1
|
Wu Y, Zhang K, Zheng Y, Jin H. A review of potential mechanisms and treatments of gastric intestinal metaplasia. Eur J Gastroenterol Hepatol 2025; 37:383-394. [PMID: 39975991 DOI: 10.1097/meg.0000000000002903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Gastric intestinal metaplasia (GIM) is a pathological process where gastric mucosal epithelial cells are replaced by intestinal-type cells, serving as a precursor lesion for gastric cancer. This transformation involves various genetic and environmental factors, affecting key genes and signaling pathways. Recent research has revealed complex mechanisms, including changes in gene expression, abnormal signaling pathway activation, and altered cell behavior. This review summarizes the latest research on GIM, discussing its pathogenesis, current treatment strategies, and potential efficacy of emerging approaches like gene editing, microbiome interventions, and integrative medicine. By exploring these strategies, we aim to provide more effective treatments for GIM and reduce gastric cancer incidence. The review also highlights the importance of interdisciplinary studies in understanding GIM mechanisms and improving treatment strategies.
Collapse
Affiliation(s)
- Yueyao Wu
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | | | | | | |
Collapse
|
2
|
Zhang M, Zhong J, Shen Y, Song Z. Crosstalk between bile acids and gut microbiota: a potential target for precancerous lesions of gastric cancer. Front Pharmacol 2025; 16:1533141. [PMID: 40183085 PMCID: PMC11965922 DOI: 10.3389/fphar.2025.1533141] [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: 11/23/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
As a critical juncture in the pathological continuum from gastritis to gastric cancer, precancerous lesions of gastric cancer (PLGC) are increasingly prevalent, significantly undermining the health of the global population. The primary constituents of bile, specifically bile acids (BAs), disrupt the equilibrium of gastric hormone secretion and compromise the structural integrity of the gastric mucosa, thereby facilitating gastric oncogenesis. Moreover, gut microbiota modulate host physiological and pathological processes through immune response regulation, metabolic pathway interference, and direct interaction with gastric tumor cells. Extensive research has elucidated that the metabolic dysregulation of BAs and gut microbiota, in concert with the resultant impairment of the gastric mucosa, are central to the pathogenesis of PLGC. In anticipation of future clinical preventive and therapeutic strategies, this review collates recent insights into the roles of BAs and gut bacteria in PLGC, examining their interplay and significance in the pathogenic mechanism of PLGC.
Collapse
Affiliation(s)
- Maofu Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jialin Zhong
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yanyun Shen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Zhongyang Song
- Department of Oncology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
3
|
Nai S, Song J, Su W, Liu X. Bidirectional Interplay Among Non-Coding RNAs, the Microbiome, and the Host During Development and Diseases. Genes (Basel) 2025; 16:208. [PMID: 40004537 PMCID: PMC11855195 DOI: 10.3390/genes16020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
It is widely known that the dysregulation of non-coding RNAs (ncRNAs) and dysbiosis of the gut microbiome play significant roles in host development and the progression of various diseases. Emerging evidence has highlighted the bidirectional interplay between ncRNAs and the gut microbiome. This article aims to review the current understanding of the molecular mechanisms underlying the crosstalk between ncRNAs, especially microRNA (miRNA), and the gut microbiome in the context of development and diseases, such as colorectal cancer, inflammatory bowel diseases, neurological disorders, obesity, and cardiovascular disease. Ultimately, this review seeks to provide a foundation for exploring the potential roles of ncRNAs and gut microbiome interactions as biomarkers and therapeutic targets for clinical diagnosis and treatment, such as ncRNA mimics, antisense oligonucleotides, and small-molecule compounds, as well as probiotics, prebiotics, and diets.
Collapse
Affiliation(s)
| | | | | | - Xiaoqian Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (S.N.); (J.S.); (W.S.)
| |
Collapse
|
4
|
Zheng S, Wang Y, Ni C, Guo R, Qiu X, Chen J, Wang L, Sun X, Chen M, Liu Y, Yuan Y, Gong Y. Helicobacter pylori SlyD stabilizes TPT1 via hnRNPK and enhances OCT1-mediated CDX2 transcriptional activation to drive gastric intestinal metaplasia. BMC Med 2025; 23:71. [PMID: 39915880 PMCID: PMC11803974 DOI: 10.1186/s12916-025-03911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Gastric intestinal metaplasia (GIM) represents an important precancerous lesion in intestinal-type gastric cancer, triggered by persistent Helicobacter pylori (H. pylori) infection. In a previous study, we unveiled SlyD as a novel virulence factor of H. pylori, establishing its role in GIM induction through TPT1. However, the underlying mechanism remains undetermined. METHODS Gastric epithelial cells were stimulated with H. pylori 26695, a SlyD inactivated mutant (ΔSlyD), and purified HpSlyD protein, respectively. Real-time qPCR and western blot were subsequently used to assess the expression levels of hnRNPK, TPT1, OCT1, and GIM markers. RNA sequencing was employed to identify differentially expressed genes associated with H. pylori SlyD infection. Protein stability was evaluated using cycloheximide. Molecular interactions were investigated through co-immunoprecipitation, chromatin immunoprecipitation, and dual-luciferase reporter assays. Additionally, molecular docking was utilized to predict TPT1 inhibitors. Immunohistochemistry staining was conducted to validate hnRNPK, TPT1, OCT1, and CDX2 expression in gastric tissue samples from both human and Mongolian gerbils. RESULTS H. pylori SlyD upregulates TPT1 and induces the expression of GIM markers through hnRNPK. The interaction between hnRNPK and TPT1 enhances TPT1 protein stability, with H. pylori SlyD intensifying this association. TPT1 promotes the expression of GIM markers mediated via OCT1, which binds to CDX2 promoter region, thereby modulating its transcriptional activity. Dihydroartemisinin has the potential to target TPT1, inhibiting the H. pylori SlyD-induced expression of GIM markers. CONCLUSIONS In vitro and in vivo experiments verified that H. pylori SlyD enhances TPT1 stability through hnRNPK, leading to OCT1-mediated transcriptional activation of CDX2 and the initiation of the GIM process. Our study offers novel perspectives on the pathogenesis of H. pylori-related gastric precancerous conditions.
Collapse
Affiliation(s)
- Shuwen Zheng
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Yingying Wang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Chuxuan Ni
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Rui Guo
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Xunan Qiu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Jijun Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Lu Wang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaohu Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Moye Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Yunen Liu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China.
| | - Yuehua Gong
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, the First Hospital of China Medical University, Shenyang, 110001, China.
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
5
|
He Y, Wang J, Deng Z, Feng H, Du M, Zhang D, Zhang G, Shi T, Chen W. FOLR2 + macrophage depletion from intestinal metaplasia to early gastric cancer: single-cell sequencing insight into gastric cancer progression. J Exp Clin Cancer Res 2024; 43:326. [PMID: 39702278 DOI: 10.1186/s13046-024-03245-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND The immune landscape associated with different subtypes of intestinal metaplasia (IM) and early gastric cancer (EGC) remains unclear. This study aimed to investigate the immune landscape of complete intestinal metaplasia (CIM), incomplete intestinal metaplasia (IIM), and EGC, as well as the underlying mechanisms of EGC progression. METHODS Gastric biopsy samples were collected from five patients with CIM, six patients with IIM, and four patients with EGC, followed by single-cell RNA sequencing. Multiplex immunohistochemical staining was employed to validate the samples from the aforementioned patients. To elucidate the potential mechanisms involved, in vitro coculture experiments were conducted using FOLR2+/FOLR2- macrophages and CD8+ T cells. Flow cytometry was utilized to investigate the biological functions of FOLR2+ macrophages in the progression of EGC. RESULTS Five subpopulations of macrophages were identified in CIM, IIM and EGC samples. FOLR2+ macrophages possess antitumor immune potential, and the proportion of FOLR2+ macrophage gradually decreased from the CIM stage to the IIM and EGC stages. FOLR2+ macrophages were significantly positively correlated with CD8+ T cells and activated the cytotoxicity of CD8+ T cells via antigen cross-presentation. Additionally, during the progression of EGC, epithelial cells progressively upregulated APP expression, thus inducing necroptosis of FOLR2+ macrophages via the APP‒TNFRSF21 axis. CONCLUSIONS Our work provides an understanding of the potential mechanisms underlying the malignant transformation of IM mediated by FOLR2+ macrophages.
Collapse
Affiliation(s)
- Yuxin He
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Zilin Deng
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Huang Feng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Mingzhan Du
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Deqing Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| |
Collapse
|
6
|
Xu Y, Wu Y, Liu J, Xiao L, Zhang Y. Caudal-Type Homeobox Transcription Factor 2 is a Favorable Prognostic Indicator in Stage II and III Gastric Cancer Following Curative Surgery. Int J Gen Med 2024; 17:4377-4394. [PMID: 39355342 PMCID: PMC11444083 DOI: 10.2147/ijgm.s471727] [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: 04/01/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024] Open
Abstract
Background The study explored the prognostic value of caudal-type homeobox transcription factor 2 (CDX2) in stage II and III gastric cancer. Methods This study evaluated the expression level of CDX2 in gastric cancer in a hospital cohort (n=197) using immunohistochemistry. According to a semiquantitative score used to determine CDX2 expression, the cases were divided into a low CDX2 group (116 cases) and a high CDX2 group (81 cases). The RNA-seq expression data from 291 patients with stage II and III gastric cancer from The Cancer Genome Atlas (TCGA) cohort were used to verify the immunohistochemistry results. Based on the median CDX2 expression value, the TCGA patients were divided into a low CDX2 group (145 cases) and a high CDX2 group (146 cases). The relationships among CDX2 expression and clinicopathological features were determined using the Chi-square test. Cox proportional hazards regression models were applied to estimate the independent prognostic factors. The probability of survival was determined using the Kaplan-Meier method and Log rank tests. Results Based on the Cox multivariate analysis, CDX2 was the independent prognostic factor in the hospital and TCGA cohorts. In the hospital cohort, CDX2 expression was associated with an improved DFS (hazard ratio [HR] = 0.4076, 95% CI, 0.2675-0.6210, P = 0.0001) and OS (HR = 0.4183, 95% CI, 0.2744-0.6375, P = 0.0002). In the TCGA cohort, CDX2 expression also was associated with an improved DFS (HR = 0.5948, 95% CI, 0.4153-0.8521, P = 0.0054) and OS (HR = 0.5976, 95% CI, 0.4172-0.8561, P = 0.0058). Furthermore, the CDX2 expression level was correlated with an improved DFS (P = 0.0025) and OS (P = 0.0015) using the Kaplan-Meier Plotter database for gastric cancer. Conclusion CDX2 is a potential prognostic biomarker for stage II and III gastric cancer. In addition, CDX2 positive cancer patients are more likely to have resectable tumors and exhibit better survival rates.
Collapse
Affiliation(s)
- Yang Xu
- Department of Gastroenterology, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163000, People's Republic of China
| | - Yue Wu
- Department of Gastroenterology, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163000, People's Republic of China
| | - Jianhong Liu
- Department of Breast and Thyroid Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163000, People's Republic of China
| | - Lili Xiao
- Department of Gastroenterology, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163000, People's Republic of China
| | - Ying Zhang
- Department of Gastroenterology, Daqing Oilfield General Hospital, Daqing, Heilongjiang, 163000, People's Republic of China
| |
Collapse
|
7
|
Zhang M, Zhong J, Song Z, Xu Q, Chen Y, Zhang Z. Regulatory mechanisms and potential therapeutic targets in precancerous lesions of gastric cancer: A comprehensive review. Biomed Pharmacother 2024; 177:117068. [PMID: 39018877 DOI: 10.1016/j.biopha.2024.117068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/19/2024] Open
Abstract
Precancerous lesions of gastric cancer (PLGC) represent a critical pathological stage in the transformation from normal gastric mucosa to gastric cancer (GC). The global incidence of PLGC has been rising over the past few decades, with a trend towards younger onset ages. Increasing evidence suggests that early prevention and treatment of PLGC can effectively reverse the malignant development of gastric mucosal epithelial cells. However, there is currently a lack of effective therapeutic drugs and methods. Recent years have witnessed substantial advancements in PLGC research, with the elucidation of novel regulatory mechanisms offering promising avenues for clinical intervention and drug development. This review aims to delineate potential targets for early prevention and diagnosis of GC while exploring innovative approaches to PLGC management. This article focuses on elucidating the regulatory mechanisms of the inflammatory microenvironment, bile acids (BA), glycolysis, autophagy, apoptosis, ferroptosis, and cellular senescence. We pay particular attention to potential therapeutic targets for PLGC, with the goal of providing insights and theoretical basis for clinical research on PLGC.
Collapse
Affiliation(s)
- Maofu Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jialin Zhong
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhongyang Song
- Department of Oncology, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730020, China
| | - Qian Xu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yuchan Chen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730050, China.
| |
Collapse
|
8
|
Zhou DS, Zhang WJ, Song SY, Hong XX, Yang WQ, Li JJ, Xu JQ, Kang JY, Cai TT, Xu YF, Guo SJ, Pan HF, Li HW. Weiwei Decoction alleviates gastric intestinal metaplasia through the olfactomedin 4/nucleotide-binding oligomerization domain 1/caudal-type homeobox gene 2 signaling pathway. World J Gastrointest Oncol 2024; 16:3211-3229. [PMID: 39072182 PMCID: PMC11271767 DOI: 10.4251/wjgo.v16.i7.3211] [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: 02/15/2024] [Revised: 04/25/2024] [Accepted: 05/11/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Gastric intestinal metaplasia (IM) is a precancerous lesion that is associated with an elevated risk of gastric carcinogenesis. Weiwei Decoction (WWD) is a promising traditional Chinese herbal formula widely employed in clinical for treating IM. Previous studies suggested the potential involvement of the olfactomedin 4 (OLFM4)/nucleotide-binding oligomerization domain 1 (NOD1)/caudal-type homeobox gene 2 (CDX2) signaling pathway in IM regulation. AIM To verify the regulation of the OLFM4/NOD1/CDX2 pathway in IM, specifically investigating WWD's effectiveness on IM through this pathway. METHODS Immunohistochemistry for OLFM4, NOD1, and CDX2 was conducted on tissue microarray. GES-1 cells treated with chenodeoxycholic acid were utilized as IM cell models. OLFM4 short hairpin RNA (shRNA), NOD1 shRNA, and OLFM4 pcDNA were transfected to clarify the pathway regulatory relationships. Protein interactions were validated by co-immunoprecipitation. To explore WWD's pharmacological actions, IM rat models were induced using N-methyl-N'-nitro-N-nitrosoguanidine followed by WWD gavage. Gastric cells were treated with WWD-medicated serum. Cytokines and chemokines content were assessed by enzyme-linked immunosorbent assay and quantitative reverse transcription polymerase chain reaction. RESULTS The OLFM4/NOD1/CDX2 axis was a characteristic of IM. OLFM4 exhibited direct binding and subsequent down-regulation of NOD1, thereby sustaining the activation of CDX2 and promoting the progression of IM. WWD improved gastric mucosal histological lesions while suppressing intestinal markers KLF transcription factor 4, villin 1, and MUCIN 2 expression in IM rats. Regarding pharmacological actions, WWD suppressed OLFM4 and restored NOD1 expression, consequently reducing CDX2 at the mRNA and protein levels in IM rats. Parallel regulatory mechanisms were observed at the protein level in IM cells treated with WWD-medicated serum. Furthermore, WWD-medicated serum treatment strengthened OLFM4 and NOD1 interaction. In case of anti-inflammatory, WWD restrained interleukin (IL)-6, interferon-gamma, IL-17, macrophage chemoattractant protein-1, macrophage inflammatory protein 1 alpha content in IM rat serum. WWD-medicated serum inhibited tumor necrosis factor alpha, IL-6, IL-8 transcriptions in IM cells. CONCLUSION The OLFM4/NOD1/CDX2 pathway is involved in the regulation of IM. WWD exerts its therapeutic efficacy on IM through the pathway, additionally attenuating the inflammatory response.
Collapse
Affiliation(s)
- Di-Shu Zhou
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Wei-Jian Zhang
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Shu-Ya Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Xin-Xin Hong
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Wei-Qin Yang
- Department of Chinese Medicine, The Eight Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, Guangdong Province, China
| | - Juan-Juan Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Jian-Qu Xu
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Jian-Yuan Kang
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Tian-Tian Cai
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Yi-Fei Xu
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Shao-Ju Guo
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Hua-Feng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong Province, China
| | - Hai-Wen Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
9
|
Jia J, Zhao H, Li F, Zheng Q, Wang G, Li D, Liu Y. Research on drug treatment and the novel signaling pathway of chronic atrophic gastritis. Biomed Pharmacother 2024; 176:116912. [PMID: 38850667 DOI: 10.1016/j.biopha.2024.116912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a global digestive system disease and one of the important causes of gastric cancer. The incidence of CAG has been increasing yearly worldwide. PURPOSE This article reviews the latest research on the common causes and future therapeutic targets of CAG as well as the pharmacological effects of corresponding clinical drugs. We provide a detailed theoretical basis for further research on possible methods for the treatment of CAG and reversal of the CAG process. RESULTS CAG often develops from chronic gastritis, and its main pathological manifestation is atrophy of the gastric mucosa, which can develop into gastric cancer. The drug treatment of CAG can be divided into agents that regulate gastric acid secretion, eradicate Helicobacter. pylori (H. pylori), protect gastric mucous membrane, or inhibit inflammatory factors according to their mechanism of action. Although there are limited specific drugs for the treatment of CAG, progress is being made in defining the pathogenesis and therapeutic targets of the disease. Growing evidence shows that NF-κB, PI3K/AKT, Wnt/ β-catenin, MAPK, Toll-like receptors (TLRs), Hedgehog, and VEGF signaling pathways play an important role in the development of CAG.
Collapse
Affiliation(s)
- Jinhao Jia
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Huijie Zhao
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Special Administrative Region of China
| | - Qiusheng Zheng
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832003, PR China
| | - Guoli Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, Xinjiang 832003, PR China.
| | - Ying Liu
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine & Binzhou Hospital of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong 264003, PR China.
| |
Collapse
|
10
|
Drnovsek J, Homan M, Zidar N, Smid LM. Pathogenesis and potential reversibility of intestinal metaplasia - a milestone in gastric carcinogenesis. Radiol Oncol 2024; 58:186-195. [PMID: 38643513 PMCID: PMC11165985 DOI: 10.2478/raon-2024-0028] [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: 11/03/2023] [Accepted: 03/19/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Non-cardia gastric cancer remains a major cause of cancer-related mortality worldwide, despite declining incidence rates in many industrialized countries. The development of intestinal-type gastric cancer occurs through a multistep process in which normal mucosa is sequentially transformed into hyperproliferative epithelium, followed by metaplastic processes leading to carcinogenesis. Chronic infection with Helicobacter pylori is the primary etiological agent that causes chronic inflammation of the gastric mucosa, induces atrophic gastritis, and can lead to intestinal metaplasia and dysplasia. Both intestinal metaplasia and dysplasia are precancerous lesions, in which gastric cancer is more likely to occur. Atrophic gastritis often improves after eradication of Helicobacter pylori; however, the occurrence of intestinal metaplasia has been traditionally regarded as "the point of no return" in the carcinogenesis sequence. Helicobacter pylori eradication heals non-atrophic chronic gastritis, may lead to regression of atrophic gastritis, and reduces the risk of gastric cancer in patients with these conditions. In this article, we discuss the pathogenesis, epigenomics, and reversibility of intestinal metaplasia and briefly touch upon potential treatment strategy. CONCLUSIONS Gastric intestinal metaplasia no longer appears to be an irreversible precancerous lesion. However, there are still many controversies regarding the improvement of intestinal metaplasia after Helicobacter pylori eradication.
Collapse
Affiliation(s)
- Jan Drnovsek
- Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matjaz Homan
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Gastroenterology, Hepatology and Nutrition, University Children’s Hospital, Ljubljana, Slovenia
| | - Nina Zidar
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Lojze M Smid
- Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
11
|
Yang X, Ye T, Rong L, Peng H, Tong J, Xiao X, Wan X, Guo J. GATA4 Forms a Positive Feedback Loop with CDX2 to Transactivate MUC2 in Bile Acids-Induced Gastric Intestinal Metaplasia. Gut Liver 2024; 18:414-425. [PMID: 36860162 PMCID: PMC11096910 DOI: 10.5009/gnl220394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 03/03/2023] Open
Abstract
Background/Aims Gastric intestinal metaplasia (GIM), a common precancerous lesion of gastric cancer, can be caused by bile acid reflux. GATA binding protein 4 (GATA4) is an intestinal transcription factor involved in the progression of gastric cancer. However, the expression and regulation of GATA4 in GIM has not been clarified. Methods The expression of GATA4 in bile acid-induced cell models and human specimens was examined. The transcriptional regulation of GATA4 was investigated by chromatin immunoprecipitation and luciferase reporter gene analysis. An animal model of duodenogastric reflux was used to confirm the regulation of GATA4 and its target genes by bile acids. Results GATA4 expression was elevated in bile acid-induced GIM and human specimens. GATA4 bound to the promoter of mucin 2 (MUC2) and stimulate its transcription. GATA4 and MUC2 expression was positively correlated in GIM tissues. Nuclear transcription factor-κB activation was required for the upregulation of GATA4 and MUC2 in bile acid-induced GIM cell models. GATA4 and caudal-related homeobox 2 (CDX2) reciprocally transactivated each other to drive the transcription of MUC2. In chenodeoxycholic acid-treated mice, MUC2, CDX2, GATA4, p50, and p65 expression levels were increased in the gastric mucosa. Conclusions GATA4 is upregulated and can form a positive feedback loop with CDX2 to transactivate MUC2 in GIM. NF-κB signaling is involved in the upregulation of GATA4 by chenodeoxycholic acid.
Collapse
Affiliation(s)
- Xiaofang Yang
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Ting Ye
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Li Rong
- Department of Gastroenterology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Peng
- Department of Gastroenterology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Tong
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Xiao Xiao
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Xiaoqiang Wan
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Jinjun Guo
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
- Department of Gastroenterology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Li K, Ma X, Li Z, Liu Y, Shen G, Luo Z, Wang D, Xia L, Wang Z, Tian M, Liu H, Geng F, Li B. A Natural Peptide from A Traditional Chinese Medicine Has the Potential to Treat Chronic Atrophic Gastritis by Activating Gastric Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304326. [PMID: 38544338 PMCID: PMC11132046 DOI: 10.1002/advs.202304326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 02/08/2024] [Indexed: 05/29/2024]
Abstract
Chronic atrophic gastritis (AG) is initiated mainly by Helicobacter pylori infection, which may progress to stomach cancer following the Correa's cascade. The current treatment regimen is H. pylori eradication, yet evidence is lacking that this treatment is effective on later stages of AG especially gastric gland atrophy. Here, using AG mouse model, patient samples, gastric organoids, and lineage tracing, this study unraveled gastric stem cell (GSC) defect as a crucial pathogenic factor in AG in mouse and human. Moreover, a natural peptide is isolated from a traditional Chinese medicine that activated GSCs to regenerate gastric epithelia in experimental AG models and revitalized the atrophic gastric organoids derived from patients. It is further shown that the peptide exerts its functions by stabilizing the EGF-EGFR complex and specifically activating the downstream ERK and Stat1 signaling. Overall, these findings advance the understanding of AG pathogenesis and open a new avenue for AG treatment.
Collapse
Affiliation(s)
- Ke Li
- Institute of Traditional Chinese Medicine and Stem Cell ResearchCollege of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
- Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Xiuying Ma
- Sichuan Engineering Research Center for Medicinal AnimalsSichuan Good Doctor Panxi Pharmaceutical Co., LtdChengdu610000China
| | - Zihao Li
- Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Ya Liu
- Institute of Traditional Chinese Medicine and Stem Cell ResearchCollege of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
| | - Guiyan Shen
- Institute of Traditional Chinese Medicine and Stem Cell ResearchCollege of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
| | - Zecheng Luo
- Institute of Traditional Chinese Medicine and Stem Cell ResearchCollege of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
| | - Dong Wang
- Institute of Traditional Chinese Medicine and Stem Cell ResearchCollege of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
| | - Li Xia
- Department of PathophysiologyKey Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhengting Wang
- Department of GastroenterologyRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Ming Tian
- Department of BurnRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200025China
| | - Huijuan Liu
- Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Funeng Geng
- Sichuan Engineering Research Center for Medicinal AnimalsSichuan Good Doctor Panxi Pharmaceutical Co., LtdChengdu610000China
| | - Baojie Li
- Institute of Traditional Chinese Medicine and Stem Cell ResearchCollege of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengdu611137China
- Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| |
Collapse
|
13
|
Zhao Y, Tang H, Xu J, Sun F, Zhao Y, Li Y. HNF4A-Bridging the Gap Between Intestinal Metaplasia and Gastric Cancer. Evol Bioinform Online 2024; 20:11769343241249017. [PMID: 38680615 PMCID: PMC11047246 DOI: 10.1177/11769343241249017] [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: 10/15/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024] Open
Abstract
Background Intestinal metaplasia (IM) of gastric epithelium has traditionally been regarded as an irreversible stage in the process of the Correa cascade. Exploring the potential molecular mechanism of IM is significant for effective gastric cancer prevention. Methods The GSE78523 dataset, obtained from the Gene Expression Omnibus (GEO) database, was analyzed using RStudio software to identify the differently expressed genes (DEGs) between IM tissues and normal gastric epithelial tissues. Subsequently, gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, Gene Set Enrichment Analysis (GESA), and protein-protein interaction (PPI) analysis were used to find potential genes. Additionally, the screened genes were analyzed for clinical, immunological, and genetic correlation aspects using single gene clinical correlation analysis (UALCAN), Tumor-Immune System Interactions Database (TISIDB), and validated through western blot experiments. Results Enrichment analysis showed that the lipid metabolic pathway was significantly associated with IM tissues and the apolipoprotein B (APOB) gene was identified in the subsequent analysis. Experiment results and correlation analysis showed that the expression of APOB was higher in IM tissues than in normal tissues. This elevated expression of APOB was also found to be associated with the expression levels of hepatocyte nuclear factor 4A (HNF4A) gene. HNF4A was also found to be associated with immune cell infiltration to gastric cancer and was linked to the prognosis of gastric cancer patients. Moreover, HNF4A was also highly expressed in both IM tissues and gastric cancer cells. Conclusion Our findings indicate that HNF4A regulates the microenvironment of lipid metabolism in IM tissues by targeting APOB. Higher expression of HNF4A tends to lead to a worse prognosis in gastric cancer patients implying it may serve as a predictive indicator for the progression from IM to gastric cancer.
Collapse
Affiliation(s)
| | | | - Jianhua Xu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Feifei Sun
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuanyuan Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Li
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
14
|
Tong QY, Pang MJ, Hu XH, Huang XZ, Sun JX, Wang XY, Burclaff J, Mills JC, Wang ZN, Miao ZF. Gastric intestinal metaplasia: progress and remaining challenges. J Gastroenterol 2024; 59:285-301. [PMID: 38242996 DOI: 10.1007/s00535-023-02073-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/26/2023] [Indexed: 01/21/2024]
Abstract
Most gastric cancers arise in the setting of chronic inflammation which alters gland organization, such that acid-pumping parietal cells are lost, and remaining cells undergo metaplastic change in differentiation patterns. From a basic science perspective, recent progress has been made in understanding how atrophy and initial pyloric metaplasia occur. However, pathologists and cancer biologists have long been focused on the development of intestinal metaplasia patterns in this setting. Arguably, much less progress has been made in understanding the mechanisms that lead to the intestinalization seen in chronic atrophic gastritis and pyloric metaplasia. One plausible explanation for this disparity lies in the notable absence of reliable and reproducible small animal models within the field, which would facilitate the investigation of the mechanisms underlying the development of gastric intestinal metaplasia (GIM). This review offers an in-depth exploration of the current state of research in GIM, shedding light on its pivotal role in tumorigenesis. We delve into the histological subtypes of GIM and explore their respective associations with tumor formation. We present the current repertoire of biomarkers utilized to delineate the origins and progression of GIM and provide a comprehensive survey of the available, albeit limited, mouse lines employed for modeling GIM and engage in a discussion regarding potential cell lineages that serve as the origins of GIM. Finally, we expound upon the myriad signaling pathways recognized for their activity in GIM and posit on their potential overlap and interactions that contribute to the ultimate manifestation of the disease phenotype. Through our exhaustive review of the progression from gastric disease to GIM, we aim to establish the groundwork for future research endeavors dedicated to elucidating the etiology of GIM and developing strategies for its prevention and treatment, considering its potential precancerous nature.
Collapse
Affiliation(s)
- Qi-Yue Tong
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Min-Jiao Pang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Xiao-Hai Hu
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Xuan-Zhang Huang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Jing-Xu Sun
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Xin-Yu Wang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China
| | - Joseph Burclaff
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, USA
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Department of Medicine, Departments of Pathology and Immunology, Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
| | - Zhi-Feng Miao
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 N. Nanjing Street, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
15
|
Pan J, Wu J, Zhang S, Wang K, Ji G, Zhou W, Dang Y. Targeted metabolomics revealed the mechanisms underlying the role of Liansu capsule in ameliorating functional dyspepsia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117568. [PMID: 38092317 DOI: 10.1016/j.jep.2023.117568] [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: 08/25/2023] [Revised: 11/26/2023] [Accepted: 12/06/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liansu capsule could alleviate dyspeptic symptoms; however, the mechanisms underlying its role in treating functional dyspepsia (FD) remain unclear. AIM OF THE STUDY To elucidate the mechanism underlying the efficacy of Liansu capsule in alleviating FD symptoms. MATERIALS AND METHODS Thirty-six male mice were randomly divided into the following six groups: control, model, low-strength Liansu, moderate-strength Liansu, high-strength Liansu, and domperidone groups. Small intestine propulsion rate, gastric residual rate and histopathological analysis were performed to evaluate efficacy of Liansu capsule. Levels of interleukin-1β, interleukin-6, tumor necrosis factor α, phosphorylation of p65, ghrelin and gastrin were verified by real-time quantitative polymerase chain reaction and immunofluorescence assays. Targeted metabolomic analyses, western blotting and immunofluorescence assays were used to explore the mechanism of Liansu capsule in ameliorating FD. RESULTS The Liansu capsule significantly ameliorated the symptoms of FD, and markedly increased the levels of ghrelin and gastrin. Moreover, Liansu capsule significantly downregulated the levels of the proinflammatory cytokine interleukin-1β, interleukin-6, tumor necrosis factor α, and inhibited the phosphorylation of p65. Targeted metabolomic analyses showed that Liansu capsule significantly reduced the levels of deoxycholic acid and hyodeoxycholic acid, which were significantly elevated in the model group. Furthermore, these results showed that deoxycholic acid and hyodeoxycholic acid markedly promoted the levels of Takeda G-protein-coupled receptor 5 (TGR5), phosphorylated signal transducer and activator of transcription 3 (STAT3), and Kruppel-like factor 5 (KLF5) in vitro. whereas, Liansu capsule significantly reduced the levels of TGR5, phosphorylated STAT3, and KLF5. CONCLUSION Our findings indicated that Liansu capsule improved FD by regulating the deoxycholic acid/hyodeoxycholic acid-TGR5-STAT3-KLF5 axis. The findings reveal a novel mechanism underlying the role of Liansu capsule, which may be a promising therapeutic strategy for FD.
Collapse
Affiliation(s)
- Jiashu Pan
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Jiaxuan Wu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Shengan Zhang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Kai Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yanqi Dang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
16
|
Li T, Ding N, Guo H, Hua R, Lin Z, Tian H, Yu Y, Fan D, Yuan Z, Gonzalez FJ, Wu Y. A gut microbiota-bile acid axis promotes intestinal homeostasis upon aspirin-mediated damage. Cell Host Microbe 2024; 32:191-208.e9. [PMID: 38237593 PMCID: PMC10922796 DOI: 10.1016/j.chom.2023.12.015] [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: 07/10/2023] [Revised: 11/18/2023] [Accepted: 12/21/2023] [Indexed: 02/17/2024]
Abstract
Aspirin-related gastrointestinal damage is of growing concern. Aspirin use modulates the gut microbiota and associated metabolites, such as bile acids (BAs), but how this impacts intestinal homeostasis remains unclear. Herein, using clinical cohorts and aspirin-treated mice, we identified an intestinal microbe, Parabacteroides goldsteinii, whose growth is suppressed by aspirin. Mice supplemented with P. goldsteinii or its BA metabolite, 7-keto-lithocholic acid (7-keto-LCA), showed reduced aspirin-mediated damage of the intestinal niche and gut barrier, effects that were lost with a P. goldsteinii hdhA mutant unable to generate 7-keto-LCA. Specifically, 7-keto-LCA promotes repair of the intestinal epithelium by suppressing signaling by the intestinal BA receptor, farnesoid X receptor (FXR). 7-Keto-LCA was confirmed to be an FXR antagonist that facilitates Wnt signaling and thus self-renewal of intestinal stem cells. These results reveal the impact of oral aspirin on the gut microbiota and intestinal BA metabolism that in turn modulates gastrointestinal homeostasis.
Collapse
Affiliation(s)
- Ting Li
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Ning Ding
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Hanqing Guo
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rui Hua
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zehao Lin
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Huohuan Tian
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yue Yu
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Daiming Fan
- Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China.
| | - Frank J Gonzalez
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Yue Wu
- Department of Cardiovascular Medicine, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Key Laboratory of Molecular Cardiology, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China.
| |
Collapse
|
17
|
Liu CX, Gao Y, Xu XF, Jin X, Zhang Y, Xu Q, Ding HX, Li BJ, Du FK, Li LC, Zhong MW, Zhu JK, Zhang GY. Bile acids inhibit ferroptosis sensitivity through activating farnesoid X receptor in gastric cancer cells. World J Gastroenterol 2024; 30:485-498. [PMID: 38414591 PMCID: PMC10895598 DOI: 10.3748/wjg.v30.i5.485] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/12/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is associated with high mortality rates. Bile acids (BAs) reflux is a well-known risk factor for GC, but the specific mechanism remains unclear. During GC development in both humans and animals, BAs serve as signaling molecules that induce metabolic reprogramming. This confers additional cancer phenotypes, including ferroptosis sensitivity. Ferroptosis is a novel mode of cell death characterized by lipid peroxidation that contributes universally to malignant progression. However, it is not fully defined if BAs can influence GC progression by modulating ferroptosis. AIM To reveal the mechanism of BAs regulation in ferroptosis of GC cells. METHODS In this study, we treated GC cells with various stimuli and evaluated the effect of BAs on the sensitivity to ferroptosis. We used gain and loss of function assays to examine the impacts of farnesoid X receptor (FXR) and BTB and CNC homology 1 (BACH1) overexpression and knockdown to obtain further insights into the molecular mechanism involved. RESULTS Our data suggested that BAs could reverse erastin-induced ferroptosis in GC cells. This effect correlated with increased glutathione (GSH) concentrations, a reduced GSH to oxidized GSH ratio, and higher GSH peroxidase 4 (GPX4) expression levels. Subsequently, we confirmed that BAs exerted these effects by activating FXR, which markedly increased the expression of GSH synthetase and GPX4. Notably, BACH1 was detected as an essential intermediate molecule in the promotion of GSH synthesis by BAs and FXR. Finally, our results suggested that FXR could significantly promote GC cell proliferation, which may be closely related to its anti-ferroptosis effect. CONCLUSION This study revealed for the first time that BAs could inhibit ferroptosis sensitivity through the FXR-BACH1-GSH-GPX4 axis in GC cells. This work provided new insights into the mechanism associated with BA-mediated promotion of GC and may help identify potential therapeutic targets for GC patients with BAs reflux.
Collapse
Affiliation(s)
- Chu-Xuan Liu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong Province, China
| | - Ying Gao
- Department of General Surgery, Linyi People's Hospital, Linyi 276034, Shandong Province, China
| | - Xiu-Fang Xu
- Department of Nursing, Huantai TCM Hospital, Zibo 256400, Shandong Province, China
| | - Xin Jin
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong Province, China
| | - Yun Zhang
- Center for Translational medical Research, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Qian Xu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Huan-Xin Ding
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Bing-Jun Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Fang-Ke Du
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Lin-Chuan Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Ming-Wei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Jian-Kang Zhu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| | - Guang-Yong Zhang
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, Shandong Province, China
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, Shandong Province, China
| |
Collapse
|
18
|
Xie D, Wu C, Wang D, Nisma Lena BA, Liu N, Ye G, Sun M. Wei-fu-chun tablet halted gastric intestinal metaplasia and dysplasia associated with inflammation by regulating the NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117020. [PMID: 37567428 DOI: 10.1016/j.jep.2023.117020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chi006Eese herbal medicine Weifuchun Tablets (WFC) approved by the State Food and Drug Administration in 1982 has been widely used in treating a variety of chronic stomach disorders including Chronic atrophic gastritis (CAG) and Gastric precancerous lesions in China clinically. This study aimed to investigate the efficacy and potential mechanism of WFC in treating Gastric intestinal metaplasia (GIM) and Gastric dysplasia (GDys). MATERIALS AND METHODS Rat GIM and GDys established by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) combined with hot paste, ethanol injury, and intermittent fasting were intervened by WFC. Body weight, histopathology, pH of gastric acid, pepsin activity, intestinal metaplasia index and inflammation were detected. Rat bone marrow derived macrophages (BMDMs) pretreated with WFC were stimulated by LPS. Inflammatory factors and the nuclear factor-kappa B (NF-κB) pathway were assessed. GES-1 cells pretreated by WFC were stimulated by MNNG and TNF-α, intestinal metaplasia index, the NF-κB pathway and interaction between P65 and CDX2 were detected. RESULTS WFC improved rat body weight, histopathology, pH value of gastric acid, activity of gastric pepsin, intestinal metaplasia (CDX2), inflammation (IL-1β, IL-6 and TNF-α), macrophage aggregation (CD68) in gastric mucosa in rat GIM and GDys. WFC inhibited inflammation (IL-1β and TNF-α) by inactivating the NF-κB pathway. WFC reduced the expression of CDX2 by inhibiting the binding of CDX2 promoter TSS upstream region with p65. CONCLUSION WFC blocked GIM and GDys associated with inflammation by regulating the NF-κB pathway.
Collapse
Affiliation(s)
- Dong Xie
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao Wu
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Wang
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bahaji Azami Nisma Lena
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ningning Liu
- Department of Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guan Ye
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, China.
| | - Mingyu Sun
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
19
|
Marzhoseyni Z, Shaghaghi Z, Alvandi M, Shirvani M. Investigating the Influence of Gut Microbiota-related Metabolites in Gastrointestinal Cancer. Curr Cancer Drug Targets 2024; 24:612-628. [PMID: 38213140 DOI: 10.2174/0115680096274860231111210214] [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: 07/30/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 01/13/2024]
Abstract
Gastrointestinal (GI) cancer is a major health concern due to its prevalence, impact on well-being, high mortality rate, economic burden, and potential for prevention and early detection. GI cancer research has made remarkable strides in understanding biology, risk factors, and treatment options. An emerging area of research is the gut microbiome's role in GI cancer development and treatment response. The gut microbiome, vital for digestion, metabolism, and immune function, is increasingly linked to GI cancers. Dysbiosis and alterations in gut microbe composition may contribute to cancer development. Scientists study how specific bacteria or microbial metabolites influence cancer progression and treatment response. Modulating the gut microbiota shows promise in enhancing treatment efficacy and preventing GI cancers. Gut microbiota dysbiosis can impact GI cancer through inflammation, metabolite production, genotoxicity, and immune modulation. Microbes produce metabolites like short-chain fatty acids, bile acids, and secondary metabolites. These affect host cells, influencing processes like cell proliferation, apoptosis, DNA damage, and immune regulation, all implicated in cancer development. This review explores the latest research on gut microbiota metabolites and their molecular mechanisms in GI cancers. The hope is that this attempt will help in conducting other relevant research to unravel the precise mechanism involved, identify microbial signatures associated with GI cancer, and develop targets.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Iran, Sari, Iran
| | - Zahra Shaghaghi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maria Shirvani
- Infectious Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
20
|
Che J, Zhao Y, Gu B, Li S, Li Y, Pan K, Sun T, Han X, Lv J, Zhang S, Fan B, Li C, Wang C, Wang J, Zhang T. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with the progression of gastroesophageal cancer. BMC Cancer 2023; 23:1238. [PMID: 38102546 PMCID: PMC10724912 DOI: 10.1186/s12885-023-11744-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Previous metabolic studies in upper digestive cancer have mostly been limited to cross-sectional study designs, which hinders the ability to effectively predict outcomes in the early stage of cancer. This study aims to identify key metabolites and metabolic pathways associated with the multistage progression of epithelial cancer and to explore their predictive value for gastroesophageal cancer (GEC) formation and for the early screening of esophageal squamous cell carcinoma (ESCC). METHODS A case-cohort study within the 7-year prospective Esophageal Cancer Screening Cohort of Shandong Province included 77 GEC cases and 77 sub-cohort individuals. Untargeted metabolic analysis was performed in serum samples. Metabolites, with FDR q value < 0.05 and variable importance in projection (VIP) > 1, were selected as differential metabolites to predict GEC formation using Random Forest (RF) models. Subsequently, we evaluated the predictive performance of these differential metabolites for the early screening of ESCC. RESULTS We found a distinct metabolic profile alteration in GEC cases compared to the sub-cohort, and identified eight differential metabolites. Pathway analyses showed dysregulation in D-glutamine and D-glutamate metabolism, nitrogen metabolism, primary bile acid biosynthesis, and steroid hormone biosynthesis in GEC patients. A panel of eight differential metabolites showed good predictive performance for GEC formation, with an area under the receiver operating characteristic curve (AUC) of 0.893 (95% CI = 0.816-0.951). Furthermore, four of the GEC pathological progression-related metabolites were validated in the early screening of ESCC, with an AUC of 0.761 (95% CI = 0.716-0.805). CONCLUSIONS These findings indicated a panel of metabolites might be an alternative approach to predict GEC formation, and therefore have the potential to mitigate the risk of cancer progression at the early stage of GEC.
Collapse
Affiliation(s)
- Jiajing Che
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yongbin Zhao
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Bingbing Gu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuting Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yunfei Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Keyu Pan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Tiantian Sun
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xinyue Han
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jiali Lv
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuai Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Bingbing Fan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chunxia Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Cheng Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Jialin Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, China.
| | - Tao Zhang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
21
|
Lei X, Cui ZY, Huang XJ. Exploration of gastric carcinogenesis from the relationship between bile acids and intestinal metaplasia and intragastric microorganisms (H. pylori and non-H. pylori). J Cancer Res Clin Oncol 2023; 149:16947-16956. [PMID: 37707577 DOI: 10.1007/s00432-023-05407-5] [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: 07/13/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Gastric cancer (GC) is a prevalent form of cancer, with Helicobacter pylori (H. pylori) infection being the most common risk factor. Recent studies have highlighted the role of long-term irritation of the gastric mucosa caused by bile reflux in the development of cancer. Bile acids (BAs), which are a significant component in bile reflux, have the potential to promote gastric carcinogenesis through various mechanisms. These mechanisms include the induction of intestinal metaplasia (IM), inhibition of H. pylori activity, modification of H. pylori colonization, and alteration of the abundance and composition of microorganisms in the stomach. Defining the mechanism of bile acid-induced gastric carcinogenesis could potentially be an effective approach to prevent GC. Hence, this paper aims to review the mechanism of bile acid-induced IM, the association between BAs and H. pylori infection as well as microorganisms in the stomach, and the correlation between BAs and gastric carcinogenesis. The ultimate goal is to elucidate the role of BAs in the development of GC.
Collapse
Affiliation(s)
- X Lei
- Department of Gastroenterology, The Lanzhou University Second Hospital, No. 82 of Linxia Street, Chengguan District, Lanzhou, 730030, China
| | - Z Y Cui
- Department of Gastroenterology, The Lanzhou University Second Hospital, No. 82 of Linxia Street, Chengguan District, Lanzhou, 730030, China
| | - X J Huang
- Department of Gastroenterology, The Lanzhou University Second Hospital, No. 82 of Linxia Street, Chengguan District, Lanzhou, 730030, China.
| |
Collapse
|
22
|
Zhai H, Zhong S, Wu R, Mo Z, Zheng S, Xue J, Meng H, Liu M, Chen X, Zhang G, Zheng X, Du F, Li R, Zhou B. Suppressing circIDE/miR-19b-3p/RBMS1 axis exhibits promoting-tumour activity through upregulating GPX4 to diminish ferroptosis in hepatocellular carcinoma. Epigenetics 2023; 18:2192438. [PMID: 36989117 PMCID: PMC10064926 DOI: 10.1080/15592294.2023.2192438] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Ferroptosis is a newly characterized form of iron-dependent non-apoptotic cell death, which is closely associated with cancer progression. However, the functions and mechanisms in regulation of escaping from ferroptosis during hepatocellular carcinoma (HCC) progression remain unknown. In this study, we reported that the RNA binding motif single stranded interacting protein 1 (RBMS1) participated in HCC development,and functioned as a regulator of ferroptosis. Clinically, the downregulation of RBMS1 occurred in HCC tissues, and low RBMS1 expression was associated with worse HCC patients survival. Mechanistically, RBMS1 overexpression inhibited HCC cell growth by attenuating the expression of glutathione peroxidase 4 (GPX4)and further facilitated ferroptosis in vitro and in vivo. More importantly, a novel circIDE (hsa_circ_0000251) was identified to elevate RBMS1 expression via sponging miR-19b-3p in HCC cells. Collectively, our findings established circIDE/miR-19b-3p/RBMS1 axis as a regulator of ferroptosis, which could be a promising therapeutic target and prognostic factor.
Collapse
Affiliation(s)
- Hang Zhai
- Department of Quality and Safety Management, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sisi Zhong
- Department of Quality and Safety Management, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Runxin Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhaohong Mo
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shiyang Zheng
- Department of Head and Neck surgery, Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - Jinhua Xue
- Department of Physiology, the School of Basic Medical Sciences of Gannan Medical University, Ganzhou, China
| | - Hongyu Meng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Maosheng Liu
- Department of Gastroentrology, the First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xianyu Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiyan Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Fei Du
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ruixi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Boxuan Zhou
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Zhang S, Shen Y, Liu H, Zhu D, Fang J, Pan H, Liu W. Inflammatory microenvironment in gastric premalignant lesions: implication and application. Front Immunol 2023; 14:1297101. [PMID: 38035066 PMCID: PMC10684945 DOI: 10.3389/fimmu.2023.1297101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Gastric precancerous lesions (GPL) are a major health concern worldwide due to their potential to progress to gastric cancer (GC). Understanding the mechanism underlying the transformation from GPL to GC can provide a fresh insight for the early detection of GC. Although chronic inflammation is prevalent in the GPL, how the inflammatory microenvironment monitored the progression of GPL-to-GC are still elusive. Inflammation has been recognized as a key player in the progression of GPL. This review aims to provide an overview of the inflammatory microenvironment in GPL and its implications for disease progression and potential therapeutic applications. We discuss the involvement of inflammation in the progression of GPL, highlighting Helicobacter pylori (H. pylori) as a mediator for inflammatory microenvironment and a key driver to GC progression. We explore the role of immune cells in mediating the progression of GPL, and focus on the regulation of inflammatory molecules in this disease. Furthermore, we discuss the potential of targeting inflammatory pathways for GPL. There are currently no specific drugs for GPL treatment, but traditional Chinese Medicine (TCM) and natural antioxidants, known as antioxidant and anti-inflammatory properties, exhibit promising effects in suppressing or reversing the progression of GPL. Finally, the challenges and future perspectives in the field are proposed. Overall, this review highlights the central role of the inflammatory microenvironment in the progression of GPL, paving the way for innovative therapeutic approaches in the future.
Collapse
Affiliation(s)
- Shengxiong Zhang
- Rehabilitation Department, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, China
- Department of Spleen and Stomach, GuangZhou Tianhe District Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Shen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Di Zhu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiansong Fang
- Science and Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huafeng Pan
- Science and Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
24
|
Nie K, Zheng Z, Li X, Chang Y, Liu F, Wang X. Explore the active ingredients and potential mechanisms of JianPi QingRe HuaYu Methods in the treatment of gastric inflammation-cancer transformation by network pharmacology and experimental validation. BMC Complement Med Ther 2023; 23:411. [PMID: 37964307 PMCID: PMC10644588 DOI: 10.1186/s12906-023-04232-0] [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: 03/28/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND JianPi QingRe HuaYu Methods (JQH) have been long used to treat chronic atrophic gastritis (CAG) and precancerous lesions of gastric cancer (PLGC). However, whether JQH can inhibit the transformation of gastritis to gastric cancer (GC) remains unclear. METHODS Herein, we first retrieved the active ingredients and targets of JQH from the TCMSP database and the targets related to the gastric inflammation-cancer transformation from public databases. Differentially expressed genes (DEGs) related to gastric inflammation-cancer transformation were identified from the Gene Expression Omnibus (GEO) database. Then, we obtained the potential therapeutic targets of JQH in treating gastric inflammation-cancer transformation by intersecting drugs and disease targets. The Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction (PPI) analyses of the potential therapeutic targets were conducted using R software. Next, we conducted molecular docking and in vitro experiments to validate our results. RESULTS We obtained 214 potential therapeutic targets of JQH by intersecting drugs and disease targets. We found that the potential mechanisms of JQH in treating gastric inflammation-cancer transformation might be related to JAK-STAT, Wnt, p53 and VEGF signaling pathways. The molecular docking indicated that quercetin, as the main active ingredient of JQH, might inhibit gastric inflammation-cancer transformation by binding with specific receptors. Our experimental results showed that quercetin inhibited cells proliferation (P < 0.001), promoted cell apoptosis (P < 0.001), reduced the secretion of pro-inflammatory cytokines (P < 0.001) and promoted the secretion of anti-inflammatory cytokines (P < 0.001) in MNNG-induced GES-1 cells. Furthermore, quercetin inhibited cells proliferation (P < 0.001) and reduced mRNA and protein level of markers of PLGC (P < 0.001) in CDCA-induced GES-1 cells. CONCLUSION These results provide the material basis and regulatory mechanisms of JQH in treating gastric inflammation-cancer transformation.
Collapse
Affiliation(s)
- Kechao Nie
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhihua Zheng
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
| | - Xiushen Li
- Shenzhen University General Hospital, Shenzhen, 518060, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yonglong Chang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - FengBin Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaoyu Wang
- School of Health Science, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
25
|
Cheng L, Yan H, Liu Y, Guan G, Cheng P. Dissecting multifunctional roles of forkhead box transcription factor D1 in cancers. Biochim Biophys Acta Rev Cancer 2023; 1878:188986. [PMID: 37716516 DOI: 10.1016/j.bbcan.2023.188986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023]
Abstract
As a member of the forkhead box (FOX) family of transcription factors (TF), FOXD1 has recently been implicated as a crucial regulator in a variety of human cancers. Accumulating evidence has established dysregulated and aberrant FOXD1 signaling as a prominent feature in cancer development and progression. However, there is a lack of systematic review on this topic. Here, we summarized the present understanding of FOXD1 functions in cancer biology and reviewed the downstream targets and upstream regulatory mechanisms of FOXD1 as well as the related signaling pathways within the context of current reports. We highlighted the functional features of FOXD1 in cancers to identify the future research consideration of this multifunctional transcription factor and potential therapeutic strategies targeting its oncogenic activity.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Haixu Yan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Gefei Guan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China.
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
26
|
Wu T, Yang Z, Chen W, Jiang M, Xiao Z, Su X, Jiao Z, Yu Y, Chen S, Song M, Yang A. miR-30e-5p-mediated FOXD1 promotes cell proliferation by blocking cellular senescence and apoptosis through p21/CDK2/Rb signaling in head and neck carcinoma. Cell Death Discov 2023; 9:295. [PMID: 37563111 PMCID: PMC10415393 DOI: 10.1038/s41420-023-01571-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023] Open
Abstract
Forkhead box D1 (FOXD1) belongs to the FOX protein family, which has been found to function as a oncogene in multiple cancer types, but its role in head and neck squamous cell carcinoma (HNSCC) requires further investigation. Our research aimed to investigate the function of FOXD1 in HNSCC. Bioinformatics analysis indicated that mRNA level of FOXD1 was highly expressed in HNSCC tissues, and over-expressed FOXD1 was related to poor prognosis. Moreover, FOXD1 knockdown increased the ratio of senescent cells but decreased the proliferation ability, while FOXD1 overexpression obtained the opposite results. In vitro experiments revealed that FOXD1 bound to the p21 promoter and inhibited its transcription, which blocked the cyclin dependent kinase 2 (CDK2)/retinoblastoma (Rb) signaling pathway, thus preventing senescence and accelerating proliferation of tumor cells. CDK2 inhibitor could reverse the process to some extent. Further research has shown that miR-3oe-5p serves as a tumor suppressant by repressing the translation of FOXD1 through combining with the 3'-untranslated region (UTR). Thus, FOXD1 resists cellular senescence and facilitates HNSCC cell proliferation by affecting the expression of p21/CDK2/Rb signaling, suggesting that FOXD1 may be a potential curative target for HNSCC.
Collapse
Affiliation(s)
- Tong Wu
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Zhongyuan Yang
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Weichao Chen
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Mingjie Jiang
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Zhichao Xiao
- Department of Otolaryngology-Head Neck Surgery, Loudi Central Hospital, Loudi, Hunan Province, China
| | - Xuan Su
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Zan Jiao
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Yongchao Yu
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China
| | - Shuwei Chen
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China.
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China.
| | - Ming Song
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China.
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China.
| | - Ankui Yang
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Oncology in Southern China, Guangzhou, 510060, China.
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
27
|
Li L, Ruan J, Ma Y, Xu X, Qin H, Tian X, Hu J. Identification of key modules and micro RNAs associated with colorectal cancer via a weighted gene co-expression network analysis and competing endogenous RNA network analysis. J Gastrointest Oncol 2023; 14:1320-1330. [PMID: 37435199 PMCID: PMC10331739 DOI: 10.21037/jgo-23-244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/09/2023] [Indexed: 07/13/2023] Open
Abstract
Background Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide, and the incidence of CRC has increased rapidly in recent years. Due to the high invasiveness of colonoscopy and the low accuracy of alternative diagnostic methods, the diagnosis of CRC remains a serious problem. Thus, molecular biomarkers for CRC need to be identified. Methods In this study, RNA-sequencing data from The Cancer Genome Atlas (TCGA) database were used to identify the long non-coding RNAs (lncRNAs), messenger RNAs (mRNAs), and micro RNAs (miRNAs) that were differentially expressed between the CRC and normal tissues. Based on the gene expression and clinical features, the results of the weighted gene co-expression network analysis (WGCNA) and the binding relationships between miRNAs and lncRNAs and mRNAs were used to establish a CRC-related competing endogenous RNA (ceRNA) network. Results The core miRNAs (i.e., mir-874, mir-92a-1, and mir-940) in the network were identified. Among them, mir-874 was negatively correlated with the overall survival (OS) of patients. The protein-coding genes in the ceRNA network included IZUMO4, WT1, NPEPL1, TEX22, PPFIA4, and SFXN3, and the lncRNAs were LINC00858 and PRR7-AS1. These genes were significantly highly expressed in CRC according to validations in other independent data sets. Conclusions In conclusion, this study established a network of the co-expressed ceRNAs associated with CRC and identified the genes and miRNAs related to the prognosis of CRC patients.
Collapse
Affiliation(s)
- Lifang Li
- Emergency Department, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jingxiong Ruan
- Clinical Laboratory, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yanfen Ma
- Clinical Laboratory, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xin Xu
- Clinical Laboratory, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hao Qin
- Clinical Laboratory, Shaanxi Provincial Cancer Hospital, Xi’an, China
| | - Xudong Tian
- Clinical Laboratory, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jian Hu
- Clinical Laboratory, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
28
|
Zeng X, Yang M, Ye T, Feng J, Xu X, Yang H, Wang X, Bao L, Li R, Xue B, Zang J, Huang Y. Mitochondrial GRIM-19 loss in parietal cells promotes spasmolytic polypeptide-expressing metaplasia through NLR family pyrin domain-containing 3 (NLRP3)-mediated IL-33 activation via a reactive oxygen species (ROS) -NRF2- Heme oxygenase-1(HO-1)-NF-кB axis. Free Radic Biol Med 2023; 202:46-61. [PMID: 36990300 DOI: 10.1016/j.freeradbiomed.2023.03.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/05/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Spasmolytic polypeptide-expressing metaplasia (SPEM), as a pre-neoplastic precursor of intestinal metaplasia (IM), plays critical roles in the development of chronic atrophic gastritis (CAG) and gastric cancer (GC). However, the pathogenetic targets responsible for the SPEM pathogenesis remain poorly understood. Gene associated with retinoid-IFN-induced mortality 19 (GRIM-19), an essential subunit of the mitochondrial respiratory chain complex I, was progressively lost along with malignant transformation of human CAG, little is known about the potential link between GRIM-19 loss and CAG pathogenesis. Here, we show that lower GRIM-19 is associated with higher NF-кB RelA/p65 and NLR family pyrin domain-containing 3 (NLRP3) levels in CAG lesions. Functionally, GRIM-19 deficiency fails to drive direct differentiation of human GES-1 cells into IM or SPEM-like cell lineages in vitro, whereas parietal cells (PCs)-specific GRIM-19 knockout disturbs gastric glandular differentiation and promotes spontaneous gastritis and SPEM pathogenesis without intestinal characteristics in mice. Mechanistically, GRIM-19 loss causes chronic mucosal injury and aberrant NRF2 (Nuclear factor erythroid 2-related factor 2)- HO-1 (Heme oxygenase-1) activation via reactive oxygen species (ROS)-mediated oxidative stress, resulting in aberrant NF-кB activation by inducing p65 nuclear translocation via an IKK/IкB partner, while NRF2-HO-1 activation contributes to GRIM-19 loss-driven NF-кB activation via a positive feedback NRF2-HO-1 loop. Furthermore, GRIM-19 loss did not cause obvious PCs loss but triggers NLRP3 inflammasome activation in PCs via a ROS-NRF2-HO-1-NF-кB axis, leading to NLRP3-dependent IL-33 expression, a key mediator for SPEM formation. Moreover, intraperitoneal administration of NLRP3 inhibitor MCC950 drastically attenuates GRIM-19 loss-driven gastritis and SPEM in vivo. Our study suggests that mitochondrial GRIM-19 maybe a potential pathogenetic target for the SPEM pathogenesis, and its deficiency promotes SPEM through NLRP3/IL-33 pathway via a ROS-NRF2-HO-1-NF-кB axis. This finding not only provides a causal link between GRIM-19 loss and SPEM pathogenesis, but offers potential therapeutic strategies for the early prevention of intestinal GC.
Collapse
Affiliation(s)
- Xin Zeng
- Institute of Paediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China; Department of Laboratory Medicine, The Third People's Hospital of Chengdu, Chengdu, 610031, China
| | - Meihua Yang
- Departments of Neurology, Washington University School of Medicine and Barnes-Jewish Hospital, Saint Louis, 63110, MO, USA
| | - Tingbo Ye
- Department of Laboratory Medicine, The Third People's Hospital of Chengdu, Chengdu, 610031, China
| | - Jinmei Feng
- Institute of Paediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Xiaohui Xu
- Institute of Paediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Huaan Yang
- Department of Urologic Surgery, Yubei District People's Hospital, Chongqing, 401120, China
| | - Xin Wang
- Ministry of Education Key Laboratory of Molecular Biology for Infectious Diseases, Chongqing Medical University, Chongqing, 40016, China
| | - Liming Bao
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, NY, 10065, USA
| | - Rui Li
- Department of Laboratory Medicine, The Third People's Hospital of Chengdu, Chengdu, 610031, China
| | - Bingqian Xue
- Department of Laboratory Medicine, The Third People's Hospital of Chengdu, Chengdu, 610031, China
| | - Jinbao Zang
- Institute of Paediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yi Huang
- Institute of Paediatrics, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
29
|
Yu L, Sui B, Zhang X, Liu J, Hao X, Zheng L. miR-92a-1-5p enriched prostate cancer extracellular vesicles regulate osteoclast function via MAPK1 and FoxO1. J Exp Clin Cancer Res 2023; 42:109. [PMID: 37131239 PMCID: PMC10152631 DOI: 10.1186/s13046-023-02685-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/25/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND We have previously reported that extracellular vesicles (EVs) derived from osteoblastic, osteoclastic and mixed prostate cancer cells promote osteoclast differentiation and inhibit osteoblast differentiation via transferring miR-92a-1-5p. In the present study, we focused on engineering miR-92a-1-5p into EVs and determining any therapeutic roles and mechanisms of the engineered EVs. METHODS A stable prostate cancer cell line (MDA PCa 2b) overexpressing miR-92a-1-5p was constructed by lentivirus, and EVs were isolated by ultracentrifugation. The overexpression of miR-92a-1-5p in both cells and EVs was tested using qPCR. Osteoclast function was evaluated by Trap staining, mRNA expression of osteoclastic markers ctsk and trap, immunolabeling of CTSK and TRAP and microCT using either in vitro and in vivo assays. Target gene of miR-92a-1-5p was proved by a dual-luciferase reporter assay system. siRNAs were designed and used for transient expression in order to determine the role of downstream genes on osteoclast differentiation. RESULTS Stable overexpression cells of miRNA-92a-5p was associated with EVs upregulating this microRNA, as confirmed by qPCR. Further, miR-92a-1-5p enriched EVs promote osteoclast differentiation in vitro by reducing MAPK1 and FoxO1 expression, associated with increased osteoclast function as shown by TRAP staining and mRNA expression of osteoclast functional genes. siRNA targeting MAPK1 or FoxO1 resulted in similar increase in osteoclast function. In vivo, the miR-92a-1-5p enriched EVs given via i.v. injection promote osteolysis, which was associated with reduction of MAPK1 and FoxO1 expression in bone marrow. CONCLUSION These experiments suggest that miR-92a-1-5p enriched EVs regulate osteoclast function via reduction of MAPK1 and FoxO1.
Collapse
Affiliation(s)
- Lijuan Yu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Institute of Laboratory Medicine Center of Chinese People's Liberation Army (PLA), Xijing Hospital, Fourth Military Medical University (Air Force Medical University), , Xi'an, 710032, China.
- Krefting Research Centre, Institute of Medicine, University of Gothenburg, 410530, Gothenburg, Sweden.
| | - Bingdong Sui
- Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University (Air Force Medical University), Xi'an, 710032, China
| | - Xin Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiayun Liu
- Institute of Laboratory Medicine Center of Chinese People's Liberation Army (PLA), Xijing Hospital, Fourth Military Medical University (Air Force Medical University), , Xi'an, 710032, China
| | - Xiaoke Hao
- Institute of Laboratory Medicine Center of Chinese People's Liberation Army (PLA), Xijing Hospital, Fourth Military Medical University (Air Force Medical University), , Xi'an, 710032, China.
- College of Medicine, Northwest University, Xi'an, 710032, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
30
|
Kotelevets SM, Chekh SA, Chukov SZ. Cancer risk stratification system and classification of gastritis: Perspectives. World J Meta-Anal 2023; 11:18-28. [DOI: 10.13105/wjma.v11.i1.18] [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: 10/26/2022] [Revised: 11/17/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
Kyoto global consensus reports that the current ICD-10 classification for gastritis is obsolete. The Kyoto classification of gastritis states that severe mucosal atrophy has a high risk of gastric cancer, while mild to moderate atrophy has a low risk. The updated Kimura-Takemoto classification of atrophic gastritis considers five histological types of multifocal corpus atrophic gastritis according to stages C2 to O3. This method of morphological diagnosis of atrophic gastritis increases sensitivity by 2.4 times for severe atrophy compared to the updated Sydney system. This advantage should be considered when stratifying the high risk of gastric cancer. The updated Kimura-Takemoto classification of atrophic gastritis should be used as a reference standard (gold standard) in studies of morpho-functional relationships to identify serological markers of atrophic gastritis with evidence-based effectiveness. The use of artificial intelligence in the serological screening of atrophic gastritis makes it possible to screen a large number of the population. During serological screening of atrophic gastritis and risk stratification of gastric cancer, it is advisable to use the Kyoto classification of gastritis with updated Kimura-Takemoto classification of atrophic gastritis.
Collapse
Affiliation(s)
- Sergey M Kotelevets
- Department of Therapy, North Caucasus State Academy, Cherkessk 369000, Karachay-Cherkess Republic, Russia
| | - Sergey A Chekh
- Department of Mathematics, North Caucasus State Academy, Cherkessk 369000, Karachay-Cherkess Republic, Russia
| | - Sergey Z Chukov
- Department of Pathological Anatomy, Stavropol State Medical University, Stavropol 355017, Stavropol region, Russia
| |
Collapse
|
31
|
Wang M, Lou E, Xue Z. The role of bile acid in intestinal metaplasia. Front Physiol 2023; 14:1115250. [PMID: 36891144 PMCID: PMC9986488 DOI: 10.3389/fphys.2023.1115250] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
A precancerous lesion of gastric cancer (GC), intestinal metaplasia (IM) is a pathological transformation of non-intestinal epithelium into an intestinal-like mucosa. It greatly raises the risk of developing the intestinal type of GC, which is frequently observed in the stomach and esophagus. It is understood that esophageal adenocarcinoma's precursor lesion, chronic gastroesophageal reflux disease (GERD), is what causes Barrett's esophagus (BE), an acquired condition. Recently, Bile acids (BAs), which are one of the compositions of gastric and duodenal contents, have been confirmed that it led to the occurrence and development of BE and gastric intestinal metaplasia (GIM). The objective of the current review is to discuss the mechanism of IM induced by bile acids. This review serves as a foundation for further research aimed at improving the way BE and GIM are currently managed.
Collapse
Affiliation(s)
- Menglei Wang
- Department of Digestive Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Enzhe Lou
- Department of Digestive Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Zengfu Xue
- Department of Digestive Diseases, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| |
Collapse
|
32
|
Zhuo XH, Sun JC, Zhong WJ, Lu Y. Negative correlations between bile reflux gastritis and Helicobacter pylori infection. Scand J Gastroenterol 2022; 57:1430-1434. [PMID: 35840539 DOI: 10.1080/00365521.2022.2094721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE There are a few articles to study the relationship between bile reflux gastritis (BRG) and H. pylori infection, and the results are debatable. This study set out to determine the relationship between BRG and Helicobacter pylori (H. pylori) infection. METHODS In this retrospective study, patients from January, 1st 2013 to January, 1st, 2021 were divided into two groups based on whether they had BRG. The control group was got by 1:1 propensity-score matching (PSM) based on age and sex. Then, the relationship between BRG and H. pylori in patients was analyzed via Chi-squared test and Phi (φ) detection. RESULTS 26449 patients were included in this study, and there were 1918 patients in each group after age and sex matching. patients with HP were responsible for 35% (9345/26449) and patients with BRG were 7% (1918/26449). Further relationship exploration, there is a negative, but weak, the relationship between BRG and HP infection (X2 = 45.62, p < .001, Phi (φ)= -0.109). CONCLUSION Patients with bile reflux may have less likely to get HP infection. HP eradication is an important thing for the prevention of gastric cancer and this study serves as a foundation and may provide directions for future research.
Collapse
Affiliation(s)
- Xian-Hua Zhuo
- Department of Otolaryngology Head and Neck Surgery, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong, China.,Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jia-Chen Sun
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wei-Jie Zhong
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yi Lu
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Guo H, Zhuang K, Ding N, Hua R, Tang H, Wu Y, Yuan Z, Li T, He S. High-fat diet induced cyclophilin B enhances STAT3/lncRNA-PVT1 feedforward loop and promotes growth and metastasis in colorectal cancer. Cell Death Dis 2022; 13:883. [PMID: 36266267 PMCID: PMC9584950 DOI: 10.1038/s41419-022-05328-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 01/23/2023]
Abstract
High-fat diet (HFD) has been implicated to promote colorectal cancer (CRC). Recently, oncogene Cyclophilin B (CypB) is reported to be induced by cholesterol. However, the role of CypB in CRC carcinogenesis and metastasis associated with HFD remains unknown. In the present study, we showed that HFD-induced CypB enhances proliferation and metastasis through an inflammation-driven circuit, including Signal Transducer and Activator of Transcription 3 (STAT3)-triggered transcription of lncRNA-PVT1, and its binding with CypB that promotes activation of STAT3. CypB was found to be upregulated in CRC, which was correlated with elevated body mass index and poor prognosis. HFD induced CypB expression and proinflammatory cytokines in colon of mice. Besides, CypB restoration facilitated growth, invasion and metastasis in CRC cells both in vitro and in vivo. Moreover, RIP sequencing data identified lncRNA-PVT1 as a functional binding partner of CypB. Mechanistically, PVT1 increased the phosphorylation and nuclear translocation of STAT3 in response to IL-6, through directly interaction with CypB, which impedes the binding of Suppressors Of Cytokine Signalling 3 (SOCS3) to STAT3. Furthermore, STAT3 in turn activated PVT1 transcription through binding to its promoter, forming a regulatory loop. Finally, this CypB/STAT3/PVT1 axis was verified in TCGA datasets and CRC tissue arrays. Our data revealed that CypB linked HFD and CRC malignancy by enhancing the CypB/STAT3/PVT1 feedforward axis and activation of STAT3.
Collapse
Affiliation(s)
- Hanqing Guo
- grid.43169.390000 0001 0599 1243First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China ,grid.43169.390000 0001 0599 1243Department of Gastroenterology, Xi’an Central Hospital, College of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Kun Zhuang
- grid.43169.390000 0001 0599 1243Department of Gastroenterology, Xi’an Central Hospital, College of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Ning Ding
- grid.43169.390000 0001 0599 1243First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Rui Hua
- grid.43169.390000 0001 0599 1243First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Hailing Tang
- grid.43169.390000 0001 0599 1243Department of Gastroenterology, Xi’an Central Hospital, College of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Yue Wu
- grid.43169.390000 0001 0599 1243First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China ,grid.452438.c0000 0004 1760 8119Department of Cardiovascular Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zuyi Yuan
- grid.43169.390000 0001 0599 1243First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China ,grid.452438.c0000 0004 1760 8119Department of Cardiovascular Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ting Li
- grid.43169.390000 0001 0599 1243First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China ,grid.452438.c0000 0004 1760 8119Department of Cardiovascular Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shuixiang He
- grid.43169.390000 0001 0599 1243First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
34
|
Zhang H, Qu X, Wang N, Zhang L, Yuan T, Shi M, Sun N, Yuan D, Ning H, Zhao M, Wang Y, Ni Z, Han C, Shi Y. Expression and significance of CDX2, FXR, and TGR5 in esophageal cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2022; 15:354-363. [PMID: 36237638 PMCID: PMC9547995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/22/2022] [Indexed: 06/16/2023]
Abstract
This study explored the expression and significance of three critical morphogenesis genes in normal esophagus, reflux esophagitis (RE), Barrett's esophagus (BE), esophageal adenocarcinoma (EA), and esophageal squamous cell carcinoma (ESCC). Esophageal tissue samples and tissue microarrays were used. CDX2, FXR, and TGR5 protein expression were measured by immunohistochemistry in normal esophageal, RE, BE, EA, and ESCC tissues. All 3 proteins had markedly changed expression during the progression of EA. The expressions of CDX2 and FXR were positively correlated in EA. In addition, TGR5 expression was positively correlated with CDX2 in RE and BE. The expressions of CDX2 and FXR were also positively correlated in ESCC. Although CDX2, FXR, and TGR5 were upregulated in ESCC, these factors might not be markers for the prognosis of ESCC. These results suggested that CDX2, FXR, and TGR5 might play different roles in EA and ESCC. They may represent novel therapeutic targets for patients with these cancers.
Collapse
Affiliation(s)
- Hongying Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
- Department of Intensive Care Unit, Shaanxi Provincial People’s HospitalXi’an 710032, Shaanxi Province, China
| | - Xiaodong Qu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Na Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Lifeng Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Ting Yuan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
- The 989 Hospital of The People’s Liberation ArmyLuoyang 471000, Henan Province, China
| | - Miao Shi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
- College of Postgraduates, Xi’an Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Nina Sun
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
- College of Postgraduates, Xi’an Medical UniversityXi’an 710032, Shaanxi Province, China
| | - Donghong Yuan
- Yanan University Affiliated HospitalYanan 716000, Shaanxi Province, China
| | - Hanbing Ning
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhou 450000, Henan Province, China
| | - Mengyun Zhao
- Xi’an No. 1 HospitalXi’an 710032, Shaanxi Province, China
| | - Yongxi Wang
- Xianyang Central HospitalXianyang 712000, Shaanxi Province, China
| | - Zhen Ni
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
- Chengdu Military Command General HospitalChengdu 610083, Sichuan Province, China
| | - Chuan Han
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
- Rocket Army Emei SanatoriumEmei 614200, Sichuan Province, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical UniversityXi’an 710032, Shaanxi Province, China
| |
Collapse
|
35
|
Bile Reflux Gastritis: Insights into Pathogenesis, Relevant Factors, Carcinomatous Risk, Diagnosis, and Management. Gastroenterol Res Pract 2022; 2022:2642551. [PMID: 36134174 PMCID: PMC9484982 DOI: 10.1155/2022/2642551] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
Bile reflux gastritis (BRG), a kind of gastrointestinal disorder in clinical practice, is characterized by regurgitation and inflammation. However, lack of guidelines leads to simple cognition and even ignorance of this disease for clinicians. Primarily, making the pathogenesis of BRG clear contributes to a correct and general understanding of this disease for physicians. Next, although recently there has been an increasing awareness among researchers in terms of the relevant factors for BRG, further studies involving large samples are still required to certify the relationship between them explicitly. Besides, researches have established that BRG is closely associated with the development of precancerous lesions and gastric cancer. Till now, there is still no golden standard for diagnosis of BRG. Nevertheless, advances in techniques, especially extensive applications of endoscopy and chemical analysis of reflux contents, have improved our ability to identify the occurrence of this disease as well as distinguishing physiological reflux from pathological reflux. Finally, it is fortunate for patients that more and more importance has been attached to the treatment of BRG. From lifestyle modification to drug therapy to surgery, all of them with the view of realizing symptomatic relief are employed for patients with BRG. In this review, we briefly evaluate this disorder based on the best available evidence, offering an overview of its complicated pathogenesis, diverse relevant factors, potential carcinomatous risk, modern diagnostic investigations, and effective therapeutic plans.
Collapse
|
36
|
Huang G, Wang S, Wang J, Tian L, Yu Y, Zuo X, Li Y. Bile reflux alters the profile of the gastric mucosa microbiota. Front Cell Infect Microbiol 2022; 12:940687. [PMID: 36159635 PMCID: PMC9500345 DOI: 10.3389/fcimb.2022.940687] [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: 05/12/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Bile reflux can cause inflammation, gastric mucosa atrophy, and diseases such as stomach cancer. Alkaline bile flowing back into the stomach affects the intragastric environment and can alter the gastric bacterial community. We sought to identify the characteristics of the stomach mucosal microbiota in patients with bile reflux. METHODS Gastric mucosal samples were collected from 52 and 40 chronic gastritis patients with and without bile reflux, respectively. The bacterial profile was determined using 16S rRNA gene analysis. RESULTS In the absence of H. pylori infection, the richness (based on the Sobs and Chao1 indices; P <0.05) and diversity (based on Shannon indices; P <0.05) of gastric mucosa microbiota were higher in patients with bile reflux patients than in those without. There was a marked difference in the microbiota structure between patients with and without bile reflux (ANOSIM, R=0.058, P=0.011). While the genera, Comamonas, Halomonas, Bradymonas, Pseudomonas, Marinobacter, Arthrobacter, and Shewanella were enriched in patients with bile reflux, the genera, Haemophilus, Porphyromonas, and Subdoligranulum, were enriched in those without bile reflux. CONCLUSION Our results demonstrate that bile reflux significantly alters the composition of the gastric microbiota.
Collapse
Affiliation(s)
| | | | | | | | - Yanbo Yu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | | | | |
Collapse
|
37
|
FOXD1 facilitates pancreatic cancer cell proliferation, invasion, and metastasis by regulating GLUT1-mediated aerobic glycolysis. Cell Death Dis 2022; 13:765. [PMID: 36057597 PMCID: PMC9440910 DOI: 10.1038/s41419-022-05213-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 01/21/2023]
Abstract
Although FOXD1 has been found to be involved in the malignant processes of several types of cancers, its role in pancreatic cancer (PC) is not well understood. This study aimed to investigate the expression and function of FOXD1 in PC. We found that FOXD1 mRNA and protein expression were upregulated in PC tissues compared with non-tumor tissues, and high expression level of FOXD1 was associated with an adverse prognostic index of PC. The results of in vitro and in vivo assays indicate that overexpression of FOXD1 promotes aerobic glycolysis and the capacity of PC cells to proliferate, invade, and metastasize, whereas FOXD1 knockdown inhibits these functions. The results of mechanistic experiments suggest that FOXD1 can not only directly promote SLC2A1 transcription but also inhibit the degradation of SLC2A1 through the RNA-induced silencing complex. As a result, FOXD1 enhances GLUT1 expression and ultimately facilitates PC cell proliferation, invasion, and metastasis by regulating aerobic glycolysis. Taken together, FOXD1 is suggested to be a potential therapeutic target for PC.
Collapse
|
38
|
Abstract
ABSTRACT Gastric intestinal metaplasia (GIM) is a precancerous lesion of gastric cancer (GC) and is considered an irreversible point of progression for GC. Helicobacter pylori infection can cause GIM, but its eradication still does not reverse the process. Bile reflux is also a pathogenic factor in GIM and can continuously irritate the gastric mucosa, and bile acids in refluxed fluid have been widely reported to be associated with GIM. This paper reviews in detail the relationship between bile reflux and GIM and the mechanisms by which bile acids induce GIM.
Collapse
|
39
|
Li S, Qu X, Zhang L, Wang N, Chen M, Zhao X, Wang J, Lv H, Qi Y, Zhang L, Liu J, Shi Y. Serum Total Bile Acids in Relation to Gastrointestinal Cancer Risk: A Retrospective Study. Front Oncol 2022; 12:859716. [PMID: 35756666 PMCID: PMC9213662 DOI: 10.3389/fonc.2022.859716] [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: 01/21/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background Bile acids (BAs) have been proposed to promote gastrointestinal cells carcinogenesis. However, studies on serum total bile acid (TBA) levels and gastrointestinal cancers (GICs) risk are rare. Methods We conducted a retrospective case-control study from 2015 to 2019 at the First Affiliated Hospital of Air Force Military Medical University, in which 4,256 GICs cases and 1,333 controls were recruited. Patients' demographic, clinical and laboratory data were collected. The odds ratios (ORs) with 95% confidence intervals (CIs) were estimated using binary logistic regression models. Results Positive associations were observed between serum TBA levels and risks of esophageal cancer (EC), gastric cancer (GC) and colorectal cancer (CRC). Overall, ORs of EC, GC and CRC risk rose with the TBA levels increasing. After adjustment for potential confounders, the OR of TBA-positive for EC risk was 4.89 (95% CI: 3.20-7.49), followed by GC (OR: 3.92, 95% CI: 2.53-6.08), and CRC (OR: 3.32, 95% CI: 2.04-5.11). Patients aged 60 years or older have a higher risk of GICs, especially for EC patients. Males are associated with a higher risk of GC, while females are associated with a higher risk of CRC. Preoperative serum TBA positive and negative was significantly different in the presence or absence of hematogenous metastasis among EC patients (P=0.014), and lymph node metastasis among GC patients (P=0.018). Conclusions This retrospective study showed positive associations between serum TBA level and GICs risk, and a higher serum TBA level constitutes a risk factor for GICs.
Collapse
Affiliation(s)
- Songbo Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiaodong Qu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Luyao Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Na Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Min Chen
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xingyu Zhao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jie Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China.,School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Huanhuan Lv
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China.,School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Ying Qi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China.,School of Clinical Medicine, Xi'an Medical University, Xi'an, China
| | - Lifeng Zhang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Junye Liu
- Department of Radiation Protective Medicine, School of Military Preventive Medicine, Air Force Medical University, Xi'an, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
40
|
Koide T, Koyanagi-Aoi M, Uehara K, Kakeji Y, Aoi T. CDX2-induced intestinal metaplasia in human gastric organoids derived from induced pluripotent stem cells. iScience 2022; 25:104314. [PMID: 35602937 PMCID: PMC9118752 DOI: 10.1016/j.isci.2022.104314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 03/14/2022] [Accepted: 04/25/2022] [Indexed: 11/03/2022] Open
Abstract
Intestinal metaplasia is related to gastric carcinogenesis. Previous studies have suggested the important role of CDX2 in intestinal metaplasia, and several reports have shown that the overexpression of CDX2 in mouse gastric mucosa caused intestinal metaplasia. However, no study has examined the induction of intestinal metaplasia using human gastric mucosa. In the present study, to produce an intestinal metaplasia model in human gastric mucosa in vitro, we differentiated human-induced pluripotent stem cells (hiPSC) to gastric organoids, followed by the overexpression of CDX2 using a tet-on system. The overexpression of CDX2 induced, although not completely, intestinal phenotypes and the enhanced expression of many, but not all, intestinal genes and previously reported intestinal metaplasia-related genes in the gastric organoids. This model can help clarify the mechanisms underlying intestinal metaplasia and carcinogenesis in human gastric mucosa and develop therapies to restitute precursor conditions of gastric cancer to normal mucosa.
Collapse
Affiliation(s)
- Takahiro Koide
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.,Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan.,Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Michiyo Koyanagi-Aoi
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.,Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan.,Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan
| | - Keiichiro Uehara
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.,Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan.,Department of Diagnostic Pathology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Takashi Aoi
- Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan.,Department of iPS Cell Applications, Graduate School of Medicine, Kobe University, Kobe, Japan.,Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan
| |
Collapse
|
41
|
Wang S, Kuang J, Zhang H, Chen W, Zheng X, Wang J, Huang F, Ge K, Li M, Zhao M, Rajani C, Zhu J, Zhao A, Jia W. Bile Acid-Microbiome Interaction Promotes Gastric Carcinogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200263. [PMID: 35285172 PMCID: PMC9165488 DOI: 10.1002/advs.202200263] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/21/2022] [Indexed: 05/11/2023]
Abstract
Bile reflux gastritis (BRG) is associated with the development of gastric cancer (GC), but the specific mechanism remains elusive. Here, a comprehensive study is conducted to explore the roles of refluxed bile acids (BAs) and microbiome in gastric carcinogenesis. The results show that conjugated BAs, interleukin 6 (IL-6), lipopolysaccharide (LPS), and the relative abundance of LPS-producing bacteria are increased significantly in the gastric juice of both BRG and GC patients. A secondary BA, taurodeoxycholic acid (TDCA), is significantly and positively correlated with the LPS-producing bacteria in the gastric juice of these patients. TDCA promotes the proliferation of normal gastric epithelial cells (GES-1) through activation of the IL-6/JAK1/STAT3 pathway. These results are further verified in two mouse models, one by gavage of TDCA, LPS, and LPS-producing bacteria (Prevotella melaninogenica), respectively, and the other by bile reflux (BR) surgery, mimicking clinical bile refluxing. Moreover, the bile reflux induced gastric precancerous lesions observed in the post BR surgery mice can be prevented by treatment with cryptotanshinone, a plant-derived STAT3 inhibitor. These results reveal an important underlying mechanism by which bile reflux promotes gastric carcinogenesis and provide an alternative strategy for the prevention of GC associated with BRG.
Collapse
Affiliation(s)
- Shouli Wang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Junliang Kuang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Hongwei Zhang
- Department of Metabolic and Bariatric SurgeryShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Wenlian Chen
- Cancer Institute, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghai200233China
| | - Xiaojiao Zheng
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Jieyi Wang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Fengjie Huang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Kun Ge
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Mengci Li
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Mingliang Zhao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Cynthia Rajani
- Cancer Biology ProgramUniversity of Hawaii Cancer CenterHonoluluHI96813USA
| | - Jinshui Zhu
- Department of GastroenterologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Aihua Zhao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
- Cancer Biology ProgramUniversity of Hawaii Cancer CenterHonoluluHI96813USA
- School of Chinese MedicineHong Kong Baptist UniversityKowloon TongHong Kong999077China
| |
Collapse
|
42
|
Liu H, Chen Y, Zhou L, Jiang X, Zhou X. MicroRNA-642b-3p functions as an oncomiR in gastric cancer by down-regulating the CUB and sushi multiple domains protein 1/smad axis. Bioengineered 2022; 13:9613-9627. [PMID: 35412956 PMCID: PMC9208452 DOI: 10.1080/21655979.2022.2056813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Aberrant expression of microRNAs (miRNAs or miRs) has been involved in the progression of gastric cancer (GC). Our analysis of GC-related gene expression profiles identified the significantly up-regulated miR-642b-3p expression, which has been reported as a mediator in various cancers but rarely mentioned in researches on GC. Herein, this study intends to investigate the role of miR-642b-3p in GC development. Bioinformatics analysis was conducted to predict the downstream target gene of miR-642b-3p. Expression patterns of miR-642b-3p and CUB and sushi multiple domains protein 1 (CSMD1) in GC tissues and cell lines was then determined. Immunofluorescence, wound healing and Transwell invasion assays were performed to observe the malignant behaviors of GC cells with altered expression of miR-642b-3p and CSMD1. Nude mice with xenograft tumors were developed for in vivo validation. miR-642b-3p expression was increased in GC tissues and cell lines. miR-642b-3p targeted CSMD1 and reduced the expression of CSMD1, thereby inhibiting the activation of Smad signaling pathway. By this mechanism, the epithelial–mesenchymal transition (EMT), invasive and migratory potentials of GC cells were repressed. Meanwhile, in vivo data verified that miR-642b-3p enhanced the tumor growth of GC cells, which was associated with blockade of CSMD1-dependent activation of the Smad signaling pathway. Overall, miR-642b-3p acts as an oncomiR promoting tumor development in GC through suppressing CSMD1 expression and inactivating the Smad signaling pathway, which may enable the development of new therapeutic strategies for treatment of GC.
Collapse
Affiliation(s)
- Haofeng Liu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou P.R. China.,Department of General Surgery, Tumor Hospital Affiliated to Nantong University & Nantong Tumor Hospital, Nantong P.R. China
| | - Yuan Chen
- Department of General Surgery, Tumor Hospital Affiliated to Nantong University & Nantong Tumor Hospital, Nantong P.R. China
| | - Linsen Zhou
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng P.R. China
| | - Xiaohui Jiang
- Department of General Surgery, Tumor Hospital Affiliated to Nantong University & Nantong Tumor Hospital, Nantong P.R. China
| | - Xiaojun Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou P.R. China
| |
Collapse
|
43
|
MiR-300 Alleviates Cell Proliferation and Migration and Facilitates Cell Apoptosis by Targeting c-Met in Gastric Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6167554. [PMID: 35419054 PMCID: PMC9001127 DOI: 10.1155/2022/6167554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/21/2022] [Accepted: 02/26/2022] [Indexed: 12/24/2022]
Abstract
c-Met is a potent oncogene, whose aberrant activation has not been fully clarified. In this study, we discover the biological function of miR-300 in gastric cancer (GC) carcinogenesis and the underlying mechanism. The overexpression, oncogenic functions, and survival analysis of c-Met in GC tissues and cells were firstly determined. miRNAs that potentially targets c-Met were then predicted by bioinformatics. The expression levels of candidate miR-300 in GC tissue pairs were investigated. Pearson analysis revealed a negative relation between miR-300 and c-Met expressions. miR-300 and c-Met expression levels were determined in three GC cell lines (MKN-45, SGC-7901, and AGS) as well. Reduced miR-300 led to increase c-Met levels. Luciferase report assay demonstrated a direct binding site of miR-300 in the 3' untranslated region (3′UTR) of c-Met. Finally, the regulatory role of miR-300 on MKN-45 cells was studied by cell proliferation, migration, and apoptosis assays. Overexpression of miR-300 attenuated viability and migration and accelerated apoptosis in MKN-45. We also induced a rescue experiment with c-Met overexpression plasmid and finally proved that miR-300 exerted a suppressing role on MKN-45 proliferation and migration but promoted MKN-45 apoptosis by directly inhibiting c-Met. This study provides a novel insight into the targeted drug development for GC therapies.
Collapse
|
44
|
He Q, Liu L, Wei J, Jiang J, Rong Z, Chen X, Zhao J, Jiang K. Roles and action mechanisms of bile acid-induced gastric intestinal metaplasia: a review. Cell Death Dis 2022; 8:158. [PMID: 35379788 PMCID: PMC8979943 DOI: 10.1038/s41420-022-00962-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 12/13/2022]
Abstract
Gastric intestinal metaplasia (IM) is a precancerous lesion that increases the risk of subsequent gastric cancer (GC) development. Therefore, the mechanism of IM has been the focus of basic and clinical research. Helicobacter pylori (H. pylori) infection has been recognized as the main pathogenesis of gastric IM. However, more and more studies have shown that chronic inflammation of gastric mucosa caused by bile reflux is the key pathogenic factor of gastric IM. Bile reflux activates the expression of IM biomarkers via the bile acid receptor. In addition, microRNAs, exosomes, and epigenetics are also involved in the occurrence and development of bile acid-induced gastric IM. Currently, the relevant research is still very few. The molecular mechanism of the phenotypic transformation of gastrointestinal epithelial cells induced by bile acids has not been fully understood. This article mainly reviews the physiology and pathology of bile acid, mechanism of gastric IM induced by bile acid, bile acid receptors, and so on, in order to provide reference for further research.
Collapse
Affiliation(s)
- Qijin He
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China
| | - Limin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China
| | - Jingge Wei
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China
| | - Jiaying Jiang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China
| | - Zheng Rong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China.
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China.
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, No. 154 Anshan Road, Tianjin, 300052, China.
| |
Collapse
|
45
|
Fan S, Huang Y, Lu G, Sun N, Wang R, Lu C, Ding L, Han J, Zhou J, Li Y, Ming T, Su X. Novel anti-hyperuricemic hexapeptides derived from Apostichopus japonicus hydrolysate and their modulation effects on the gut microbiota and host microRNA profile. Food Funct 2022; 13:3865-3878. [PMID: 35274663 DOI: 10.1039/d1fo03981d] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hyperuricemia (HUA) is the second most common metabolic disease nowadays, and is characterized by permanently increased concentrations of serum uric acid. In this study, two novel hexapeptides (GPAGPR and GPSGRP) were identified from Apostichopus japonicus hydrolysate and predicted to have xanthine oxidase (XOD) inhibitory activity by molecular docking. Their in vitro XOD inhibition rates reached 37.3% and 48.6%, respectively, at a concentration of 40 mg mL-1. Subsequently, in vivo experiments were carried out in a HUA mouse model, and we found that both peptides reduced the serum uric acid by inhibiting uric acid biosynthesis and reabsorption, as well as alleviated renal inflammation via suppressing the activation of the NLRP3 inflammasome. 16S rDNA sequencing indicated that both peptide treatments reduced the richness and diversity of the gut microbiota, altered the composition in the phylum and genus levels, but different change trends were observed in the phylum Verrucomicrobia and genera Akkermansia, Dubosiella, Alloprevotella, Clostridium unclassified and Alistipes. In addition, changes in the renal microRNA (miRNA) profiles induced by GPSGRP treatment were analyzed; 21 differentially expressed (DE) miRNAs were identified among groups, and KEGG pathway analysis indicated that their potential target genes were involved in pluripotency of stem cell regulation, mTOR signaling pathway and proteoglycans. Moreover, ten miRNAs involved in the HUA onset and alleviation were identified, which showed a high correlation with genera related to the metabolism of short-chain fatty acids, bile acids and tryptophan. This study delineated two hexapeptides as potential microbiota modulators and miRNA regulators that can ameliorate HUA.
Collapse
Affiliation(s)
- Siqing Fan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
| | - Yumeng Huang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,School of Marine Science, Ningbo University, Ningbo, China
| | - Guoding Lu
- Ningbo Green-Health Pharmaceutical Co., Ltd, Ningbo, China
| | - Na Sun
- Ningbo Green-Health Pharmaceutical Co., Ltd, Ningbo, China
| | - Rui Wang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
| | - Chenyang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,School of Marine Science, Ningbo University, Ningbo, China
| | - Lijian Ding
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
| | - Jiaojiao Han
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,School of Marine Science, Ningbo University, Ningbo, China
| | - Jun Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,School of Marine Science, Ningbo University, Ningbo, China
| | - Ye Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,School of Marine Science, Ningbo University, Ningbo, China
| | - Tinghong Ming
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,School of Marine Science, Ningbo University, Ningbo, China
| | - Xiurong Su
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China. .,School of Marine Science, Ningbo University, Ningbo, China
| |
Collapse
|
46
|
Li J, Xie B, Wang H, Chen C, Pan C, Jia J. Research on Function of Exosome of miR-328-3p Secreted by Bone Marrow Mesenchymal Stem Cells (BMSCs) on Restraining the Gastric Cancer Through Being Down-Regulated with Trefoil Factor 3. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Certain progress has been made in the therapeutic method against gastric cancer such as surgical operation combined with chemotherapy and radiation therapy in recent years. But the therapeutic efficacy and prognosis on gastric cancer was still not satisfactory. The function of exosome
of miR-328–3p secreted by bone marrow stromal cells (BMSCs) on restraining the gastric cancer was studied in the present study. The BMSCs with highly-expressed miR-328-3p was established. The exosome in cell supernatant was collected. The exosome of BMSCs and MSCs with highlyexpressed
miR-328-3p was added into SGC-7901 cells followed by analysis of miR-328-3p level by Real-time PCR and TFF3 (Trefoil Factor 3) level in exosome by Western blot, cell proliferation, expression of E-cadherin, Vimentin and Caspase-3. miR-328-39 expression was reduced and TFF3 was elevated in
gastric cancer tissue (P < 0.05). miR-328-3p was upregulated and TFF3 was downregulated after addition of BMSCs exosomes along with increased cell proliferation and reduced E-cadherin and Caspase3 expression (P < 0.05). In conclusion, exosome of BMSCs could be regulated
by miR-328-3p and TFF3 expression is restrained so as to regulate the biological behaviors of gastric cancer cell.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Bo Xie
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Hu Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Chengsong Chen
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Chengwu Pan
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Jianguang Jia
- Department of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| |
Collapse
|
47
|
Xie D, Sun MY. Mechanism of action of NF-κB related cell signaling pathways in progression of gastritis to carcinoma. Shijie Huaren Xiaohua Zazhi 2022; 30:255-259. [DOI: 10.11569/wcjd.v30.i6.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a major global public health problem. The evolvement pattern of "superficial gastritis-chronic atrophic gastritis-intestinal metaplasia-dysplasia-gastric carcinoma" is common in related gastric diseases. As a key factor involved in systemic stress response, inflammatory response, and apoptosis, the regulation of NF-κB related to inflammation and apoptosis is a necessary link between inflammation and cancer progression. NF-κB is activated in most solid tumors and lymphomas. In the critical mechanism of gastric cancer induced by gastritis with various etiologies, the upstream and downstream molecules in the NF-κB signaling pathway are changed, and the cells are exposed to the microenvironment of inflammatory response for a long time, which ultimately leads to the development of their carcinogenic potential. This article discusses the mechanism of NF-κB in the key risk factors for the progression of gastric disease.
Collapse
Affiliation(s)
- Dong Xie
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Chinese Medicine, Shanghai 201203, China
| | - Ming-Yu Sun
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
48
|
Zhang L, Wang N, Chen M, Wu S, Zeng J, Zhou F, Wu Q, Liu J, Shi Y. HDAC6/FOXP3/HNF4α axis promotes bile acids induced gastric intestinal metaplasia. Am J Cancer Res 2022; 12:1409-1422. [PMID: 35411233 PMCID: PMC8984877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/05/2022] [Indexed: 06/14/2023] Open
Abstract
Bile reflux is one of the main causes of gastric intestinal metaplasia (IM) which is an important precancerous lesion. Our previous study has shown that ectopic expression of Histone deacetylase 6 (HDAC6) promotes the activation of intestinal markers in bile acids (BA) induced gastric IM cells; however, the mechanism underlying how HDAC6-mediated epigenetic modifications regulate intestinal markers is not clear. In this study, we aimed to investigate the downstream targets of HDAC6 and the underlying mechanism in the process of BA induced gastric IM. We demonstrated that deoxycholic acid (DCA) upregulated HDAC6 in gastric cells, which further inhibited the transcription of Forkhead box protein 3 (FOXP3). Then, FOXP3 transcriptionally inhibited Hepatocyte nuclear factor 4α (HNF4α), which further inhibits the expression of downstream intestinal markers. These molecules have been shown to be clinically relevant, as FOXP3 levels were negatively correlated with HDAC6 and HNF4α in IM tissues. Transgenic mice experiments confirmed that HNF4α overexpression combined with DCA treatment induced gastric mucosa to secrete intestinal mucus and caused an abnormal mucosal structure. Our findings suggest that HDAC6 reduces FOXP3 through epigenetic modification, thus forming a closed loop HDAC6/FOXP3/HNF4α to promote gastric IM. Inhibition of HDAC6 may be a potential approach to prevent gastric IM in patients with bile reflux.
Collapse
Affiliation(s)
- Luyao Zhang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Na Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Min Chen
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Siran Wu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Jiaoxia Zeng
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Fenli Zhou
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Qiong Wu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Junye Liu
- Department of Radiation Protective Medicine, Fourth Military Medical UniversityXi’an, Shaanxi, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical UniversityXi’an, Shaanxi, China
| |
Collapse
|
49
|
Tang SY, Zhou PJ, Meng Y, Zeng FR, Deng GT. Gastric cancer: An epigenetic view. World J Gastrointest Oncol 2022; 14:90-109. [PMID: 35116105 PMCID: PMC8790429 DOI: 10.4251/wjgo.v14.i1.90] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/17/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) poses a serious threat worldwide with unfavorable prognosis mainly due to late diagnosis and limited therapies. Therefore, precise molecular classification and search for potential targets are required for diagnosis and treatment, as GC is complicated and heterogeneous in nature. Accumulating evidence indicates that epigenetics plays a vital role in gastric carcinogenesis and progression, including histone modifications, DNA methylation and non-coding RNAs. Epigenetic biomarkers and drugs are currently under intensive evaluations to ensure efficient clinical utility in GC. In this review, key epigenetic alterations and related functions and mechanisms are summarized in GC. We focus on integration of existing epigenetic findings in GC for the bench-to-bedside translation of some pivotal epigenetic alterations into clinical practice and also describe the vacant field waiting for investigation.
Collapse
Affiliation(s)
- Si-Yuan Tang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Pei-Jun Zhou
- Cancer Research Institute, School of Basic Medicine Science, Central South University, School of Basic Medicine Science, Central South University 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Fu-Rong Zeng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guang-Tong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
50
|
Chen L, Zhu G, She L, Ding Y, Yang C, Zhu F. Analysis of Risk Factors and Establishment of a Prediction Model for Endoscopic Primary Bile Reflux: A Single-Center Retrospective Study. Front Med (Lausanne) 2021; 8:758771. [PMID: 34859013 PMCID: PMC8631358 DOI: 10.3389/fmed.2021.758771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/07/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Endoscopic primary bile reflux is one of the main diagnostic criteria for bile reflux gastritis (BRG). Presently, the risk factors and prediction models of endoscopic primary bile reflux (EPBR) in gastropathy patients who cannot or will not undergo endoscopy due to contraindications are not clear. Thus, this study aimed to evaluate the risk factors of EPBR and to establish and verify a prediction model. Methods: A series of 844 patients (564 subjects with EPBR and 280 control subjects) were retrospectively selected for this study and divided into a training set (n = 591) and a validation set (n = 253) according to the usual ratio of 70:30% for the subsequent internal validation of the logistic regression model for EPBR. Fifteen parameters that might affect the occurrence of EPBR were collected. Subsequently, univariate and stepwise logistic regression analyses were introduced to reveal the risk factors and the multivariate prediction model. An R package was dedicated to the corresponding internal validation of the EPBR model. Results: The univariate analysis showed that gender, age, smoking, Helicobacter pylori (H. pylori) infection status, metabolic syndrome (MS), non-steroidal anti-inflammatory drugs (NSAIDs) use history, and previous medical histories of chronic liver diseases, cholelithiasis, and erosive gastritis were statistically significant between the two groups (P < 0.05). Multivariate regression described that being a male [OR (95%confidence interval (CI)) = 2.29 (1.50–3.50), P < 0.001], age≥45 years old [OR (95% CI) = 4.24 (2.59–6.96), P < 0.001], H. pylori infection status [OR (95% CI) = 2.34 (1.37–4.01), P = 0.002], MS [OR (95% CI) = 3.14 (1.77–5.54), P < 0.001], NSAIDs use history [OR (95% CI) = 1.87 (1.03–3.40), P = 0.04], cholelithiasis history [OR (95% CI) = 3.95 (2.18–7.18), P < 0.001] and erosive gastritis history [OR (95% CI) = 6.77 (3.73–12.29), P < 0.001] were the risk factors for the occurrence of EPBR. Based on the results of these risk factors, an EPBR prediction model with an adequate calibration and excellent discrimination was established [area under the curve (AUC): 0.839, 95% CI = 0.806–0.872]. Conclusions: Being a male, age ≥ 45 years old, H. pylori infection, histories of MS, NSAIDs use, cholelithiasis, and erosive gastritis appear to be the risk factors for EPBR, and our favorable prediction model might be an option for the prediction of EPBR.
Collapse
Affiliation(s)
- Li Chen
- Department of Gastroenterology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China.,State Key Laboratory of Pathogenesis, Prevention, and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Ürümqi, China.,Department of Gastroenterology, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Guoying Zhu
- Department of Clinical Nutrition, School of Medicine, Putuo People's Hospital, Tongji University, Shanghai, China
| | - Ling She
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Ürümqi, China.,Department of Gastroenterology, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Yongnian Ding
- State Key Laboratory of Pathogenesis, Prevention, and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Ürümqi, China.,Department of Gastroenterology, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Changqing Yang
- Department of Gastroenterology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Fengshang Zhu
- Department of Gastroenterology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China.,State Key Laboratory of Pathogenesis, Prevention, and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Ürümqi, China.,Department of Gastroenterology, Second Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| |
Collapse
|