse
Affiliation(s)
- Jingna Tao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongmian Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Linghan Meng
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liju Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaqi Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Kolenda T, Białas P, Guglas K, Stasiak M, Kozłowska-Masłoń J, Tylkowska K, Zapłata A, Poter P, Janiczek-Polewska M, Mantaj P, Gieremek P, Kazimierczak U, Przybyła A, Regulska K, Stanisz B, Leporowska E, Mackiewicz A, Mackiewicz J, Kazmierska J, Cybulski Z, Teresiak A. lncRNA EGOT Is the Marker of HPV Infection and a Prognostic Factor for HNSCC Patients. Biomedicines 2025; 13:798. [PMID: 40299341 PMCID: PMC12025276 DOI: 10.3390/biomedicines13040798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/02/2025] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
Background: High-risk human papillomavirus (HPV) contributes to oropharyngeal cancers through mechanisms involving the deregulation of host cell functions by oncoproteins E6 and E7. Changes in the epigenome, particularly involving long non-coding RNAs (lncRNAs), are crucial for understanding HPV-related carcinogenesis. Methods: This study aimed to analyze the expression levels of lncRNAs in HPV-related head and neck squamous cell carcinoma (HNSCC) to determine their biological and clinical significance, addressing the current gap in clinically validated biomarkers for early screening and therapeutic interventions. Results: The study highlights the significant overexpression of the EGOT gene in HPV-positive HNSCC samples, suggesting its potential as a marker to distinguish between HPV-negative and HPV-positive cases. Furthermore, high EGOT expression correlates with better overall survival (OS) and indicates possible resistance to therapy, making it a valuable prognostic factor. Conclusions: These findings underscore the potential of incorporating EGOT expression analysis in clinical practice for improved patient stratification and treatment outcomes in HNSCC.
Collapse
Affiliation(s)
- Tomasz Kolenda
- Research and Implementation Unit, Greater Poland Cancer Centre, Garbary 15, 61-866 Poznan, Poland; (K.G.); (K.R.); (Z.C.); (A.T.)
- Microbiology Laboratory, Greater Poland Cancer Centre, Garbary Street 15, 61-866 Poznan, Poland;
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznan, Poland; (M.S.); (U.K.); (A.M.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland
| | - Piotr Białas
- Department of Cell Biology, Poznan University of Medical Sciences, 5D Rokietnicka, 60-806 Poznan, Poland
| | - Kacper Guglas
- Research and Implementation Unit, Greater Poland Cancer Centre, Garbary 15, 61-866 Poznan, Poland; (K.G.); (K.R.); (Z.C.); (A.T.)
- Microbiology Laboratory, Greater Poland Cancer Centre, Garbary Street 15, 61-866 Poznan, Poland;
| | - Maciej Stasiak
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznan, Poland; (M.S.); (U.K.); (A.M.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland
| | - Joanna Kozłowska-Masłoń
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland;
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, 61-614 Poznan, Poland
| | - Karina Tylkowska
- Microbiology Laboratory, Greater Poland Cancer Centre, Garbary Street 15, 61-866 Poznan, Poland;
- Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
| | - Anna Zapłata
- Department of Laboratory Diagnostics, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland; (A.Z.); (E.L.)
| | - Paulina Poter
- Department of Oncologic Pathology and Prophylaxis, Poznan University of Medical Sciences, Greater Poland Cancer Center, 15 Garbary Street, 61-866 Poznan, Poland;
| | - Marlena Janiczek-Polewska
- Department of Clinical Oncology, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland;
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Patrycja Mantaj
- Radiation Protection Department, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland;
| | - Paulina Gieremek
- Departament of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka Street 3, 60-806 Poznan, Poland; (P.G.); (B.S.)
- Pharmacy, Greater Poland Cancer Centre, Garbary 15, 61-866 Poznan, Poland
| | - Urszula Kazimierczak
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznan, Poland; (M.S.); (U.K.); (A.M.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland
| | - Anna Przybyła
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznan, Poland; (M.S.); (U.K.); (A.M.)
| | - Katarzyna Regulska
- Research and Implementation Unit, Greater Poland Cancer Centre, Garbary 15, 61-866 Poznan, Poland; (K.G.); (K.R.); (Z.C.); (A.T.)
- Pharmacy, Greater Poland Cancer Centre, Garbary 15, 61-866 Poznan, Poland
| | - Beata Stanisz
- Departament of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka Street 3, 60-806 Poznan, Poland; (P.G.); (B.S.)
| | - Ewa Leporowska
- Department of Laboratory Diagnostics, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland; (A.Z.); (E.L.)
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznan, Poland; (M.S.); (U.K.); (A.M.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznan University of Medical Sciences, 60-512 Poznan, Poland;
| | - Joanna Kazmierska
- Radiotherapy Department II, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland;
| | - Zefiryn Cybulski
- Research and Implementation Unit, Greater Poland Cancer Centre, Garbary 15, 61-866 Poznan, Poland; (K.G.); (K.R.); (Z.C.); (A.T.)
- Microbiology Laboratory, Greater Poland Cancer Centre, Garbary Street 15, 61-866 Poznan, Poland;
| | - Anna Teresiak
- Research and Implementation Unit, Greater Poland Cancer Centre, Garbary 15, 61-866 Poznan, Poland; (K.G.); (K.R.); (Z.C.); (A.T.)
- Laboratory of Cancer Genetics, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznan, Poland;
| |
Collapse
|
7
|
Sun HT. Helicobacter pylori-related serum indicators: Cutting-edge advances to enhance the efficacy of gastric cancer screening. World J Gastrointest Oncol 2025; 17:100739. [PMID: 40092953 PMCID: PMC11866254 DOI: 10.4251/wjgo.v17.i3.100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/08/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025] Open
Abstract
Helicobacter pylori (H. pylori) infection induces pathological changes via chronic inflammation and virulence factors, thereby increasing the risk of gastric cancer development. Compared with invasive examination methods, H. pylori-related serum indicators are cost-effective and valuable for the early detection of gastric cancer (GC); however, large-scale clinical validation and sufficient understanding of the specific molecular mechanisms involved are lacking. Therefore, a comprehensive review and analysis of recent advances in this field is necessary. In this review, we systematically analyze the relationship between H. pylori and GC and discuss the application of new molecular biomarkers in GC screening. We also summarize the screening potential and application of anti-H. pylori immunoglobulin G and virulence factor-related serum antibodies for identifying GC risk. These indicators provide early warning of infection and enhance screening accuracy. Additionally, we discuss the potential combination of multiple screening indicators for the comprehensive analysis and development of emerging testing methods to improve the accuracy and efficiency of GC screening. Although this review may lack sufficient evidence due to limitations in existing studies, including small sample sizes, regional variations, and inconsistent testing methods, it contributes to advancing personalized precision medicine in high-risk populations and developing GC screening strategies.
Collapse
Affiliation(s)
- Hao-Tian Sun
- Cancer Institute, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
8
|
Zhang Z, Chen S, Li S, Zheng Y, Mai L, Zhang X. Association of Helicobacter pylori related chronic atrophic gastritis and gastric cancer risk: a literature review. Front Med (Lausanne) 2025; 12:1504749. [PMID: 40051725 PMCID: PMC11882515 DOI: 10.3389/fmed.2025.1504749] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/06/2025] [Indexed: 03/09/2025] Open
Abstract
Chronic atrophic gastritis (CAG) is considered to be closely related to Helicobacter pylori (H. pylori) infection and characterized by the atrophy and/or intestinal metaplasia (IM) of the gastric mucosa in pathology. CAG is often regarded as the precancerous lesion of gastric cancer and H. pylori infection stimulates the development of atrophy and IM and the progression of gastric cancer through the persistent effect acting on the gastric mucosa, including releasing inflammatory factors such as Interleukin-8(IL-8). From the molecular biology perspective, growing evidence shows that H. pylori probably induce the expression of NF-κB, miR-204, miR-27a, hnRNPA2B1, and JARID1B, which play crucial roles in the progression of CAG into gastric cancer. In addition, H. pylori can increase Epstein-Barr virus (EBV) infection, and the co-infection will jointly increase gastric cancer risk. Furthermore, H. pylori induces cellular senescence and promotes atrophy progression and finally increases the gastric cancer risk. This review aims to explore the carcinogenic mechanisms of H. pylori related CAG in order to provide theoretical foundations for the pathogenesis mechanism and early detection and prevention of gastric cancer.
Collapse
Affiliation(s)
- Zefeng Zhang
- Department of Digestive Endoscopy Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Sitong Chen
- Southern Medical University, Guangzhou, Guangdong, China
| | - Shudan Li
- Southern Medical University, Guangzhou, Guangdong, China
| | - Yadan Zheng
- Southern Medical University, Guangzhou, Guangdong, China
| | - Lifei Mai
- Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoguang Zhang
- Department of Digestive Endoscopy Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Cheng X, Gu H, Chong Y, Li F, Bei S, Li H, Jiang J, Pan M, Feng L, Zhang X. Vitamin C Mediates IGFBP7 to Alleviate Chronic Atrophic Gastritis via the HIF-1α/VEGF Pathway. J Cell Mol Med 2025; 29:e70392. [PMID: 40012220 PMCID: PMC11865351 DOI: 10.1111/jcmm.70392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Chronic atrophic gastritis (CAG) is a precancerous lesion characterised by gastric mucosal atrophy and inflammation. Identifying key molecular mechanisms and potential therapeutic targets is essential to improve patient outcomes. Key modules and differentially expressed genes (DEGs) were recognised in the GSE153224 dataset using weighted gene co-expression network analysis (WGCNA) and examination of differential expression. IGFBP7 was identified as a hub gene by protein-protein interaction (PPI) network and expression validation. CAG patients' blood parameters and gastric mucosal health status were evaluated before and after the treatment of vitamin C (VC). In addition, we investigated the effects of VC and N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) on GES-1 cells, including cell viability, apoptosis and the expression of inflammatory and angiogenic markers. WGCNA identified that the blue module was significantly associated with CAG with a correlation coefficient 0.924. Among 93 overlapping genes, IGFBP7 was notably underexpressed and selected as a hub gene. ROC analysis confirmed the high diagnostic performance of IGFBP7. CAG patients treated with VC showed significant improvement in blood parameters and improved gastric mucosal health. In vitro, VC increased cell viability, reduced cytotoxicity and apoptosis and lowered COX-2 and apoptosis-related protein expression in MNNG-treated GES-1 cells. Knockdown of IGFBP7 further influenced these effects. MNNG upregulated HIF-1α/VEGF signalling proteins, which VC attenuated. Combined VC and IGFBP7 knockdown showed potential protective effects. This study highlights the regulatory role of VC and IGFBP7 in CAG and demonstrates their potential as therapeutic targets for improving gastric mucosal health and mitigating inflammation.
Collapse
Affiliation(s)
- Xun Cheng
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Hao Gu
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Yulin Chong
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Fan Li
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Songhua Bei
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Huanqing Li
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Jun Jiang
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Ming Pan
- Department of Traditional Chinese Medicine, Minhang HospitalFudan UniversityShanghaiChina
| | - Li Feng
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| | - Xiaohong Zhang
- Endoscopy Center, Minhang HospitalFudan UniversityShanghaiChina
| |
Collapse
|
10
|
Huang Y, Lu X, Wang Y, Zhang Y, Cen M. Increasing miR-1260b predicts the risk of gastric cancer in atrophic gastritis patients and regulates cell growth and metastasis of gastric cancer. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2025. [PMID: 39784713 DOI: 10.17235/reed.2024.10527/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Atrophic gastritis is of high risk of progressing to gastric cancer. Screening early gastric cancer and predicting the risk of atrophic gastritis developing into gastric cancer could improve the prognosis of gastric cancer. OBJECTIVE This study evaluated the significance of miR-1260b in early gastric cancer and in the progression of atrophic gastritis to gastric cancer aiming to explore a reliable biomarker. MATERIALS AND METHODS The study enrolled 78 early gastric cancer patients and 77 atrophic gastritis patients. The expression of miR-1260b was detected in serum and tissue samples by PCR. The risk of atrophic gastritis patients progressing to gastric cancer was assessed and correlated with miR-1260b levels. The potential of miR-1260b in distinguishing early gastric cancer was evaluated by ROC. In vitro, gastric cancer cells were infected with Helicobacter pylori, and the regulatory effect of miR-1260b on cell growth and metastasis was evaluated by CCK8 and Transwell assay. RESULTS Significant upregulation of miR-1260b was observed in early gastric cancer patients relative to atrophic gastritis patients, which distinguishes early gastric cancer patients and showed a positive correlation with the risk of atrophic gastritis patients developing gastric cancer. Early gastric cancer patients with positive H. pylori infection showed a higher miR-1260b level, and increasing miR-1260b was also observed in H. pylori-infected gastric cancer cells. H. pylori promoted cell growth and metastasis of gastric cancer while silencing miR-1260b could alleviate these effects. miR-1260b negatively regulated ZNF302, and the knockdown of ZNF302 reversed the protective effect of miR-1260 knockdown on gastric cancer cells. CONCLUSION Increasing miR-1260b can assist diagnose early gastric cancer and predict the risk of gastric cancer in atrophic gastritis patients. Silencing miR-1260b could alleviate the promotion of gastric cancer induced by H. pylori via negatively modulating ZNF302.
Collapse
Affiliation(s)
- Yesheng Huang
- Gastroenterology, The First Affiliated Hospital of Jinan University
| | - Xiayang Lu
- Gastroenterology, Taizhou Traditional Chinese Medicine Hospital
| | - Yu Wang
- Minimally Invasive Endoscopy Center, The Affiliated Hospital of Panzhihua University, China
| | - Yichuan Zhang
- Minimally Invasive Endoscopy Center, The Affiliated Hospital of Panzhihua University, China
| | - Meini Cen
- Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities
| |
Collapse
|
11
|
Yun H, Dong F, Wei X, Yan X, Zhang R, Zhang X, Wang Y. Role and value of the tumor microenvironment in the progression and treatment resistance of gastric cancer (Review). Oncol Rep 2025; 53:14. [PMID: 39611496 PMCID: PMC11622107 DOI: 10.3892/or.2024.8847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/30/2024] Open
Abstract
Gastric cancer (GC) is characterized by a complex and heterogeneous tumor microenvironment (TME) that significantly influences disease progression and treatment outcomes. The tumor stroma, which is composed of a variety of cell types such as cancer‑associated fibroblasts, immune cells and vascular components, displays significant spatial and temporal diversity. These stromal elements engage in dynamic crosstalk with cancer cells, shaping their proliferative, invasive and metastatic potential. Furthermore, the TME is instrumental in facilitating resistance to traditional chemotherapy, specific treatments and immunotherapy strategies. Understanding the underlying mechanisms by which the GC microenvironment evolves and supports tumor growth and therapeutic resistance is critical for developing effective treatment strategies. The present review explores the latest progress in understanding the intricate interactions between cancer cells and their immediate environment in GC, highlighting the implications for disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Heng Yun
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Fangde Dong
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiaoqin Wei
- Department of Pain, The Second People's Hospital of Baiyin, Baiyin, Gansu 730900, P.R. China
| | - Xinyong Yan
- Department of Proctology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Ronglong Zhang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Xiuyu Zhang
- Department of Gastroenterology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| | - Yulin Wang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, Gansu 730900, P.R. China
| |
Collapse
|
12
|
Luo Y, Xue H, Chen H, Gao Y, Ji G, Wu T. Metabolomics advances in chronic atrophic gastritis diagnosis and the integration of traditional Chinese medicine. Saudi Pharm J 2024; 32:102213. [DOI: 10.1016/j.jsps.2024.102213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025] Open
|
13
|
Andersen GT, Ianevski A, Resell M, Pojskic N, Rabben HL, Geithus S, Kodama Y, Hiroyuki T, Kainov D, Grønbech JE, Hayakawa Y, Wang TC, Zhao CM, Chen D. Multi-bioinformatics revealed potential biomarkers and repurposed drugs for gastric adenocarcinoma-related gastric intestinal metaplasia. NPJ Syst Biol Appl 2024; 10:127. [PMID: 39496635 PMCID: PMC11535201 DOI: 10.1038/s41540-024-00455-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/13/2024] [Indexed: 11/06/2024] Open
Abstract
Biomarkers associated with the progression from gastric intestinal metaplasia (GIM) to gastric adenocarcinoma (GA), i.e., GA-related GIM, could provide valuable insights into identifying patients with increased risk for GA. The aim of this study was to utilize multi-bioinformatics to reveal potential biomarkers for the GA-related GIM and predict potential drug repurposing for GA prevention in patients. The multi-bioinformatics included gene expression matrix (GEM) by microarray gene expression (MGE), ScType (a fully automated and ultra-fast cell-type identification based solely on a given scRNA-seq data), Ingenuity Pathway Analysis, PageRank centrality, GO and MSigDB enrichments, Cytoscape, Human Protein Atlas and molecular docking analysis in combination with immunohistochemistry. To identify GA-related GIM, paired surgical biopsies were collected from 16 GIM-GA patients who underwent gastrectomy, yielding 64 samples (4 biopsies per stomach x 16 patients) for MGE. Co-analysis was performed by including scRNAseq and immunohistochemistry datasets of endoscopic biopsies of 37 patients. The results of the present study showed potential biomarkers for GA-related GIM, including GEM of individual patients, individual genes (such as RBP2 and CD44), signaling pathways, network of molecules, and network of signaling pathways with key topological nodes. Accordingly, potential treatment targets with repurposed drugs were identified including epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase Src, paxillin, transcription factor Jun, breast cancer type 1 susceptibility protein, cellular tumor antigen p53, mouse double minute 2, and CD44.
Collapse
Affiliation(s)
- Gøran Troseth Andersen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
- Department of Surgery, Namsos Hospital, Namsos, Norway
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Mathilde Resell
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Naris Pojskic
- Laboratory for Bioinformatics and Biostatistics, University of Sarajevo - Institute for Genetic Engineering and Biotechnology, Sarajevo, Bosnia and Herzegovina
| | - Hanne-Line Rabben
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Synne Geithus
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Yosuke Kodama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tomita Hiroyuki
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jon Erik Grønbech
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
| | - Yoku Hayakawa
- Department of Gastroenterology, Tokyo University Hospital, Tokyo, Japan
| | - Timothy C Wang
- Department of Digestive and Liver Diseases and Herbert Iring Comprehensive Cancer Center, Columbia University Medical Center, New York, USA
| | - Chun-Mei Zhao
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Duan Chen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
14
|
Ou L, Hao Y, Liu H, Zhu Z, Li Q, Chen Q, Wei R, Feng Z, Zhang G, Yao M. Chebulinic acid isolated from aqueous extracts of Terminalia chebula Retz inhibits Helicobacter pylori infection by potential binding to Cag A protein and regulating adhesion. Front Microbiol 2024; 15:1416794. [PMID: 39421559 PMCID: PMC11483367 DOI: 10.3389/fmicb.2024.1416794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Terminalia chebula Retz, known as the King of Tibet, is considered a functional food in China, celebrated for its antioxidant, immune-modulating, antibacterial, and anti-inflammatory properties. Chebulinic acid, derived from aqueous extracts of Terminalia chebula Retz, is known for its anti-inflammatory properties. However, its potential as an anti-Helicobacter pylori (HP) agent has not been fully explored. METHODS Herein, we extracted the main compound from Terminalia chebula Retz using a semi-preparative liquid chromatography (LC) system and identified compound 5 as chebulinic acid through Ultra-high performance liquid chromatography-MS/MS (UPLC-MS/MS) and Nuclear Magnetic Resonance (NMR). To evaluate its role, we conducted minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) assays, scanning electron microscope (SEM) imaging, inhibiting kinetics curves, urea fast test, cell counting kit-8 (CCK-8) assay, western blot analysis, griess reagent system, and molecular docking. RESULTS Our results showed that chebulinic acid effectively inhibited the growth of the HP strain ATCC 700392, damaged the HP structure, and exhibited selective antimicrobial activity without affecting normal epithelial cells GES-1. Importantly, it suppressed the expression of Cytotoxin-associated gene A (Cag A) protein, a crucial factor in HP infection. Molecular docking analysis predicted a strong affinity (-9.7 kcal/mol) between chebulinic acid and Cag A protein. CONCLUSION Overall, our findings suggest that chebulinic acid acts as an anti-adhesive agent, disrupting the adhesion of HP to host cells, which is a critical step in HP infection. It also suppresses the Cag A protein. These results highlight the potential of chebulinic acid against HP infections.
Collapse
Affiliation(s)
- Ling Ou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| | - Yajie Hao
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
| | - Hengrui Liu
- Cancer Institute, Jinan University, Guangzhou, China
- Yinuo Biomedical Company, Tianjin, China
| | - Zhixiang Zhu
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qingwei Li
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
| | - Qingchang Chen
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Ruixia Wei
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Zhong Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
- International Pharmaceutical Engineering Lab of Shandong Province, Feixian, China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Guimin Zhang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Meicun Yao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
15
|
Xia RJ, Du XY, Shen LW, Ma JG, Xu SM, Fan RF, Qin JW, Yan L. Roles of the tumor microenvironment in the resistance to programmed cell death protein 1 inhibitors in patients with gastric cancer. World J Gastrointest Oncol 2024; 16:3820-3831. [PMID: 39350980 PMCID: PMC11438768 DOI: 10.4251/wjgo.v16.i9.3820] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/27/2024] [Accepted: 08/09/2024] [Indexed: 09/09/2024] Open
Abstract
Despite the continuous developments and advancements in the treatment of gastric cancer (GC), which is one of the most prevalent types of cancer in China, the overall survival is still poor for most patients with advanced GC. In recent years, with the progress in tumor immunology research, attention has shifted toward immunotherapy as a therapeutic approach for GC. Programmed cell death protein 1 (PD-1) inhibitors, as novel immunosuppressive medications, have been widely utilized in the treatment of GC. However, many patients are still resistant to PD-1 inhibitors and experience recurrence in the advanced stages of PD-1 immunotherapy. To reduce the occurrence of drug resistance and recurrence in GC patients receiving PD-1 immunotherapy, to maximize the clinical activity of immunosuppressive drugs, and to elicit a lasting immune response, it is essential to research the tumor microenvironment mechanisms leading to PD-1 inhibitor resistance in GC patients. This article reviews the progress in studying the factors influencing the resistance to PD-1 inhibitors in the GC tumor microenvironment, aiming to provide insights and a basis for reducing resistance to PD-1 inhibitors for GC patients in the future.
Collapse
Affiliation(s)
- Ren-Jie Xia
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Xiao-Yu Du
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Li-Wen Shen
- Department of Medical Support Center, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Guo Ma
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Shu-Mei Xu
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Rui-Fang Fan
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Wei Qin
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Long Yan
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| |
Collapse
|
16
|
Qian ST, Zhao HY, Xie FF, Liu QS, Cai DL. Streptococcus anginosus in the development and treatment of precancerous lesions of gastric cancer. World J Gastrointest Oncol 2024; 16:3771-3780. [PMID: 39350992 PMCID: PMC11438778 DOI: 10.4251/wjgo.v16.i9.3771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/22/2024] [Accepted: 06/06/2024] [Indexed: 09/09/2024] Open
Abstract
The microbiota is strongly association with cancer. Studies have shown significant differences in the gastric microbiota between patients with gastric cancer (GC) patients and noncancer patients, suggesting that the microbiota may play a role in the development of GC. Although Helicobacter pylori (H. pylori) infection is widely recognized as a primary risk factor for GC, recent studies based on microbiota sequencing technology have revealed that non-H. pylori microbes also have a significant impact on GC. A recent study discovered that Streptococcus anginosus (S. anginosus) is more prevalent in the gastric mucosa of patients with GC than in that of those without GC. S. anginosus infection can spontaneously induce chronic gastritis, mural cell atrophy, mucoid chemotaxis, and heterotrophic hyperplasia, which promote the development of precancerous lesions of GC (PLGC). S. anginosus also disrupts the gastric barrier function, promotes the proliferation of GC cells, and inhibits apoptosis. However, S. anginosus is underrepresented in the literature. Recent reports suggest that it may cause precancerous lesions, indicating its emerging pathogenicity. Modern novel molecular diagnostic techniques, such as polymerase chain reaction, genetic testing, and Ultrasensitive Chromosomal Aneuploidy Detection, can be used to gastric precancerous lesions via microbial markers. Therefore, we present a concise summary of the relationship between S. anginosus and PLGC. Our aim was to further investigate new methods of preventing and treating PLGC by exploring the pathogenicity of S. anginosus on PLGC.
Collapse
Affiliation(s)
- Su-Ting Qian
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Hao-Yu Zhao
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Fei-Fei Xie
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Qing-Sheng Liu
- Science and Education Section, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Dan-Li Cai
- Intensive Care Unit, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 311122, Zhejiang Province, China
| |
Collapse
|
17
|
Liu L, Wang X, He Q, Yu B, Wang J, Shen H. Differential diagnosis of gastric low- and high grade dysplasia using C6orf15 protein. Ann Diagn Pathol 2024; 71:152298. [PMID: 38547762 DOI: 10.1016/j.anndiagpath.2024.152298] [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: 02/07/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 06/09/2024]
Abstract
OBJECTIVE To investigate the expression of C6orf15 protein in gastric endoscopic biopsy specimens and its usage as an ancillary diagnostic biomarker in determining the grade of gastric dysplasia. METHODS We selected 102 patients with gastric endoscopic biopsy specimens from Jinling Hospital. These were divided into four groups: 22 cases of gastric mucosal benign lesions, 28 with low-grade dysplasia (LGD, intestinal-type: 21 cases,foveolar-type: 7cases), 28 with high-grade dysplasia (HGD, intestinal-type: 20 cases,foveolar-type: 8 cases), and 24 cases of gastric adenocarcinoma. We examined the expressions of C6orf15, P53, and Ki67 in 102 gastric endoscopic biopsy specimens, including 47 cases with accompanying endoscopic submucosal dissection (ESD) specimens, using immunohistochemistry. RESULTS In gastric HGD and gastric adenocarcinoma, the c6orf15 protein exhibits diffuse and strong cytoplasmic expression in tumor cells. Conversely, in gastric LGD and benign gastric mucosal lesions, the c6orf15 protein shows negative or faint yellow cytoplasmic staining. The expression rate of C6orf15 in high-grade gastric dysplasia (HGD, 93 %) and gastric adenocarcinoma (100 %) was significantly higher than in the gastric mucosal benign lesion group (0 %) and the low-grade dysplasia (LGD, 7 %) group (P < 0.001). CONCLUSION The detection of C6orf15 protein expression could serve as a valuable adjunctive diagnostic tool for distinguishing between gastric HGD, LGD, and benign lesions. The combined assessment of C6orf15, P53, and Ki67 expressions may be beneficial in determining the grade of gastric dysplasia and evaluating the risk of progression in gastric mucosal lesions in clinical practice.
Collapse
Affiliation(s)
- Leilei Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing 210029, China; Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Xuan Wang
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Qibin He
- Department of Gastroenterology, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211199, China
| | - Bo Yu
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Jiandong Wang
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China.
| | - Hong Shen
- Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing 210029, China.
| |
Collapse
|
18
|
Lin DY, Chen XM, Jiang X. Effect and mechanism of Proanthocyanidins on Helicobacter pylori-induced gastric mucosal injury in mice. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:368-375. [DOI: 10.11569/wcjd.v32.i5.368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
|
19
|
Wang YK, Li YY, Wang B, Ran DM, Zhu CY, Li P, Jiang B, Wang SN. Histopathological staging of atrophic lesions of gastric mucosa. Heliyon 2024; 10:e27845. [PMID: 38560685 PMCID: PMC10979135 DOI: 10.1016/j.heliyon.2024.e27845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Objective To study the histopathological staging of atrophic lesions of the gastric mucosa. Methods Histology and immunohistochemistry were used to closely examine 2144 specimens of atrophic gastric mucosa that were taken from endoscopic biopsies. Results When the gastric mucosa epithelium is affected by infection, chemical stimulation, immune factors, genetic factors, and other factors, it may cause an atrophy of gastric mucosa epithelium and a decrease in the number of glands, intestinal metaplasia, hyperplasia of smooth muscle fibers, and atrophy of stem cells in the proliferative zone. In this study, we characterized the above lesions as atrophic lesions of the gastric mucosa. Based on the morphological and histological characteristics of the lesion, as well as the law of cell proliferation and transformation during its occurrence and development, we propose five stages. We also noted the onset age, gender correlation, and histopathological characteristics of each stage of gastric mucosal atrophies. Conclusion Understanding the pathological staging of gastric mucosal atrophy is essential for treating patients correctly and keeping track of changes in malignant cells. It is also very important in preventing the initiation of gastric cancer or from getting worse.
Collapse
Affiliation(s)
- Yang-kun Wang
- The Fourth People"s Hospital of Longgang District, Shenzhen, 518123, China
| | - Ying-ying Li
- Shenzhen Polytechnic University, Shenzhen, 518055, China
| | - Bin Wang
- Department of Radiation Therapy, Cancer Center, Shanghai Jiahui International Hospital, Shanghai, 200000, China
| | - Dong-mei Ran
- Department of Pathology, Southern University of Science and Technology Hospital, Shenzhen, 518055, China
| | - Chao-ya Zhu
- Third Affiliated Hospital of Zhengzhou University, Shenzhen, 450052, China
| | - Ping Li
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Bo Jiang
- Department of Pathology, No. 990 Hospital of the PLA Joint Logistics Support Force, Zhumadian, 463000, China
| | - Su-nan Wang
- Shenzhen Polytechnic University, Shenzhen, 518055, China
| |
Collapse
|