1
|
Xia K, Zhou Y, Wang W, Cai Y. Streptococcus anginosus: the potential role in the progression of gastric cancer. J Cancer Res Clin Oncol 2025; 151:143. [PMID: 40252119 PMCID: PMC12009222 DOI: 10.1007/s00432-025-06201-1] [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: 03/13/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
Gastric cancer (GC) is among the most common and aggressive malignancies worldwide, characterized by a poor prognosis. Research on its pathogenesis and progression continues to evolve. Streptococcus anginosus (S. anginosus, SA) is a Gram-positive coccus commonly found in the oral cavity and upper respiratory tract, serving as a commensal bacterium in the oral, gastrointestinal, and genitourinary tracts. It is frequently associated with abscess formation in various organs and tissues, as well as other purulent infections. In recent years, S. anginosus has gained increasing attention for its role in GC progression, potentially leading to chronic gastric inflammation and precancerous lesions, and ultimately promoting the development of GC. Emerging evidence indicates a strong association between S. anginosus and the malignant progression and unfavorable prognosis of GC. This review summarizes the role and underlying mechanisms of S. anginosus in GC and proposes that S. anginosus plays a pivotal role in its initiation and progression, underscoring its potential therapeutic significance.
Collapse
Affiliation(s)
- Kun Xia
- Department of General Surgery, People's Hospital of Ningxiang City, 209, North Road of 1st Ring, Ningxiang, Hunan, 410600, P. R. China
| | - Yaoxiang Zhou
- Department of General Surgery, People's Hospital of Ningxiang City, 209, North Road of 1st Ring, Ningxiang, Hunan, 410600, P. R. China
| | - Wei Wang
- Department of General Surgery, People's Hospital of Ningxiang City, 209, North Road of 1st Ring, Ningxiang, Hunan, 410600, P. R. China
| | - Yinzhong Cai
- Department of General Surgery, People's Hospital of Ningxiang City, 209, North Road of 1st Ring, Ningxiang, Hunan, 410600, P. R. China.
| |
Collapse
|
2
|
Xiong Y, Shan S, Fu X, Zhao W, Han Y, Xu Y, Qu Y, Sun X, Lu S, Guo J, Lu W. Multi-omics analysis of the protective effects of Platycodon grandiflorum -derived inulin-type fructan against low-concentration PM 2.5-induced lung microenvironment changes in rats. Int J Biol Macromol 2025:142484. [PMID: 40220836 DOI: 10.1016/j.ijbiomac.2025.142484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 03/15/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025]
Abstract
In northern China, haze events frequently occur during winter, and PM2.5 is recognized as the most significant particulate matter in haze, posing a major threat to human health. Therefore, we employed a PM2.5 inhalation exposure system to investigate the protective effects of Platycodon grandiflorum inulin-type fructan (PGPI-1-a) on low-concentration PM2.5-induced lung microenvironment changes. Our findings revealed that long-term (4-month) PM2.5 exposure did not cause apparent pathological alterations in rat lungs but induced lung inflammation, which was alleviated by PGPI-1-a intervention. Multi-omics analysis demonstrated that PGPI-1-a restored abnormally expressed lung proteins, improved lung microbiota disorders, and regulated serum metabolite imbalances related to lipid and amino acid metabolism, ameliorating low-concentration PM2.5-induced lung microenvironment changes. These results suggest that Platycodon grandiflorum inulin-type fructan could serve as a potential dietary supplement for mitigating PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Yi Xiong
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Shan Shan
- Food Processing Research Institute, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xinjing Fu
- Chinese Acad Med Sci, Inst Lab Anim Sci, Beijing Key Lab Anim Models Emerging & Remerging, Key Lab Human Dis Comparat Med, Chinese Minist Hlt, Beijing, China
| | - Wenjie Zhao
- Chinese Acad Med Sci, Inst Lab Anim Sci, Beijing Key Lab Anim Models Emerging & Remerging, Key Lab Human Dis Comparat Med, Chinese Minist Hlt, Beijing, China
| | - Yunlin Han
- Chinese Acad Med Sci, Inst Lab Anim Sci, Beijing Key Lab Anim Models Emerging & Remerging, Key Lab Human Dis Comparat Med, Chinese Minist Hlt, Beijing, China
| | - Yanfeng Xu
- Chinese Acad Med Sci, Inst Lab Anim Sci, Beijing Key Lab Anim Models Emerging & Remerging, Key Lab Human Dis Comparat Med, Chinese Minist Hlt, Beijing, China
| | - Yajin Qu
- Chinese Acad Med Sci, Inst Lab Anim Sci, Beijing Key Lab Anim Models Emerging & Remerging, Key Lab Human Dis Comparat Med, Chinese Minist Hlt, Beijing, China
| | - Xiuping Sun
- Chinese Acad Med Sci, Inst Lab Anim Sci, Beijing Key Lab Anim Models Emerging & Remerging, Key Lab Human Dis Comparat Med, Chinese Minist Hlt, Beijing, China
| | - Shuwen Lu
- Food Processing Research Institute, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Jianguo Guo
- Chinese Acad Med Sci, Inst Lab Anim Sci, Beijing Key Lab Anim Models Emerging & Remerging, Key Lab Human Dis Comparat Med, Chinese Minist Hlt, Beijing, China.
| | - Weihong Lu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China.
| |
Collapse
|
3
|
Lin L, Li W, Yan L, Guo X, Zhuang M, Chen F, Ye W. Comparison of gastric microbiota in patients with different gastric lesions in high and low risk areas of gastric cancer. BMC Microbiol 2025; 25:202. [PMID: 40205356 PMCID: PMC11984132 DOI: 10.1186/s12866-025-03926-4] [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: 08/17/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Variation in gastric cancer (GC) incidence across different geographic areas persists, even when there are similar prevalence rates of Helicobacter pylori (H. pylori) infection. An extensive examination of the gastric microbiota in populations from both high- and low-risk regions of GC could help explain the geographical disparities in GC incidence. METHODS This study enrolled a total of 130 patients with superficial gastritis (SG) and precancerous lesions of gastric cancer (PLGC) from a high-risk area for GC and 205 patients from a low-risk area. Gastric microbial profiling was performed using 16 S rRNA gene sequencing to analyze differences in microbial composition between regions and lesion types. RESULTS The study revealed significant differences in gastric microbial profiles between patients from high- and low-risk regions, particularly in PLGC patients. PLGC patients from the low-risk region exhibited higher microbial richness than those from the high-risk area, with marked distinctions in microbial community structure between the two regions. Specific differences in microbial composition were observed at the phylum and genus levels between different regions. Six bacterial genera, including Selenomonas and Peptostreptococcus, were identified as enriched in PLGC patients from the high-risk area. Additionally, there was a noticeable imbalance in the microbiota of the gastric mucosal lining during the progression of gastric lesions. CONCLUSION This comparative analysis highlights the potential impact of the gastric microbiome in the development of GC and suggests that regional differences in microbial profiles may provide clues to the varying incidence rates of GC.
Collapse
Affiliation(s)
- Liying Lin
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Wanxin Li
- Department of Epidemiology and Health Statistics & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Lingjun Yan
- Department of Neurosurgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Department of Neurosurgery, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Xiaoxiong Guo
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Mingkai Zhuang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Fenglin Chen
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| | - Weimin Ye
- Department of Epidemiology and Health Statistics & Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 17177, Sweden.
| |
Collapse
|
4
|
Ge Y, Janson V, Dong Z, Liu H. Role and mechanism of IL-33 in bacteria infection related gastric cancer continuum: From inflammation to tumor progression. Biochim Biophys Acta Rev Cancer 2025; 1880:189296. [PMID: 40058506 DOI: 10.1016/j.bbcan.2025.189296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Gastric cancer, a globally prevalent malignant tumor, is characterized by low early diagnosis rate, high metastasis rate, and poor prognosis, particularly in East Asia, Eastern Europe, and South America. Helicobacter pylori (H. pylori) is recognized as the primary risk factor for gastric cancer. However, the fact that fewer than 3 % of infected individuals develop cancer suggests that other bacteria may also influence gastric carcinogenesis. A diverse community of microorganisms may interact with H. pylori, thereby driving disease progression. Here, the role of the cytokine IL-33, a member of the IL-1 family, is scrutinized. Its production can be induced by H. pylori through the activation of specific signaling pathways, and it contributes to the inflammatory environment by promoting the release of pro-inflammatory cytokines. This article reviews the conflicting evidence regarding IL-33's role in the progression from gastritis to gastric cancer and discusses the potential therapeutic implications of targeting the IL-33/ST2 axis, with various antibodies and inhibitors in development or undergoing clinical trials for inflammatory diseases. However, the role of IL-33 in gastric cancer treatment remains to be fully elucidated, with its effects potentially dependent on the cellular context and stage of cancer progression. In summary, this review provides a comprehensive overview of the intricate relationship between gastric microbiota, IL-33, and gastritis - gastric cancer transition, offering insights into potential therapeutic targets and the development of novel treatment strategies.
Collapse
Affiliation(s)
- Yunxiao Ge
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Victor Janson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China
| | - Hui Liu
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, Henan 450008, China.
| |
Collapse
|
5
|
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
|
6
|
Yan Y, Satoh-Takayama N. New perspectives on gastric disorders: the relationship between innate lymphoid cells and microbes in the stomach. Cell Mol Life Sci 2025; 82:113. [PMID: 40074935 PMCID: PMC11904066 DOI: 10.1007/s00018-025-05632-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/28/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025]
Abstract
A growing number of studies in recent years have revealed the changes in the gastric microbiota during the development of gastric diseases, breaking the stereotype that the stomach is hostile to microorganisms beyond H. pylori. After a decade of intensive research, the discovery of innate lymphoid cells (ILCs) has provided a new perspective on the immune response in many diseases. In the context of defense against infectious pathogens, the pre-existing innate defense mechanism of tissue-resident ILCs can rapidly recognize and respond to microbes to eliminate infection at the earliest stages. Here, we outline the basic function of ILCs in the gastric mucosa and in shaping the gastric microbiome. We discuss the interactions between the gastric microbiota and ILCs, explaining how the ILCs actively drive the immune response against bacterial pathogens that can lead to the development of the gastric disease.
Collapse
Affiliation(s)
- Yunzi Yan
- Precision Immune Regulation RIKEN ECL Research Unit, Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Naoko Satoh-Takayama
- Precision Immune Regulation RIKEN ECL Research Unit, Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan.
- Immunobiology Laboratory, Graduate School of Medical Life Sciences, Yokohama City University, Yokohama, Kanagawa, Japan.
| |
Collapse
|
7
|
Wang Y, Qiu X, Chu A, Chen J, Wang L, Sun X, Wang B, Yuan Y, Gong Y. Advances in 16S rRNA-Based Microbial Biomarkers for Gastric Cancer Diagnosis and Prognosis. Microb Biotechnol 2025; 18:e70115. [PMID: 39992270 PMCID: PMC11849407 DOI: 10.1111/1751-7915.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025] Open
Abstract
Gastric cancer (GC) is a malignant tumour with high morbidity and mortality worldwide, and there is an urgent need for early diagnosis and precision treatment. In recent years, the role of microbiota in the occurrence and development of GC has drawn extensive attention. Particularly, the in-depth study of GC-related microbiota by 16S rRNA sequencing technology has offered valuable tools and new perspectives for exploring the microbial characteristics of GC patients. This review systematically summarises the microbial diversity and composition of GC and non-GC samples based on 16S rRNA data, outlines the progress in identifying GC-related microbial biomarkers, explores the potential mechanisms by which diagnostic microbial biomarkers influence the development of GC, and reflects on the limitations of present studies. By integrating the current evidence, this review intends to offer a new theoretical foundation and further direction for the clinical translation of microbiota research in the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Yingying Wang
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Xunan Qiu
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Aining Chu
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Jijun Chen
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Lu Wang
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Xiaohu Sun
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Bengang Wang
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| | - Yuehua Gong
- Tumor Etiology and Screening Department of Cancer Institute and General SurgeryThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education DepartmentThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
8
|
Ye Y, Bin B, Chen P, Chen J, Meng A, Yu L, Yang F, Cui H. Advances in the study of the role of gastric microbiota in the progression of gastric cancer. Microb Pathog 2025; 199:107240. [PMID: 39708981 DOI: 10.1016/j.micpath.2024.107240] [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: 07/10/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Gastric cancer (GC) is a common malignant tumor and the third most common cancer in China in terms of mortality. Stomach microorganisms play complex roles in the development of GC. The carcinogenic mechanism of Helicobacter pylori has been elucidated, and there is much evidence that other microorganisms in the gastric mucosa are also heavily involved in the disease progression of this cancer. However, their carcinogenic mechanisms have not yet been fully elucidated. The microbial compositions associated with the normal stomach, precancerous lesions, and GC are distinctly different and have a complex evolutionary mechanism. The dysregulation of gastric microbiota may play a key role in the oncogenic process from precancerous lesions to malignant gastric tumors. In this review, we explore the potential translational and clinical implications of intragastric microbes in the diagnosis, prognosis, and treatment of GC. Finally, we summarize the research dilemmas and solutions concerning intragastric microbes, emphasizing that they should be at the forefront of strategies for GC prevention and treatment.
Collapse
Affiliation(s)
- Yu Ye
- Inner Mongolia Medical University, No 60, Xi Lin Guo Le South Road, Hohhot, 010020, Inner Mongolia Autonomous Region, PR China
| | - Ba Bin
- Department of Oncology, Ordos Hospital of Traditional Chinese Medicine, No 5, Yongning Street, Kangbashi District, Ordos City, Inner Mongolia Autonomous Region, PR China
| | - Pengfei Chen
- The Affiliated Hospital of Inner Mongolia Medical University, PR China
| | - Jing Chen
- Medical Department of Ordos College of Applied Technology, PR China
| | - Aruna Meng
- Inner Mongolia Medical University, No 60, Xi Lin Guo Le South Road, Hohhot, 010020, Inner Mongolia Autonomous Region, PR China
| | - Lei Yu
- Department of Pharmacy, Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region, 010020, PR China
| | - Fan Yang
- Inner Mongolia Autonomous Region Blood Central, PR China.
| | - Hongwei Cui
- Peking University Cancer Hospital (Inner Mongolia Campus) & Affiliated Cancer Hospital of Inner Mongolia Medical University, No 42, Zhao Wu Da Road, Hohhot, 010020, Inner Mongolia Autonomous Region, PR China.
| |
Collapse
|
9
|
Wang B, Luan J, Zhao W, Yu J, Li A, Li X, Zhong X, Cao H, Wang R, Liu B, Lu S, Shi M. Comprehensive multiomics analysis of the signatures of gastric mucosal bacteria and plasma metabolites across different stomach microhabitats in the development of gastric cancer. Cell Oncol (Dordr) 2025; 48:139-159. [PMID: 38963518 PMCID: PMC11850404 DOI: 10.1007/s13402-024-00965-3] [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] [Accepted: 05/30/2024] [Indexed: 07/05/2024] Open
Abstract
PURPOSE As an important component of the microenvironment, the gastric microbiota and its metabolites are associated with tumour occurrence, progression, and metastasis. However, the relationship between the gastric microbiota and the development of gastric cancer is unclear. The present study investigated the role of the gastric mucosa microbiome and metabolites as aetiological factors in gastric carcinogenesis. METHODS Gastric biopsies from different stomach microhabitats (n = 70) were subjected to 16S rRNA gene sequencing, and blood samples (n = 95) were subjected to untargeted metabolome (gas chromatography‒mass spectrometry, GC‒MS) analyses. The datasets were analysed using various bioinformatics approaches. RESULTS The microbiota diversity and community composition markedly changed during gastric carcinogenesis. High Helicobacter. pylori colonization modified the overall diversity and composition of the microbiota associated with gastritis and cancer in the stomach. Most importantly, analysis of the functional features of the microbiota revealed that nitrate reductase genes were significantly enriched in the tumoral microbiota, while urease-producing genes were significantly enriched in the microbiota of H. pylori-positive patients. A panel of 81 metabolites was constructed to discriminate gastric cancer patients from gastritis patients, and a panel of 15 metabolites was constructed to discriminate H. pylori-positive patients from H. pylori-negative patients. receiver operator characteristic (ROC) curve analysis identified a series of gastric microbes and plasma metabolites as potential biomarkers of gastric cancer. CONCLUSION The present study identified a series of signatures that may play important roles in gastric carcinogenesis and have the potential to be used as biomarkers for diagnosis and for the surveillance of gastric cancer patients with minimal invasiveness.
Collapse
Affiliation(s)
- Bingsen Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
| | - Jiahui Luan
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, 255400, China
| | - Weidong Zhao
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Junbao Yu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
| | - Anqing Li
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Xinxin Li
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Xiaoqin Zhong
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of gastroenterology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Hongyun Cao
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, 255400, China
| | - Ruicai Wang
- Department of Pathology, Zibo Municipal Hospital, Zibo, 255400, China
| | - Bo Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
- Department of Clinical Microbiology, Zibo City Key Laboratory of Respiratory Infection and Clinical Microbiology, Zibo City Engineering Technology Research Center of Etiology Molecular Diagnosis, Zibo Municipal Hospital, Zibo, 255400, China
- Department of Pulmonary and Critical Care Medicine, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China
| | - Shiyong Lu
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China
| | - Mei Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
- Shandong University-Zibo Municipal Hospital Research Center of Human Microbiome and Health, Zibo, 255400, China.
| |
Collapse
|
10
|
Yang P, Liang G, Ni Y, Chu X, Zhang X, Wang Z, Khan A, Jin F, Shen H, Li M, Xu Z. Investigating the role of intratumoral Streptococcus mitis in gastric cancer progression: insights into tumor microenvironment. J Transl Med 2025; 23:126. [PMID: 39875915 PMCID: PMC11773703 DOI: 10.1186/s12967-025-06142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Growing evidence implicates that intratumoral microbiota are closely linked to cancer progression; however, research on the role of these microbiota in the development of gastric cancer remains limited. Here, using 16 S rRNA sequencing, tumor tissue proteomics and serum cytokines analysis, we identified enrichment of specific microbial communities within tumors of gastric cancer patients, possibly affecting the tumor microenvironment by immune modulation, metabolic processes, and inflammatory responses. Based on the results of in vivo experiments and intratumoral microbiota analysis, we found that Streptococcus mitis can inhibit gastric cancer progression via suppressing M2 macrophage polarization and infiltration, as well as altering the intratumoral microbial community. In summary, our findings suggest that the intratumoral microbiota, exemplified by Streptococcus mites, may be involved in regulating the progression of gastric cancer, thereby emerging as potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Ping Yang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Gaoli Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Yangyue Ni
- Department of Pathogen Biology, Key Laboratory for Pathogen Infection and Control of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, P.R. China
| | - Xiaojie Chu
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
| | - Xiaoshan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Physiology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, School of Life Sciences, NJU Advanced Institute of Life Sciences (NAILS), Nanjing University, Nanjing, 210008, Jiangsu, P. R. China
| | - Zhongyu Wang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China
| | - Adeel Khan
- Department of Biotechnology, University of Science and Technology Bannu, Bannu, 28100, KP, Pakistan
| | - Fangfang Jin
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China.
| | - Han Shen
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China.
| | - Miao Li
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, China.
| | - Zhipeng Xu
- Department of Pathogen Biology, Key Laboratory for Pathogen Infection and Control of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, P.R. China.
| |
Collapse
|
11
|
Chen Y, Lei L, Xia M, Cheng R, Cai H, Hu T. The association between oral microbiome and gastric precancerous lesions. mSystems 2025; 10:e0132224. [PMID: 39629992 PMCID: PMC11748542 DOI: 10.1128/msystems.01322-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Gastric precancerous lesions are thought to be precursors in the occurrence and development of gastric cancer through Correa's cascade. Recent studies have investigated the association between the oral microbiome and gastric precancerous lesions. However, there has yet to be a comprehensive synthesis review of the existing literature on the relationship between oral microbiome and gastric precancerous lesions. A systematic review was conducted to characterize the literature on the association between oral microbiome and gastric precancerous lesions. The studies show that oral microbiome is dynamic in individuals with gastric precancerous lesions. Oral-derived microorganisms were colonized in the gastric precancerous lesions. Interactions between oral and gastric microbiomes affect the response of the host immunity. The abnormal proliferation of oral-associated microorganisms may be linked to the reduction of gastric acid. The present review supports the potential association between oral microbiome and gastric precancerous lesions. However, the interactions are complex and multifaceted, which require further investigation.
Collapse
Affiliation(s)
- Yifei Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Mengying Xia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ran Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - He Cai
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Tao Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Al-Matouq J, Al-Ghafli H, Alibrahim NN, Alsaffar N, Radwan Z, Ali MD. Unveiling the Interplay Between the Human Microbiome and Gastric Cancer: A Review of the Complex Relationships and Therapeutic Avenues. Cancers (Basel) 2025; 17:226. [PMID: 39858007 PMCID: PMC11763844 DOI: 10.3390/cancers17020226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The human microbiota plays a crucial role in maintaining overall health and well-being. The gut microbiota has been implicated in developing and progressing various diseases, including cancer. This review highlights the related mechanisms and the compositions that influence cancer pathogenesis with a highlight on gastric cancer. We provide a comprehensive overview of the mechanisms by which the microbiome influences cancer development, progression, and response to treatment, with a focus on identifying potential biomarkers for early detection, prevention strategies, and novel therapeutic interventions that leverage microbiome modulation. This comprehensive review can guide future research and clinical practices in understanding and harnessing the microbiome to optimize gastric cancer therapies.
Collapse
Affiliation(s)
- Jenan Al-Matouq
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Hawra Al-Ghafli
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Noura N. Alibrahim
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Nida Alsaffar
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Zaheda Radwan
- Department of Medical Laboratory Sciences, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia; (H.A.-G.); (N.N.A.); (N.A.); (Z.R.)
| | - Mohammad Daud Ali
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Al Safa, Dammam 34222, Saudi Arabia;
| |
Collapse
|
13
|
Xia M, Lei L, Zhao L, Xu W, Zhang H, Li M, Hu J, Cheng R, Hu T. The dynamic oral-gastric microbial axis connects oral and gastric health: current evidence and disputes. NPJ Biofilms Microbiomes 2025; 11:1. [PMID: 39747247 PMCID: PMC11696714 DOI: 10.1038/s41522-024-00623-4] [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: 04/02/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025] Open
Abstract
Emerging evidence indicates that oral microbes are closely related to gastric microbes and gastric lesions, including gastric atrophy, intestinal metaplasia and gastric cancer (GC). Helicobacter pylori is a key pathogen involved in GC. However, the increasing prevalence of H. pylori-negative GC and gastric dysbiosis in GC patients emphasize the potential role of other microbial factors. In this review, we discussed the current evidence about the relationship between the oral-gastric microbial axis and oral and gastric health. Epidemiologic evidence indicates that poor oral hygiene is related to greater GC risk. Multiple oral-associated microbes are enriched in the stomach of GC patients. Once colonizing the stomach, oral-associated microbes Streptococcus anginosus and Prevotella melaninogenica, are involved in gastric inflammation or carcinogenesis. Microbial metabolites such as lactate, nitrite, and acetaldehyde promote malignant transformation. The stomach, as a checkpoint of microbial transmission in the digestive tract, is of great importance since the link between oral microbes and intestinal diseases has been emphasized. Still, new technologies and standardized metrics are necessary to identify potential pathogenetic microbes for GC and the core microbiota, interactions, richness, colonization, location and effect (CIRCLE). In the future, oral microbes could be candidates for noninvasive indicators to predict gastric diseases.
Collapse
Affiliation(s)
- Mengying Xia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Microbiology, ADA Forsyth Institute, Cambridge, USA
| | - Linyong Zhao
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenqing Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - Hongyu Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Mingming Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiankun Hu
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ran Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Tao Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Kuang W, Xu J, Xu F, Huang W, Majid M, Shi H, Yuan X, Ruan Y, Hu X. Current study of pathogenetic mechanisms and therapeutics of chronic atrophic gastritis: a comprehensive review. Front Cell Dev Biol 2024; 12:1513426. [PMID: 39720008 PMCID: PMC11666564 DOI: 10.3389/fcell.2024.1513426] [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: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Chronic atrophic gastritis (CAG) is a prevalent digestive system disease characterized by atrophy of the gastric mucosa and the disappearance of inherent gastric glands. According to the theory of Correa's cascade, CAG is an important pathological stage in the transformation from normal condition to gastric carcinoma. In recent years, the global incidence of CAG has been increasing due to pathogenic factors, including Helicobacter pylori infection, bile reflux, and the consumption of processed meats. In this review, we comprehensively described the etiology and clinical diagnosis of CAG. We focused on elucidating the regulatory mechanisms and promising therapeutic targets in CAG, with the expectation of providing insights and theoretical support for future research on CAG.
Collapse
Affiliation(s)
- Weihong Kuang
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jialin Xu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Fenting Xu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Weizhen Huang
- Cancer Center, The First Huizhou Affiliated Hospital, Guangdong Medical University, Huizhou, China
| | - Muhammad Majid
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Hui Shi
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
| | - Xia Yuan
- Cancer Center, The First Huizhou Affiliated Hospital, Guangdong Medical University, Huizhou, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
- Cancer Center, The First Huizhou Affiliated Hospital, Guangdong Medical University, Huizhou, China
| | - Xianjing Hu
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Traditional Chinese Medicines for Prevention and Treatment of Digestive Diseases, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Department of Acupuncture, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| |
Collapse
|
15
|
Li Y, He C, Lu N. Impacts of Helicobacter pylori infection and eradication on gastrointestinal microbiota: An up-to-date critical review and future perspectives. Chin Med J (Engl) 2024; 137:2833-2842. [PMID: 39501846 DOI: 10.1097/cm9.0000000000003348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT Helicobacter pylori ( H. pylori ) infects approximately half of the population worldwide and causes chronic gastritis, peptic ulcers, and gastric cancer. Test-and-treat strategies have been recommended for the prevention of H. pylori -associated diseases. Advancements in high-throughput sequencing technologies have broadened our understanding of the complex gastrointestinal (GI) microbiota and its role in maintaining host homeostasis. Recently, an increasing number of studies have indicated that the colonization of H. pylori induces dramatic alterations in the gastric microbiota, with a predominance of H. pylori and a reduction in microbial diversity. Dysbiosis of the gut microbiome has also been observed after H. pylori infection, which may play a role in the development of colorectal cancer. However, there is concern regarding the impact of antibiotics on the gut microbiota during H. pylori eradication. In this review, we summarize the current literature concerning how H. pylori infection reshapes the GI microbiota and the underlying mechanisms, including changes in the gastric environment, immune responses, and persistent inflammation. Additionally, the impacts of H. pylori eradication on GI microbial homeostasis and the use of probiotics as adjuvant therapy are also discussed. The shifts in the GI microbiota and their crosstalk with H. pylori may provide potential targets for H. pylori -related gastric diseases and extragastric manifestations.
Collapse
Affiliation(s)
- Yu Li
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
- HuanKui Academy, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Cong He
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Nonghua Lu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| |
Collapse
|
16
|
Zeng R, Sung JJY, Yu J. New pathogen for gastric cancer: Streptococcus anginosus. Clin Transl Med 2024; 14:e70104. [PMID: 39605236 PMCID: PMC11602752 DOI: 10.1002/ctm2.70104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Affiliation(s)
- Ruijie Zeng
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research InstituteThe Chinese University of Hong KongHong Kong SARChina
| | - Joseph J. Y. Sung
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research InstituteThe Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
17
|
Liu Z, Xu H, You W, Pan K, Li W. Helicobacter pylori eradication for primary prevention of gastric cancer: progresses and challenges. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:299-310. [PMID: 39735441 PMCID: PMC11674435 DOI: 10.1016/j.jncc.2024.06.006] [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: 04/26/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 12/31/2024] Open
Abstract
Gastric cancer remains a significant global health challenge, causing a substantial number of cancer-related deaths, particularly in China. While the exact causes of gastric cancer are still being investigated, Helicobacter pylori (H. pylori) infection has been identified as the primary risk factor, which triggers chronic inflammation and a multistage progression of gastric lesions that may lead to carcinogenesis over a long latency time. Since the 1990s, numerous efforts have focused on assessing the effectiveness of H. pylori eradication in preventing new cases of gastric cancer among both the general population and patients who have undergone early-stage cancer treatment. This body of work, including several community-based interventions and meta-analyses, has shown a reduction in both the incidence of and mortality from gastric cancer following H. pylori treatment, alongside a decreased risk of metachronous gastric cancer. In this review, we seek to consolidate current knowledge on the effects of H. pylori treatment on gastric cancer prevention, its systemic consequences, cost-effectiveness, and the influence of antibiotic resistance and host characteristics on treatment outcomes. We further discuss the potential for precision primary prevention of H. pylori treatment and comment on the efficient implementation of test-and-treat policies and allocation of health resources towards minimizing the burden of gastric cancer globally.
Collapse
Affiliation(s)
- Zongchao Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hengmin Xu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weicheng You
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kaifeng Pan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wenqing Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Cancer Epidemiology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
18
|
Yuan L, Pan L, Wang Y, Zhao J, Fang L, Zhou Y, Xia R, Ma Y, Jiang Z, Xu Z, Hu C, Wang Y, Zhang S, Zhang B, Ding H, Chen M, Cheng H, Goel A, Zhang Z, Cheng X. Characterization of the landscape of the intratumoral microbiota reveals that Streptococcus anginosus increases the risk of gastric cancer initiation and progression. Cell Discov 2024; 10:117. [PMID: 39587089 PMCID: PMC11589709 DOI: 10.1038/s41421-024-00746-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
As a critical component of the tumour immune microenvironment (TIME), the resident microbiota promotes tumorigenesis across a variety of cancer types. Here, we integrated multiple types of omics data, including microbiome, transcriptome, and metabolome data, to investigate the functional role of intratumoral bacteria in gastric cancer (GC). The microbiome was used to categorize GC samples into six subtypes, and patients with a high abundance of Streptococcus or Pseudomonas had a markedly worse prognosis. Further assays revealed that Streptococcus anginosus (SA) promoted tumour cell proliferation and metastasis while suppressing the differentiation and infiltration of CD8+ T cells. However, antibiotic treatment significantly suppressed tumorigenesis in SA+ mice in vivo. We further demonstrated that the SA arginine pathway increased the abundance of ornithine, which may be a major contributor to reshaping of the TIME. Our findings demonstrated that SA, a novel risk factor, plays significant roles in the initiation and progression of GC, suggesting that SA might be a promising target for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Li Yuan
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Libin Pan
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Yunzhe Wang
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Zhao
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Luo Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Ying Zhou
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Ruihong Xia
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yubo Ma
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Zhengchen Jiang
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yanan Wang
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Shengjie Zhang
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Bo Zhang
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Haiying Ding
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Mengxuan Chen
- Shanghai Analytical Applications Center, Shimadzu (China) Co., LTD, Shanghai, China
| | - Haibo Cheng
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Zhao Zhang
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Xiangdong Cheng
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
19
|
Zeng R, Gou H, Lau HCH, Yu J. Stomach microbiota in gastric cancer development and clinical implications. Gut 2024; 73:2062-2073. [PMID: 38886045 PMCID: PMC11672014 DOI: 10.1136/gutjnl-2024-332815] [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: 05/07/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Gastric cancer (GC) is one of the most common malignancies and a prominent cause of cancer mortality worldwide. A distinctive characteristic of GC is its intimate association with commensal microbial community. Although Helicobacter pylori is widely recognised as an inciting factor of the onset of gastric carcinogenesis, increasing evidence has indicated the substantial involvement of microbes that reside in the gastric mucosa during disease progression. In particular, dysregulation in gastric microbiota could play pivotal roles throughout the whole carcinogenic processes, from the development of precancerous lesions to gastric malignancy. Here, current understanding of the gastric microbiota in GC development is summarised. Potential translational and clinical implications of using gastric microbes for GC diagnosis, prognosis and therapeutics are also evaluated, with further discussion on conceptual haziness and limitations at present. Finally, we highlight that modulating microbes is a novel and promising frontier for the prevention and management of GC, which necessitates future in-depth investigations.
Collapse
Affiliation(s)
- Ruijie Zeng
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongyan Gou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Wong CC, Yu J. Redefining the Gastric Microbes in Promoting Gastric Tumorigenesis: The Rise of the Non-H. pylori Microbiome. Cancer Discov 2024; 14:2051-2054. [PMID: 39485255 DOI: 10.1158/2159-8290.cd-24-0835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 11/03/2024]
Abstract
Gastric cancer remains one of the top cancers in China compared with Western countries, mainly attributed to the high rates of Helicobacter pylori infection. However, recent discoveries on the non-H. pylori gastric microbiome have led to a paradigm shift in our understanding of microbial risk factors driving gastric cancer, which will impact future screening and prevention strategies.
Collapse
Affiliation(s)
- Chi Chun Wong
- State Key Laboratory of Digestive Disease, Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
21
|
Wu M, Tian C, Zou Z, Jin M, Liu H. Gastrointestinal Microbiota in Gastric Cancer: Potential Mechanisms and Clinical Applications-A Literature Review. Cancers (Basel) 2024; 16:3547. [PMID: 39456641 PMCID: PMC11506470 DOI: 10.3390/cancers16203547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Emerging evidence highlights the crucial role of gastrointestinal microbiota in the pathogenesis of gastric cancer. Helicobacter pylori (H. pylori) infection stands out as a primary pathogenic factor. However, interventions such as anti-H. pylori therapy, gastric surgeries, immunotherapy, and chronic inflammation significantly remodel the gastric microbiome, implicating a broader spectrum of microorganisms in cancer development. These microbial populations can modulate gastric carcinogenesis through various mechanisms, including sustained chronic inflammation, bacterial genotoxins, alterations in short-chain fatty acids, elevated gastrointestinal bile acids, impaired mucus barrier function, and increased concentrations of N-nitrosamines and lactic acid. The dynamic changes in gut microbiota also critically influence the outcomes of anti-cancer therapies by modifying drug bioavailability and metabolism, thus affecting therapeutic efficacy and side effect profiles. Additionally, the effectiveness of radiotherapy can be significantly impacted by gut microbiota alterations. Novel therapeutic strategies targeting the microbiome, such as dietary interventions, probiotic and synbiotic supplementation, and fecal microbiota transplantation, are showing promise in cancer treatment. Understanding the intricate relationship between the gut microbiota and gastric cancer is essential for developing new, evidence-based approaches to the prevention and treatment of this malignancy.
Collapse
Affiliation(s)
- Mengjiao Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenjun Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China;
| | - Zhenwei Zou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- The Eighth Hospital of Wuhan, Wuhan 430012, China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
22
|
Hou M, Chen J, Yang L, Qin L, Liu J, Zhao H, Guo Y, Yu QQ, Zhang Q. Identification of Fatty Acid Metabolism-Related Subtypes in Gastric Cancer Aided by Machine Learning. Cancer Manag Res 2024; 16:1463-1473. [PMID: 39439917 PMCID: PMC11495201 DOI: 10.2147/cmar.s483577] [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: 08/12/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Gastric cancer, the fifth most common malignant tumor in the world, poses a serious threat to human health. However, the role of fatty acid metabolism (FAM) in gastric cancer remains incompletely understood. We aim to provide guidance for clinical decisions by utilizing public database of gastric adenocarcinoma to establish an FAM-related gene subtypes via machine learning algorithm. METHODS The intersection of FMGs from KEGG, Hallmark, and Reactome bioinformatics databases and the DEGs of the TCGA-STAD cohort was used to decompose the gene matrix related to establish FAM-related gene subtypes by NMF. Comparison of immune infiltrating differences between subtypes using ESTIMATE and Cibersort algorithms. The multifactor Cox regression to identify independent risk genes for patient prognosis based on the subtypes. A prognostic model including independent risk genes was built using random survival forest and Cox regression. IHC validation in gastric cancer and adjacent tissues confirmed the above gene expression level. RESULTS 71 DEGs related to FMGs of STAD were identified, which was used to established the FAM-related gene subtypes, C1 and C2. The immune infiltrating analysis showed that most immune features of C2 were significantly upregulated compared to C1. The independent risk genes were CGβ8, UPK1B, and OR51G based on the subtypes. A gastric cancer prognostic model consisting of independent risk genes was constructed and patients were classified into high-risk and low-risk groups with survival differential analysis. Finally, IHC showed that CGβ8 and UPK1B expression were upregulated in gastric cancer, while OR51G2 did not detect differences in expression. CONCLUSION The study developed a machine learning-based gastric cancer prognosis risk model using FMGs. This model effectively stratifies patients according to their risk levels and provides valuable insights for clinical decision-making, enabling accurate evaluation of patient prognosis.
Collapse
Affiliation(s)
- Maolin Hou
- Department of Internal Medicine, Siziwangqi People’s Hospital, Wulancabu, 011800, People’s Republic of China
| | - Jinghua Chen
- Department of Oncology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, 250000, People’s Republic of China
| | - Le Yang
- Department of Gastrointestinal Surgery, Jining, 272000, People’s Republic of China
| | - Lei Qin
- Department of Gastrointestinal Surgery, Jining, 272000, People’s Republic of China
| | - Jie Liu
- Department of Pediatric Intensive Care Unit, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250000, People’s Republic of China
| | - Haibo Zhao
- Department of Oncology, Jining, 272000, People’s Republic of China
| | - Yujin Guo
- Department of Clinical Pharmacology, Jining, 272000, People’s Republic of China
| | - Qing-Qing Yu
- Department of Clinical Pharmacology, Jining, 272000, People’s Republic of China
| | - Qiujie Zhang
- Department of Oncology, Jining, 272000, People’s Republic of China
| |
Collapse
|
23
|
Qian ST, Zhao HY, Xie FF, Liu QS, Cai DL. Streptococcus anginosus in the development and treatment of precancerous lesions of gastric cancer. World J Gastrointest Oncol 2024; 16:3771-3780. [PMID: 39350992 PMCID: PMC11438778 DOI: 10.4251/wjgo.v16.i9.3771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/22/2024] [Accepted: 06/06/2024] [Indexed: 09/09/2024] Open
Abstract
The microbiota is strongly association with cancer. Studies have shown significant differences in the gastric microbiota between patients with gastric cancer (GC) patients and noncancer patients, suggesting that the microbiota may play a role in the development of GC. Although Helicobacter pylori (H. pylori) infection is widely recognized as a primary risk factor for GC, recent studies based on microbiota sequencing technology have revealed that non-H. pylori microbes also have a significant impact on GC. A recent study discovered that Streptococcus anginosus (S. anginosus) is more prevalent in the gastric mucosa of patients with GC than in that of those without GC. S. anginosus infection can spontaneously induce chronic gastritis, mural cell atrophy, mucoid chemotaxis, and heterotrophic hyperplasia, which promote the development of precancerous lesions of GC (PLGC). S. anginosus also disrupts the gastric barrier function, promotes the proliferation of GC cells, and inhibits apoptosis. However, S. anginosus is underrepresented in the literature. Recent reports suggest that it may cause precancerous lesions, indicating its emerging pathogenicity. Modern novel molecular diagnostic techniques, such as polymerase chain reaction, genetic testing, and Ultrasensitive Chromosomal Aneuploidy Detection, can be used to gastric precancerous lesions via microbial markers. Therefore, we present a concise summary of the relationship between S. anginosus and PLGC. Our aim was to further investigate new methods of preventing and treating PLGC by exploring the pathogenicity of S. anginosus on PLGC.
Collapse
Affiliation(s)
- Su-Ting Qian
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Hao-Yu Zhao
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Fei-Fei Xie
- Department of Digestive, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Qing-Sheng Liu
- Science and Education Section, Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, Zhejiang Province, China
| | - Dan-Li Cai
- Intensive Care Unit, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 311122, Zhejiang Province, China
| |
Collapse
|
24
|
Liang B, Deng Y, Huang Y, Zhong Y, Li Z, Du J, Ye R, Feng Y, Bai R, Fan B, Chen X, Huang X, Yang X, Xian H, Yang X, Huang Z. Fragile Guts Make Fragile Brains: Intestinal Epithelial Nrf2 Deficiency Exacerbates Neurotoxicity Induced by Polystyrene Nanoplastics. ACS NANO 2024; 18:24044-24059. [PMID: 39158845 DOI: 10.1021/acsnano.4c03874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Oral ingestion is the primary route for human exposure to nanoplastics, making the gastrointestinal tract one of the first and most impacted organs. Given the presence of the gut-brain axis, a crucial concern arises regarding the potential impact of intestinal damage on the neurotoxic effects of nanoplastics (NPs). The intricate mechanisms underlying NP-induced neurotoxicity through the microbiome-gut-brain axis necessitate further investigation. To address this, we used mice specifically engineered with nuclear factor erythroid-derived 2-related factor 2 (Nrf2) deficiency in their intestines, a strain whose intestines are particularly susceptible to polystyrene NPs (PS-NPs). We conducted a 28-day repeated-dose oral toxicity study with 2.5 and 250 mg/kg of 50 nm PS-NPs in these mice. Our study delineated how PS-NP exposure caused gut microbiota dysbiosis, characterized by Mycoplasma and Coriobacteriaceae proliferation, resulting in increased levels of interleukin 17C (IL-17C) production in the intestines. The surplus IL-17C permeated the brain via the bloodstream, triggering inflammation and brain damage. Our investigation elucidated a direct correlation between intestinal health and neurological outcomes in the context of PS-NP exposure. Susceptible mice with fragile guts exhibited heightened neurotoxicity induced by PS-NPs. This phenomenon was attributed to the elevated abundance of microbiota associated with IL-17C production in the intestines of these mice, such as Mesorhizobium and Lwoffii, provoked by PS-NPs. Neurotoxicity was alleviated by in vivo treatment with anti-IL-17C-neutralizing antibodies or antibiotics. These findings advanced our comprehension of the regulatory mechanisms governing the gut-brain axis in PS-NP-induced neurotoxicity and underscored the critical importance of maintaining intestinal health to mitigate the neurotoxic effects of PS-NPs.
Collapse
Affiliation(s)
- Boxuan Liang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yanhong Deng
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yuji Huang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yizhou Zhong
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhiming Li
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jiaxin Du
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Rongyi Ye
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yu Feng
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Ruobing Bai
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Bingchi Fan
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiaoqing Chen
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiyun Huang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiaohong Yang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hongyi Xian
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xingfen Yang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Zhenlie Huang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
25
|
Fang X, Liu S, Muhammad B, Zheng M, Ge X, Xu Y, Kan S, Zhang Y, Yu Y, Zheng K, Geng D, Liu CF. Gut microbiota dysbiosis contributes to α-synuclein-related pathology associated with C/EBPβ/AEP signaling activation in a mouse model of Parkinson's disease. Neural Regen Res 2024; 19:2081-2088. [PMID: 38227539 DOI: 10.4103/1673-5374.391191] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202409000-00042/figure1/v/2024-01-16T170235Z/r/image-tiff Parkinson's disease is a neurodegenerative disease characterized by motor and gastrointestinal dysfunction. Gastrointestinal dysfunction can precede the onset of motor symptoms by several years. Gut microbiota dysbiosis is involved in the pathogenesis of Parkinson's disease, whether it plays a causal role in motor dysfunction, and the mechanism underlying this potential effect, remain unknown. CCAAT/enhancer binding protein β/asparagine endopeptidase (C/EBPβ/AEP) signaling, activated by bacterial endotoxin, can promote α-synuclein transcription, thereby contributing to Parkinson's disease pathology. In this study, we aimed to investigate the role of the gut microbiota in C/EBPβ/AEP signaling, α-synuclein-related pathology, and motor symptoms using a rotenone-induced mouse model of Parkinson's disease combined with antibiotic-induced microbiome depletion and fecal microbiota transplantation. We found that rotenone administration resulted in gut microbiota dysbiosis and perturbation of the intestinal barrier, as well as activation of the C/EBP/AEP pathway, α-synuclein aggregation, and tyrosine hydroxylase-positive neuron loss in the substantia nigra in mice with motor deficits. However, treatment with rotenone did not have any of these adverse effects in mice whose gut microbiota was depleted by pretreatment with antibiotics. Importantly, we found that transplanting gut microbiota derived from mice treated with rotenone induced motor deficits, intestinal inflammation, and endotoxemia. Transplantation of fecal microbiota from healthy control mice alleviated rotenone-induced motor deficits, intestinal inflammation, endotoxemia, and intestinal barrier impairment. These results highlight the vital role that gut microbiota dysbiosis plays in inducing motor deficits, C/EBPβ/AEP signaling activation, and α-synuclein-related pathology in a rotenone-induced mouse model of Parkinson's disease. Additionally, our findings suggest that supplementing with healthy microbiota may be a safe and effective treatment that could help ameliorate the progression of motor deficits in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoli Fang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Sha Liu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Bilal Muhammad
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yan Xu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Shu Kan
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yang Zhang
- Department of Neurology, Xuzhou Central Hospital, Xuzhou, Jiangsu Province, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Jiangsu Key Laboratory of Neuropsychiatric Disease and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
26
|
Song N, Zhang X, Su J, Chen L, Jin Q, Liu C, Dai Z. Nature and Determinants of Fear of Cancer Recurrence After Endoscopic Submucosal Dissection for Early Gastric Cancer: A Cross-Sectional Study. Gastroenterol Nurs 2024; 47:358-367. [PMID: 39356122 DOI: 10.1097/sga.0000000000000812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/08/2024] [Indexed: 10/03/2024] Open
Abstract
Gastric cancer is one of the most prevalent tumors in China and other countries, with high morbidity and mortality. Fear of cancer recurrence is common among cancer survivors. Fear of cancer recurrence experiences and psychological interventions have been investigated in breast and other cancers. However, this phenomenon and associated factors have not been evaluated in early gastric cancer survivors in China. The objective of this study was to investigate the nature of fear of cancer recurrence and influencing factors in Chinese patients with early gastric cancer treated with endoscopic submucosal dissection. This cross-sectional study in two centers included 312 early gastric cancer patients who answered self-report questionnaires and were treated with endoscopic submucosal dissection between June 2022 and May 2023 to assess fear of cancer recurrence. Gender, family history of gastrointestinal tumor, tumor recurrence, Helicobacter pylori infection, disease perception, and self-perceived burden were significant factors influencing fear of cancer recurrence (p < .05). More than half of early gastric cancer patients have fear of cancer recurrence, and how to deal with it has become a key issue in the postoperative care of patients. Medical professionals should address these factors to reduce fear of cancer recurrence in at-risk patients.
Collapse
Affiliation(s)
- Nian Song
- Nian Song, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Xiaotao Zhang, MD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jie Su, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Lu Chen, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Qianhong Jin, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Chengcheng Liu, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Zhengxiang Dai, MD, RN, Department of Hospital-Acquired Infection Control, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Xiaotao Zhang
- Nian Song, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Xiaotao Zhang, MD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jie Su, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Lu Chen, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Qianhong Jin, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Chengcheng Liu, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Zhengxiang Dai, MD, RN, Department of Hospital-Acquired Infection Control, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Jie Su
- Nian Song, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Xiaotao Zhang, MD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jie Su, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Lu Chen, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Qianhong Jin, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Chengcheng Liu, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Zhengxiang Dai, MD, RN, Department of Hospital-Acquired Infection Control, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Lu Chen
- Nian Song, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Xiaotao Zhang, MD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jie Su, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Lu Chen, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Qianhong Jin, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Chengcheng Liu, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Zhengxiang Dai, MD, RN, Department of Hospital-Acquired Infection Control, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Qianhong Jin
- Nian Song, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Xiaotao Zhang, MD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jie Su, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Lu Chen, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Qianhong Jin, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Chengcheng Liu, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Zhengxiang Dai, MD, RN, Department of Hospital-Acquired Infection Control, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Chengcheng Liu
- Nian Song, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Xiaotao Zhang, MD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jie Su, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Lu Chen, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Qianhong Jin, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Chengcheng Liu, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Zhengxiang Dai, MD, RN, Department of Hospital-Acquired Infection Control, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Zhengxiang Dai
- Nian Song, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Xiaotao Zhang, MD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Jie Su, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Lu Chen, BD, Department of Digestive Endoscopy, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Qianhong Jin, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Chengcheng Liu, MD, School of Nursing, Nanjing University of Chinese Medicine, Jiangsu, China
- Zhengxiang Dai, MD, RN, Department of Hospital-Acquired Infection Control, the Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu, China
| |
Collapse
|
27
|
Bricca L, Porcari S, Savarino E, Rugge M. Microbiota in gastrointestinal malignancies. Best Pract Res Clin Gastroenterol 2024; 72:101953. [PMID: 39645287 DOI: 10.1016/j.bpg.2024.101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 12/09/2024]
Abstract
This manuscript provides an overview of the microbiota profile associated with precancerous lesions in the esophagus, stomach, and large bowel. The critical review of the available data reveals significant variability in the methods used for microbiota profiling. This variability may affect the reliable identification of specific biological links between histologically profiled neoplastic diseases and the microbiota population. Overall, this critical review reveals significant links between microbiota communities and the different lesions within the spectrum of the oncogenetic cascade in various epidemiological contexts and anatomical districts.
Collapse
Affiliation(s)
- Ludovica Bricca
- Department of Surgical Oncological and Gastroenterological Science (DiSCOG), Gastroenterology Unit, University of Padova, Padova, Italy
| | - Serena Porcari
- Department of Medical and Surgical Sciences, University Cattolica del Sacro Cuore - IRCCS Policlinico A. Gemelli, Roma, Italy
| | - Edoardo Savarino
- Department of Surgical Oncological and Gastroenterological Science (DiSCOG), Gastroenterology Unit, University of Padova, Padova, Italy
| | - Massimo Rugge
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy.
| |
Collapse
|
28
|
Niu Y, Li J, Qian H, Liang C, Shi X, Bu S. Evaluation of efficacy and safety of Lacticaseibacillus rhamnosus LRa05 in the eradication of Helicobacter pylori: a randomized, double-blind, placebo-controlled trial. Front Immunol 2024; 15:1450414. [PMID: 39234246 PMCID: PMC11371625 DOI: 10.3389/fimmu.2024.1450414] [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: 06/17/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024] Open
Abstract
Aim This study aims to evaluate the efficacy of Lacticaseibacillus rhamnosus LRa05 supplementation in enhancing Helicobacter pylori (H. pylori) eradication rate and alleviating the gastrointestinal side effects associated with bismuth quadruple therapy. Methods H. pylori-positive patients were randomized to receive levofloxacin-based bismuth quadruple therapy combined either probiotic LRa05 or a placebo for two weeks, followed by LRa05 (1 × 1010 CFU) or maltodextrin for the next two weeks. H. pylori infection was detected by 13C breath test pre- and post-treatment. Blood and stool samples were collected at week 0 and week 4 for routine and biochemical analysis, and serum inflammatory markers. Gastrointestinal symptoms were evaluated using the gastrointestinal symptom rating scale (GSRS). Intestinal microbiota was analyzed using 16S rRNA sequencing. The research was listed under the Chinese Clinical Trial Registry (ChiCTR2300072220), and written informed consent was obtained from all participants. Results The LRa05 group exhibited a trend toward higher H. pylori eradication rates (86.11%) compared to the placebo group (82.86%), though the difference was not statistically significant. Significant reductions in neutrophil count, alanine aminotransferase, aspartate aminotransferase, pepsinogen I, interleukin-6 (IL-6), tumor necrosis factor α (TNF-α) (p < 0.05) suggest that LRa05 supplementation may mitigate inflammation, enhance liver function, and potential aid in early cancer prevention. GSRS symptom scores showed that LRa05 alleviated abdominal pain, acid reflux, bloating, and diarrhea, enhancing patient compliance. Furthermore, 16S rRNA sequencing showed that LRa05 countered the antibiotic-induced disruption of gut microbiota diversity, primarily by increasing beneficial bacteria. Conclusion Although LRa05 did not significantly improve the success rate of H. pylori eradication therapy, it has the potential to improve liver function and reduced levels of inflammatory markers such as IL-6 and TNF-α in the body, regulating the inflammatory response. In addition, it played a positive role in alleviating the adverse symptoms and gut microbiota disturbances caused by eradication therapy, providing a possible way to improve the overall health of patients and demonstrating promising clinical potential. Clinical Trial Registration http://www.chictr.org.cn, identifier ChiCTR2300072220.
Collapse
Affiliation(s)
- Yue Niu
- Department of Gastroenterology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jing Li
- Department of Gastroenterology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hongwei Qian
- Department of General Practice, Shihua Community Health Service Center in Jinshan District, Shanghai, China
| | - Chunli Liang
- Department of Gastroenterology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xinyi Shi
- Department of General Practice, Shihua Community Health Service Center in Jinshan District, Shanghai, China
| | - Shurui Bu
- Department of Gastroenterology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Fathi Kisomi M, Yadegar A, Shekari T, Amin M, Llopis-Lorente A, Liu C, Haririan I, Aghdaei HA, Shokrgozar MA, Zali MR, Rad-Malekshahi M, Miri AH, Hamblin MR, Wacker MG. Unveiling the potential role of micro/nano biomaterials in the treatment of Helicobacter pylori infection. Expert Rev Anti Infect Ther 2024; 22:613-630. [PMID: 39210553 DOI: 10.1080/14787210.2024.2391910] [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: 05/30/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Helicobacter pylori causes stubborn infections and leads to a variety of stomach disorders, such as peptic ulcer, chronic atrophic gastritis, and gastric cancer. Although antibiotic-based approaches have been widely used against H. pylori, some challenges such as antibiotic resistance are increasing in severity. Therefore, simpler but more effective strategies are needed. AREAS COVERED In this review, basic information on functionalized and non-functionalized micro/nano biomaterials and routes of administration for H. pylori inhibition are provided in an easy-to-understand format. Afterward, in vitro and in vivo studies of some promising bio-platforms including metal-based biomaterials, biopolymers, small-molecule saccharides, and vaccines for H. pylori inhibition are discussed in a holistic manner. EXPERT OPINION Functionalized or non-functionalized micro/nano biomaterials loaded with anti-H. pylori agents can show efficient bactericidal activity with no/slight negative influence on the host gastrointestinal microbiota. However, this claim needs to be substantiated with hard data such as assessment of the biopharmaceutical parameters of anti-H. pylori systems and the measurement of diversity/abundance of bacterial genera in the host gastric/gut microbiota before and after H. pylori eradication.
Collapse
Affiliation(s)
- Misagh Fathi Kisomi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tara Shekari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, and the Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Antoni Llopis-Lorente
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, P.R. China
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, 4 Science Drive 2, Singapore 117545, Singapore
| |
Collapse
|
30
|
Zhang W, Zhong Y, Wang Z, Tang F, Zheng C. Apple polysaccharide improves age-matched cognitive impairment and intestinal aging through microbiota-gut-brain axis. Sci Rep 2024; 14:16215. [PMID: 39003416 PMCID: PMC11246462 DOI: 10.1038/s41598-024-67132-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
The Apple polysaccharides (AP), extracted from the fruit of apple, has been used to treat multiple pathological diseases. In this study, we evaluated the effects of AP on cognitive impairment and intestinal aging in naturally aging mice. As a result, it was found that AP could improve spatial learning and memory impairment in aging mice through the Morris water maze experiment. Additionally, AP intervention can upregulate the expression of nerve growth factor (BDNF), postsynaptic marker (PSD95), and presynaptic marker (SYP) proteins. Moreover, AP can enhance total antioxidant capacity, reduce the level of pro-inflammatory cytokine, and inhibit the activation of the NF-κB signaling pathway, exerting anti-inflammatory and antioxidant functions. And the administration of AP restored intestinal mucosal barrier function, reduced the expression of aging and apoptosis related proteins. The administration of AP also altered the gut microbiota of mice. At the genus level, AP decreased the abundance of Helicobacter and Bilophila, while increased the abundance of Lactobacillus and Bacteroides. In summary, these data demonstrate that AP treatment can alleviate cognitive impairment, oxidative stress, and inflammatory reactions, repair the intestinal mucosal barrier, reduce intestinal aging, and alter specific microbial characteristics, ultimately improving the health of the elderly.
Collapse
Affiliation(s)
- Wenming Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 330000, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yuchun Zhong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 330000, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zhuoya Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Furui Tang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Cihua Zheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, 330000, Nanchang, Jiangxi, People's Republic of China.
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China.
- The Institute of Translational Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
31
|
Berard M, Chassain K, Méry C, Gillaizeau F, Carton T, Humeau H, Navasiolava N, Rocour S, Schurgers L, Kempf M, Martin L. Changes in the gut microbiota of pseudoxanthoma elasticum patients. Ann Dermatol Venereol 2024; 151:103290. [PMID: 39003978 DOI: 10.1016/j.annder.2024.103290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 03/11/2024] [Accepted: 05/13/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE Pseudoxanthoma elasticum (PXE) is a rare autosomal disorder with a variable phenotype that may be modulated by environmental factors. Plasma vitamin K (VK) levels may be involved in the ectopic calcification process observed in PXE. Since VK2 is predominantly produced by the gut microbiota, we hypothesized that changes in the gut microbiota of PXE patients might exacerbate the calcification process and disease symptoms. METHODS Twenty PXE patients were included in the study and 60 gut microbiota profiles from the Biofortis laboratory database were used as controls. RESULTS The Rhodospirillaceae family was more abundant in the PXE group while the Sphingomonadaceae family was more abundant in the control group. In a PXE severity subgroup analysis, microbiota dispersion was lower in "severe" than in "non-severe" patients, which was confirmed by permutation multivariate analysis of variance at the phylum, family and genus ranks. However, no significant association was found in a model incorporating relative abundance of bacterial families, severity score, and different blood and fecal VK species. CONCLUSION These results suggest slight compositional changes in the gut microbiota of PXE patients. Further studies are needed to substantiate their impact on VK metabolism and the calcification process.
Collapse
Affiliation(s)
- M Berard
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - K Chassain
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - C Méry
- Biofortis SAS, 44800 Saint Herblain, France
| | | | - T Carton
- Biofortis SAS, 44800 Saint Herblain, France
| | - H Humeau
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - N Navasiolava
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - S Rocour
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - L Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, University of Maastricht, Netherlands
| | - M Kempf
- Laboratory of Bacteriology, Dept. of Infectious Agents, Angers University Hospital, F-49000 Angers, France; Nantes University, Angers University, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, F-44000 Nantes, France
| | - L Martin
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France; Angers University, MitoVasc (INSERM U1083, CNRS 6015), SFR ICAT, F-49000 Angers, France.
| |
Collapse
|
32
|
Kawai T, Kawai Y, Akimito Y, Hamada M, Iwata E, Niikura R, Nagata N, Yanagisawa K, Fukuzawa M, Itoi T, Sugimoto M. Intragastric bacterial infection and endoscopic findings in Helicobacter pylori-negative patients. J Clin Biochem Nutr 2024; 75:65-70. [PMID: 39070535 PMCID: PMC11273264 DOI: 10.3164/jcbn.24-56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 07/30/2024] Open
Abstract
In the present study, the authors examined the association between gastric bacterial infection and gastric endoscopic findings in Helicobacter pylori (H. pylori)-negative patients. The subjects were 105 H. pylori-negative patients. The mean age was 72.8 ± 9.1 years. Endoscopy and gastric juice culture were performed. The presence or absence of endoscopic findings was checked according to the Kyoto classification of gastritis. Culture was positive in 69 patients (65.7%), with Streptococcus α-hemolytic being the most common (51 patients), followed by Neisseria sp. (43 patients). According to the univariate analysis, there was a significant difference between the results of culture and background factors in the use of gastric antisecretory drugs and between the results of culture and various endoscopic findings in atrophic gastritis, intestinal metaplasia, regular arrangement of collecting venule, mucosal swelling, sticky mucus, hyperplastic polyps, hematin, and gastric cobblestone-like lesions. Furthermore, multivariate analysis revealed significant differences in background factors such as the use of gastric antisecretory drugs and endoscopic findings only in patients with mucosal swelling. Endoscopic findings of non-H. pylori bacteria-positive gastritis differed from endoscopic findings of H. pylori-infected gastritis in several respects. In conclusion, our results suggest that non-H. pylori bacteria may infect the stomach and cause gastric inflammation, especially in patients who long term use gastric antisecretory drugs.
Collapse
Affiliation(s)
- Takashi Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Yusuke Kawai
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Yoshika Akimito
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Mariko Hamada
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Eri Iwata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Ryota Niikura
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Kyosuke Yanagisawa
- Department of Gastroenterological Endoscopy, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Masakatsu Fukuzawa
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Mitsushige Sugimoto
- Division of Genome-Wide Infectious Diseases, Research Center for GLOBAL and LOCAL Infectious Disease, Oita University, 1-1 Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| |
Collapse
|
33
|
Tohumcu E, Kaitsas F, Bricca L, Ruggeri A, Gasbarrini A, Cammarota G, Ianiro G. Helicobacter pylori and the Human Gastrointestinal Microbiota: A Multifaceted Relationship. Antibiotics (Basel) 2024; 13:584. [PMID: 39061266 PMCID: PMC11274338 DOI: 10.3390/antibiotics13070584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Helicobacter pylori is a type of Gram-negative bacteria belonging to the Proteobacteria phylum which is known to cause gastrointestinal disorders such as gastritis and gastric ulcers. Its treatment is based on current eradication regimens, which are composed of combinations of antibiotics such as clarithromycin, metronidazole, levofloxacin and amoxicillin, often combined with a proton pump inhibitor (PPI). With the development of sequencing technologies, it has been demonstrated that not only does the colonization of the gastric and gut environment by H. pylori cause microbial changes, but also the treatment regimens used for its eradication have a significant altering effect on both the gastric and gut microbiota. Here, we review current knowledge on microbiota modulations of current therapies in both environments. We also summarize future perspectives regarding H. pylori infection, the integration of probiotics into therapy and what challenges are being faced on a global basis when we talk about eradication.
Collapse
Affiliation(s)
- Ege Tohumcu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (E.T.); (F.K.); (A.R.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Kaitsas
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (E.T.); (F.K.); (A.R.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Ludovica Bricca
- Department of Surgical, Oncological and Gastroenterological Sciences (DiSCOG), Padua Univeristy, 35123 Padova, Italy;
| | - Alessandro Ruggeri
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (E.T.); (F.K.); (A.R.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (E.T.); (F.K.); (A.R.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (E.T.); (F.K.); (A.R.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (E.T.); (F.K.); (A.R.); (A.G.); (G.C.)
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
34
|
Montironi ID, Arsaute S, Roma DA, Cecchini ME, Pinotti A, Mañas F, Bessone FA, de Moreno de LeBlanc A, Alustiza FE, Bellingeri RV, Cariddi LN. Evaluation of oral supplementation of free and nanoencapsulated Minthostachys verticillata essential oil on immunological, biochemical and antioxidants parameters and gut microbiota in weaned piglets. Vet Res Commun 2024; 48:1641-1658. [PMID: 38453821 DOI: 10.1007/s11259-024-10347-7] [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: 01/12/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
Early weaning is an important stressor that impairs the piglet´s health, and essential oils appear as promising candidates to improve it instead of antibiotics. The aim of this study was to evaluate the effect of oral supplementation of free and nanoencapsulated Minthostachys verticillata essential oil (EO and NEO, respectively) on immunological, biochemical and antioxidants parameters as well as on gut microbiota in weaned piglets. EO was extracted by hydrodistillation and nanoencapsulation was performed by high-energy method using Tween 80 and Span 60 as surfactants. EO and NEO were chemically analyzed by gas chromatography-mass spectrometry (GC-MS). The cytotoxic effects of both EO and NEO was evaluated on Caco-2 cell line. For in vivo assay, male weaned piglets (age: 28 days, mean initial body weight: 11.63 ± 0.37 kg) were randomly distributed in six groups of six animals each (n = 6) and received orally EO (10.0 mg/kg/day) or NEO (2.5, 5.0 and 10.0 mg/kg/day), named hereinafter as EO-10, NEO-2.5, NEO-5 and NEO-10, for 30 consecutive days. Animals not treated or treated with surfactants mixture were evaluated as control and vehicle control. Subsequently, histological, hematological and biochemical parameters, cytokines production, oxidative markers, CD4+/CD8+ T cells and gut microbiota were evaluated. GC-MS analysis was similar in both EO and NEO. The NEO was more toxic on Caco-2 cells than EO. Oral supplementation of EO-10 or NEO-10 improved growth performance compared to control group NEO-2.5 or NEO-5 (p < 0.05) groups. NEO-2.5, NEO-5 and NEO-10 did not alter the morpho-physiology of digestive organs and decreased malondialdehyde (MDA) levels in liver compared to control (p < 0.05) or EO-10 groups (p < 0.05, p < 0.01). In addition, NEO-10 showed an increase in CD4+/CD8+ T cells ratio (p < 0.001), and induced the highest serum levels of IL-10 (p < 0.01). Serum triglycerides levels were significantly lower in animals treated with EO-10 or NEO-2.5, NEO-5 and NEO-10 compared to control group (p < 0.001). Gut microbiota analysis showed that NEO-10 favor the development of beneficial intestinal microorganisms to improve parameters related to early weaning of piglets. In conclusion, EO and NEO improved parameters altered by early weaning in piglets however, NEO was safer and powerful. Therefore, NEO should be further studied to be applied in swine health.
Collapse
Affiliation(s)
- Ivana D Montironi
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Río Cuarto, Córdoba, 5800, Argentina
| | - Sofía Arsaute
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Río Cuarto, Córdoba, 5800, Argentina
| | - Dardo A Roma
- Facultad de Agronomía y Veterinaria. Cátedra de Farmacología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Ciencias Veterinarias (INCIVET), Río Cuarto, Córdoba, 5800, Argentina
| | - María E Cecchini
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Río Cuarto, Córdoba, 5800, Argentina
| | - Agustina Pinotti
- Instituto Nacional de Tecnología Agropecuaria (INTA), Estación Experimental Agropecuaria Marcos Juárez, Marcos Juárez 2580, Córdoba, Argentina
| | - Fernando Mañas
- Facultad de Agronomía y Veterinaria. Cátedra de Farmacología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Ciencias Veterinarias (INCIVET), Río Cuarto, Córdoba, 5800, Argentina
| | - Fernando A Bessone
- Instituto Nacional de Tecnología Agropecuaria (INTA), Estación Experimental Agropecuaria Marcos Juárez, Marcos Juárez 2580, Córdoba, Argentina
| | - Alejandra de Moreno de LeBlanc
- Centro de Referencia para Lactobacilos (CERELA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Miguel de Tucumán, Tucumán, 4000, Argentina
| | - Fabrisio E Alustiza
- Instituto Nacional de Tecnología Agropecuaria (INTA), Estación Experimental Agropecuaria Marcos Juárez, Marcos Juárez 2580, Córdoba, Argentina
| | - Romina V Bellingeri
- Facultad de Agronomía y Veterinaria, Departamento de Anatomía Animal, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Río Cuarto, Córdoba, 5800, Argentina
| | - Laura Noelia Cariddi
- Facultad de Ciencias Exactas Físico-Químicas y Naturales, Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Río Cuarto, Córdoba, 5800, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Río Cuarto, Córdoba, 5800, Argentina.
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biotecnología Ambiental y Salud (INBIAS), Ruta 36 Km 601, Río Cuarto, Córdoba, CP: 5800, Argentina.
| |
Collapse
|
35
|
Wang JL, Jing DD. Gastric microbiome and gastric cancer: Relationship, mechanism, and clinical significance. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:327-332. [DOI: 10.11569/wcjd.v32.i5.327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
|
36
|
Leoni C, Vinci L, Marzano M, D’Erchia AM, Dellino M, Cox SN, Vitagliano A, Visci G, Notario E, Filomena E, Cicinelli E, Pesole G, Ceci LR. Endometrial Cancer: A Pilot Study of the Tissue Microbiota. Microorganisms 2024; 12:1090. [PMID: 38930472 PMCID: PMC11205883 DOI: 10.3390/microorganisms12061090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The endometrium remains a difficult tissue for the analysis of microbiota, mainly due to the low bacterial presence and the sampling procedures. Among its pathologies, endometrial cancer has not yet been completely investigated for its relationship with microbiota composition. In this work, we report on possible correlations between endometrial microbiota dysbiosis and endometrial cancer. METHODS Women with endometrial cancer at various stages of tumor progression were enrolled together with women with a benign polymyomatous uterus as the control. Analyses were performed using biopsies collected at two specific endometrial sites during the surgery. This study adopted two approaches: the absolute quantification of the bacterial load, using droplet digital PCR (ddPCR), and the analysis of the bacterial composition, using a deep metabarcoding NGS procedure. RESULTS ddPCR provided the first-ever assessment of the absolute quantification of bacterial DNA in the endometrium, confirming a generally low microbial abundance. Metabarcoding analysis revealed a different microbiota distribution in the two endometrial sites, regardless of pathology, accompanied by an overall higher prevalence of pathogenic bacterial genera in cancerous tissues. CONCLUSIONS These results pave the way for future studies aimed at identifying potential biomarkers and gaining a deeper understanding of the role of bacteria associated with tumors.
Collapse
Affiliation(s)
- Claudia Leoni
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| | - Lorenzo Vinci
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Marinella Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| | - Anna Maria D’Erchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Miriam Dellino
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Sharon Natasha Cox
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Amerigo Vitagliano
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Grazia Visci
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Elisabetta Notario
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| | - Ermes Filomena
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Ettore Cicinelli
- 2nd Unit of Obstetrics and Gynaecology, Department of Biomedical Science and Human Oncology, University of Bari “A. Moro”, Piazza G. Cesare, 70124 Bari, Italy; (L.V.); (M.D.); (A.V.); (E.C.)
| | - Graziano Pesole
- Department of Biosciences, Biotechnologies and Environment, University of Bari A. Moro, Via Orabona n. 4, 70126 Bari, Italy; (A.M.D.); (S.N.C.); (G.V.); (E.F.); (G.P.)
| | - Luigi Ruggiero Ceci
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council (CNR), Via Amendola n. 122/O, 70126 Bari, Italy; (M.M.); (E.N.)
| |
Collapse
|
37
|
Sgamato C, Rocco A, Compare D, Priadko K, Romano M, Nardone G. Exploring the Link between Helicobacter pylori, Gastric Microbiota and Gastric Cancer. Antibiotics (Basel) 2024; 13:484. [PMID: 38927151 PMCID: PMC11201017 DOI: 10.3390/antibiotics13060484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Gastric cancer (GC) still represents one of the leading causes of cancer-related mortality and is a major public health issue worldwide. Understanding the etiopathogenetic mechanisms behind GC development holds immense potential to revolutionize patients' treatment and prognosis. Within the complex web of genetic predispositions and environmental factors, the connection between Helicobacter pylori (H. pylori) and gastric microbiota emerges as a focus of intense research investigation. According to the most recent hypotheses, H. pylori triggers inflammatory responses and molecular alterations in gastric mucosa, while non-Helicobacter microbiota modulates disease progression. In this review, we analyze the current state of the literature on the relationship between H. pylori and non-Helicobacter gastric microbiota in gastric carcinogenesis, highlighting the mechanisms by which microecological dysbiosis can contribute to the malignant transformation of the mucosa.
Collapse
Affiliation(s)
- Costantino Sgamato
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, 80131 Naples, Italy; (C.S.); (D.C.); (G.N.)
| | - Alba Rocco
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, 80131 Naples, Italy; (C.S.); (D.C.); (G.N.)
| | - Debora Compare
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, 80131 Naples, Italy; (C.S.); (D.C.); (G.N.)
| | - Kateryna Priadko
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (K.P.); (M.R.)
| | - Marco Romano
- Hepatogastroenterology Unit, Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (K.P.); (M.R.)
| | - Gerardo Nardone
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, 80131 Naples, Italy; (C.S.); (D.C.); (G.N.)
| |
Collapse
|
38
|
Arai J, Fujiwara H, Aoki T, Niikura R, Ihara S, Suzuki N, Hayakawa Y, Kasuga M, Fujishiro M. Metabolic Factors Associated with Endoscopic Atrophy, Intestinal Metaplasia, and Gastric Neoplasms in Helicobacter pylori-Positive Patients. Clin Pract 2024; 14:779-788. [PMID: 38804394 PMCID: PMC11130883 DOI: 10.3390/clinpract14030062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Previous studies demonstrate an association between metabolic factors and Helicobacter pylori-related gastric cancer. However, the association of gastric atrophy or intestinal metaplasia (IM) with these factors remains unknown. METHODS Data on 1603 Helicobacter pylori-positive patients who underwent esophagogastroduodenoscopy between 2001 and 2021 were evaluated. The outcome measures were endoscopic atrophy, IM grade, and the incidence of endoscopically diagnosed and pathologically confirmed gastric neoplasms. Clinical factors associated with these findings were also determined. RESULTS Advanced age; successful Helicobacter pylori eradication; and comorbidities including diabetes mellitus (DM), hypertension, dyslipidemia, and fib4 index were significantly associated with endoscopic gastric atrophy grade. Male sex; advanced age; and comorbidities including DM, hypertension, dyslipidemia, hyperuricemia, fatty liver, aortic calcification, and fib4 index were also significantly associated with endoscopic IM grade, whereas advanced age, successful Helicobacter pylori eradication, DM, fatty liver, and fib4 index were significantly associated with the incidence of gastric neoplasms. CONCLUSION Several metabolic disorders, including DM, hypertension, dyslipidemia, hyperuricemia, and fatty liver disease, are risk factors for advanced-grade gastric atrophy, intestinal metaplasia, and gastric neoplasms. Risk stratification according to these factors, particularly those with metabolic disorders, would affect EGD surveillance for Helicobacter pylori-positive patients.
Collapse
Affiliation(s)
- Junya Arai
- Division of Gastroenterology, The Institute of Medical Science, Asahi Life Foundation, Tokyo 103-0002, Japan;
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan (S.I.); (N.S.); (Y.H.); (M.F.)
| | - Hiroaki Fujiwara
- Division of Gastroenterology, The Institute of Medical Science, Asahi Life Foundation, Tokyo 103-0002, Japan;
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan (S.I.); (N.S.); (Y.H.); (M.F.)
| | - Tomonori Aoki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan (S.I.); (N.S.); (Y.H.); (M.F.)
| | - Ryota Niikura
- Department of Endoscopy, Graduate School of Medicine, Tokyo Medical University, Tokyo 160-0023, Japan;
| | - Sozaburo Ihara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan (S.I.); (N.S.); (Y.H.); (M.F.)
| | - Nobumi Suzuki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan (S.I.); (N.S.); (Y.H.); (M.F.)
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan (S.I.); (N.S.); (Y.H.); (M.F.)
| | - Masato Kasuga
- The Institute of Medical Science, Asahi Life Foundation, Tokyo 103-0002, Japan;
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan (S.I.); (N.S.); (Y.H.); (M.F.)
| |
Collapse
|
39
|
Chen WQ, Fan QF, He YJ, Li F, Wu X, Li YP, Yang XJ. Yiwei Xiaoyu granules for treatment of chronic atrophic gastritis with deficiency syndrome of the spleen and stomach. World J Clin Cases 2024; 12:2201-2209. [PMID: 38808353 PMCID: PMC11129131 DOI: 10.12998/wjcc.v12.i13.2201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/03/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND The Correa sequence, initiated by Helicobacter pylori (H. pylori), commonly progresses to gastric cancer through the stage of chronic atrophic gastritis (CAG). Although eradication of H. pylori only reduces the risk of gastric cancer, it does not eliminate the risk for neoplastic progression. Yiwei Xiaoyu granules (YWXY) are a commonly used composite preparation in Chinese clinics. However, the pursuit of excellence in clinical trials and the establishment of standardized animal experiments are still needed to contribute to full understanding and application of traditional Chinese medicine in the treatment of CAG. AIM To demonstrate the effectiveness of YWXY in patients with CAG and spleen-stomach deficiency syndrome (DSSS), by alleviating histological scores, improving response rates for pathological lesions, and achieving clinical efficacy in relieving DSSS symptoms. METHODS We designed a double-blind, randomized, controlled trial. The study enrolled seventy-two H. pylori-negative patients (mean age, 52.3 years; 38 men) who were randomly allocated to either the treatment group or control group in a 1:1 ratio, and treated with 15 g YWXY or 0.36 g Weifuchun (WFC) tablet combined with the respective dummy for 24 wk. The pre-randomization phase resulted in the exclusion of 72 patients: 50 participants did not meet the inclusion criteria, 12 participants declined to participate, and 10 participants were excluded for various other reasons. Seven visits were conducted during the study, and histopathological examination with target endoscopic biopsy of narrow-band imaging was requested before the first and seventh visits. We also evaluated endoscopic performance scores, total symptom scores, serum pepsinogen and gastrin-17. RESULTS Six patients did not complete the trial procedures. Treatment with YWXY improved the Operative Link on Gastric Intestinal Metaplasia Assessment (OLGIM) stage, compared with WFC (P < 0.05). YWXY provided better relief from symptoms of DSSS and better improvement in serum gastric function, compared with WFC (P < 0.05). CONCLUSION YWXY compared with WFC significantly reduced the risk of mild or moderate atrophic disease, according to OLGIM stage, significantly relieved symptoms of DSSS, and improved serum gastric function.
Collapse
Affiliation(s)
- Wan-Qun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| | - Qing-Feng Fan
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| | - Yuan-Jun He
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| | - Fei Li
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| | - Xin Wu
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| | - Yan-Ping Li
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| | - Xiao-Jun Yang
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China
| |
Collapse
|
40
|
Zheng W, Gan Y, Yang Y, Peng K, Li F, Zhao H, Gu W, Jiang M. Clinicopathological features and mucosal microbiota in gastric mucosal damage between nodular and non-nodular gastritis in children with Helicobacter pylori infection. Int Immunopharmacol 2024; 131:111813. [PMID: 38493689 DOI: 10.1016/j.intimp.2024.111813] [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: 12/09/2023] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024]
Abstract
AIMS Nodular gastritis (NG) represents a frequently observed clinical presentation of Helicobacter pylori (H. pylori) infection in pediatric patients. This investigation aimed to explore the microbiota and histological features of the gastric mucosa in children with H. pylori colonized NG. MAIN METHODS The current investigation examined a sample of 120 children who underwent gastroscopy due to symptoms of gastrointestinal distress, which showed that 64 were patients with H. pylori infection. Endoscopic procedures were conducted to acquire mucosal biopsies for the purpose of DNA extraction and histopathological analysis. The 16S rRNA profiling was utilized to examine the gastric mucosal microbiota. KEY FINDINGS In conjunction with endoscopic evaluation, 26 of 64 patients were diagnosed with NG. The NG group had significantly higher inflammation scores and activity scores on histological assessment than the non-NG group. The NG group exhibited a significant reduction in the richness levels of the five genera. In terms of the predicted functions, the pathways of synthesis and degradation of ketone bodies and phagosome in the NG group were less abundant compared with the non-NG group, while the Wnt signaling pathway was significantly enriched. NG does not increase a microbial community that possesses genotoxic potential within the gastric mucosa. SIGNIFICANCE In conclusion, NG group exhibited significant severe inflammation and reduced abundance levels of several bacterial genera compared to the non-NG group. However, individuals with NG did not have a dysregulated microbial community with genotoxic potential.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Yongjie Gan
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Yaofeng Yang
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Kerong Peng
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Fubang Li
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Hong Zhao
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Weizhong Gu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Mizu Jiang
- Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China; Pediatric Endoscopy Center and Gastrointestinal Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China.
| |
Collapse
|
41
|
Liu Z, Zhang D, Chen S. Unveiling the gastric microbiota: implications for gastric carcinogenesis, immune responses, and clinical prospects. J Exp Clin Cancer Res 2024; 43:118. [PMID: 38641815 PMCID: PMC11027554 DOI: 10.1186/s13046-024-03034-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/29/2024] [Indexed: 04/21/2024] Open
Abstract
High-throughput sequencing has ushered in a paradigm shift in gastric microbiota, breaking the stereotype that the stomach is hostile to microorganisms beyond H. pylori. Recent attention directed toward the composition and functionality of this 'community' has shed light on its potential relevance in cancer. The microbial composition in the stomach of health displays host specificity which changes throughout a person's lifespan and is subject to both external and internal factors. Distinctive alterations in gastric microbiome signature are discernible at different stages of gastric precancerous lesions and malignancy. The robust microbes that dominate in gastric malignant tissue are intricately implicated in gastric cancer susceptibility, carcinogenesis, and the modulation of immunosurveillance and immune escape. These revelations offer fresh avenues for utilizing gastric microbiota as predictive biomarkers in clinical settings. Furthermore, inter-individual microbiota variations partially account for differential responses to cancer immunotherapy. In this review, we summarize current literature on the influence of the gastric microbiota on gastric carcinogenesis, anti-tumor immunity and immunotherapy, providing insights into potential clinical applications.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Dachuan Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Siyu Chen
- Department of Oncology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
42
|
Ke Y, Tan C, Zhen J, Dong W. Global status and trends of gastric cancer and gastric microbiota research: a bibliometric analysis. Front Microbiol 2024; 15:1341012. [PMID: 38655079 PMCID: PMC11037409 DOI: 10.3389/fmicb.2024.1341012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/12/2024] [Indexed: 04/26/2024] Open
Abstract
Background Numerous studies have cast light on the relationship between the gastric microbiota and gastric carcinogenesis. In this study, we conducted a bibliometric analysis of the relevant literature in the field of gastric cancer and the gastric microbiota and clarified its research status, hotspots, and development trends. Materials and methods Publications were retrieved from the Web of Science Core Collection on 18 July 2023. CiteSpace 6.2.R4, VOSviewer 1.6.19.0, and Biblioshiny were used for the co-occurrence and cooperation analyses of countries, institutions, authors, references, and keywords. A keyword cluster analysis and an emergence analysis were performed, and relevant knowledge maps were drawn. Results The number of published papers in this field totaled 215 and showed an increasing trend. The analysis of funding suggested that the input in this field is increasing steadily. China had the highest number of publications, while the United States had the highest betweenness centrality. Baylor College of Medicine published the most articles cumulatively. Both Ferreira RM and Cooker OO had the highest citation frequency. The journal Helicobacter showed the most interest in this field, while Gut provided a substantial research foundation. A total of 280 keywords were obtained using CiteSpace, which were primarily focused on the eradication and pathogenic mechanisms of Helicobacter pylori, as well as the application of the gastric microbiota in the evaluation and treatment of gastric cancer. The burst analysis suggested that in the future, research may focus on the application of gastric microorganisms, particularly Fusobacterium nucleatum, in the diagnosis and treatment of gastric cancer, along with their pathogenic mechanisms. Conclusion Current studies have been tracking the eradication of Helicobacter pylori and its pathogenic mechanisms, as well as changes in the gastric microbiota during gastric carcinogenesis. Future research may focus on the clinical application and pathogenesis of stomach microorganisms through bacteria such as Fusobacterium nucleatum.
Collapse
Affiliation(s)
- Yujia Ke
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junhai Zhen
- Department of General Practice, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
43
|
Jiang W, Tan J, Zhang J, Deng X, He X, Zhang J, Liu T, Sun R, Sun M, Chen K, Xu T, Yan Y, Moazzami A, Wu EJ, Zhan J, Hu B. Polysaccharides from Dendrobium officinale improve obesity-induced insulin resistance through the gut microbiota and the SOCS3-mediated insulin receptor substrate-1 signaling pathway. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:3437-3447. [PMID: 38111200 DOI: 10.1002/jsfa.13229] [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: 06/27/2023] [Revised: 11/24/2023] [Accepted: 12/19/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Obesity induces insulin resistance and chronic inflammation, impacting human health. The relationship between obesity, gut microbiota, and regulatory mechanisms has been studied extensively. Dendrobium officinale polysaccharide (DOP), a traditional Chinese herbal medicine, potentially reduces insulin resistance. However, the mechanism through which DOP affects gut microbiota and alleviates obesity-induced insulin resistance in rats requires further investigation. RESULTS The current study aimed to assess the impact of DOP on gut microbiota and insulin resistance in rats on a high-fat diet. The results revealed that DOP effectively reduced blood lipids, glucose disorders, oxidative stress, and inflammatory infiltration in the liver of obese Sprague Dawley rats. This was achieved by downregulating SOCS3 expression and upregulating insulin receptor substrate-1 (IRS-1) by regulating the JAK/STAT/SOCS3 signaling pathway. Notably, DOP intervention enhanced the abundance of beneficial gut microbiota and reduced harmful microbiota. Correlation analysis demonstrated significant associations among intestinal microbiota, SOCS3-mediated IRS-1 expression, and inflammatory factors. CONCLUSION Dendrobium officinale polysaccharide regulated the gut microbiota, enhanced IRS-1 expression, and mitigated liver injury and insulin resistance due to a high-fat diet. These findings depict the potential anti-insulin resistance properties of DOP and offer further evidence for addressing obesity and its complications. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wei Jiang
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
- Sichuan Provincial key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, China
| | - Jin Tan
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Jiacheng Zhang
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Deng
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Xinyue He
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, USA
| | - Tong Liu
- Department of Forest Ecology and Management, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Rong Sun
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Mengxun Sun
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Kuo Chen
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tingjia Xu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Yuling Yan
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Ali Moazzami
- Department of Molecular Sciences, Uppsala Biocenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - E-Jiao Wu
- Institute of Pomology, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Jiangsu Key Laboratory for Horticultural Crop Genetic Improvement, Nanjing, China
| | - Jiasui Zhan
- Department of Forest Mycology and Plant pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Binhong Hu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
- Sichuan Provincial key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, China
- Department of Forest Mycology and Plant pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
44
|
Zhang J, Liu X, Usman T, Tang Y, Mi S, Li W, Yang M, Yu Y. Integrated analysis of transcriptome and milk metagenome in subclinical mastitic and healthy cows. Anim Biosci 2024; 37:709-717. [PMID: 35073659 PMCID: PMC10915226 DOI: 10.5713/ab.21.0495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/14/2021] [Accepted: 01/18/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Abnormally increased somatic cell counts (SCCs) in milk is usually a sign of bovine subclinical mastitis. Mutual interaction between the host and its associated microbiota plays an important role in developing such diseases. The main objective of this study was to explore the difference between cows with elevated SCCs and healthy cattle from the perspective of host-microbe interplay. METHODS A total of 31 milk samples and 23 bovine peripheral blood samples were collected from Holstein dairy cattle to conduct an integrated analysis of transcriptomic and metagenomics. RESULTS The results showed that Ralstonia and Sphingomonas were enriched in cows with subclinical mastitis. The relative abundance of the two bacteria was positively correlated with the expression level of bovine transcobalamin 1 and uridine phosphorylase 1 encoding gene. Moreover, functional analysis revealed a distinct alternation in some important microbial biological processes. CONCLUSION These results reveal the relative abundance of Ralstonia and Sphingomonas other than common mastitis-causing pathogens varied from healthy cows to those with subclinical mastitis and might be associated with elevated SCCs. Potential association was observed between bovine milk microbiota composition and the transcriptional pattern of some genes, thus providing new insights to understand homeostasis of bovine udder.
Collapse
Affiliation(s)
- Jinning Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193
China
| | - Xueqin Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193
China
| | - Tahir Usman
- College of Veterinary Sciences and Animal Husbandry, Abdul Wali Khan University, Mardan, 23200,
Pakistan
| | - Yongjie Tang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193
China
| | - Siyuan Mi
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193
China
| | - Wenlong Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193
China
| | - Mengyou Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193
China
| | - Ying Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193
China
| |
Collapse
|
45
|
Lei C, Xu Y, Zhang S, Huang C, Qin J. The role of microbiota in gastric cancer: A comprehensive review. Helicobacter 2024; 29:e13071. [PMID: 38643366 DOI: 10.1111/hel.13071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Gastric cancer (GC) continues to pose a significant global threat in terms of cancer-related fatalities. Despite notable advancements in medical research and therapies, further investigation is warranted to elucidate its underlying etiology and risk factors. Recent times have witnessed an escalated emphasis on comprehending the role of the microbiota in cancer development. METHODS This review briefly delves into recent developments in microbiome-related research pertaining to gastric cancer. RESULTS According to studies, the microbiota can influence GC growth by inciting inflammation, disrupting immunological processes, and generating harmful microbial metabolites. Furthermore, there is ongoing research into how the microbiome can impact a patient's response to chemotherapy and immunotherapy. CONCLUSION The utilization of the microbiome for detecting, preventing, and managing stomach cancer remains an active area of exploration.
Collapse
Affiliation(s)
- Changzhen Lei
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yitian Xu
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shaopeng Zhang
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Qin
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
46
|
Rugge M, Genta RM, Malfertheiner P, Dinis-Ribeiro M, El-Serag H, Graham DY, Kuipers EJ, Leung WK, Park JY, Rokkas T, Schulz C, El-Omar EM. RE.GA.IN.: the Real-world Gastritis Initiative-updating the updates. Gut 2024; 73:407-441. [PMID: 38383142 DOI: 10.1136/gutjnl-2023-331164] [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: 09/18/2023] [Accepted: 12/18/2023] [Indexed: 02/23/2024]
Abstract
At the end of the last century, a far-sighted 'working party' held in Sydney, Australia addressed the clinicopathological issues related to gastric inflammatory diseases. A few years later, an international conference held in Houston, Texas, USA critically updated the seminal Sydney classification. In line with these initiatives, Kyoto Global Consensus Report, flanked by the Maastricht-Florence conferences, added new clinical evidence to the gastritis clinicopathological puzzle.The most relevant topics related to the gastric inflammatory diseases have been addressed by the Real-world Gastritis Initiative (RE.GA.IN.), from disease definitions to the clinical diagnosis and prognosis. This paper reports the conclusions of the RE.GA.IN. consensus process, which culminated in Venice in November 2022 after more than 8 months of intense global scientific deliberations. A forum of gastritis scholars from five continents participated in the multidisciplinary RE.GA.IN. consensus. After lively debates on the most controversial aspects of the gastritis spectrum, the RE.GA.IN. Faculty amalgamated complementary knowledge to distil patient-centred, evidence-based statements to assist health professionals in their real-world clinical practice. The sections of this report focus on: the epidemiology of gastritis; Helicobacter pylori as dominant aetiology of environmental gastritis and as the most important determinant of the gastric oncogenetic field; the evolving knowledge on gastric autoimmunity; the clinicopathological relevance of gastric microbiota; the new diagnostic horizons of endoscopy; and the clinical priority of histologically reporting gastritis in terms of staging. The ultimate goal of RE.GA.IN. was and remains the promotion of further improvement in the clinical management of patients with gastritis.
Collapse
Affiliation(s)
- Massimo Rugge
- Department of Medicine-DIMED, University of Padova, Padua, Italy
- Azienda Zero, Veneto Tumour Registry, Padua, Italy
| | - Robert M Genta
- Gastrointestinal Pathology, Inform Diagnostics Research Institute, Dallas, Texas, USA
- Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Malfertheiner
- Medizinische Klinik und Poliklinik II, Ludwig Maximilian Universität Klinikum München, Munich, Germany
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | - Mario Dinis-Ribeiro
- Porto Comprehensive Cancer Center & RISE@CI-IPO, University of Porto, Porto, Portugal
- Gastroenterology Department, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Hashem El-Serag
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Houston VA Health Services Research & Development Center of Excellence, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - David Y Graham
- Department of Medicine, Michael E DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Ernst J Kuipers
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Jin Young Park
- International Agency for Research on Cancer, Lyon, France
| | - Theodore Rokkas
- Gastroenterology, Henry Dunant Hospital Center, Athens, Greece
| | | | - Emad M El-Omar
- Microbiome Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Fu K, Cheung AHK, Wong CC, Liu W, Zhou Y, Wang F, Huang P, Yuan K, Coker OO, Pan Y, Chen D, Lam NM, Gao M, Zhang X, Huang H, To KF, Sung JJY, Yu J. Streptococcus anginosus promotes gastric inflammation, atrophy, and tumorigenesis in mice. Cell 2024; 187:882-896.e17. [PMID: 38295787 DOI: 10.1016/j.cell.2024.01.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/13/2023] [Accepted: 01/02/2024] [Indexed: 02/18/2024]
Abstract
Streptococcus anginosus (S. anginosus) was enriched in the gastric mucosa of patients with gastric cancer (GC). Here, we show that S. anginosus colonized the mouse stomach and induced acute gastritis. S. anginosus infection spontaneously induced progressive chronic gastritis, parietal cell atrophy, mucinous metaplasia, and dysplasia in conventional mice, and the findings were confirmed in germ-free mice. In addition, S. anginosus accelerated GC progression in carcinogen-induced gastric tumorigenesis and YTN16 GC cell allografts. Consistently, S. anginosus disrupted gastric barrier function, promoted cell proliferation, and inhibited apoptosis. Mechanistically, we identified an S. anginosus surface protein, TMPC, that interacts with Annexin A2 (ANXA2) receptor on gastric epithelial cells. Interaction of TMPC with ANXA2 mediated attachment and colonization of S. anginosus and induced mitogen-activated protein kinase (MAPK) activation. ANXA2 knockout abrogated the induction of MAPK by S. anginosus. Thus, this study reveals S. anginosus as a pathogen that promotes gastric tumorigenesis via direct interactions with gastric epithelial cells in the TMPC-ANXA2-MAPK axis.
Collapse
Affiliation(s)
- Kaili Fu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alvin Ho Kwan Cheung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Weixin Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yunfei Zhou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Feixue Wang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pingmei Huang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kai Yuan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Olabisi Oluwabukola Coker
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yasi Pan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Danyu Chen
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Nga Man Lam
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mengxue Gao
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - He Huang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joseph Jao Yiu Sung
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
48
|
Apaza CJ, Días M, García Tejedor A, Boscá L, Laparra Llopis JM. Contribution of Nucleotide-Binding Oligomerization Domain-like (NOD) Receptors to the Immune and Metabolic Health. Biomedicines 2024; 12:341. [PMID: 38397943 PMCID: PMC10886542 DOI: 10.3390/biomedicines12020341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Nucleotide-binding oligomerization domain-like (NOD) receptors rely on the interface between immunity and metabolism. Dietary factors constitute critical players in the activation of innate immunity and modulation of the gut microbiota. The latter have been involved in worsening or improving the control and promotion of diseases such as obesity, type 2 diabetes, metabolic syndrome, diseases known as non-communicable metabolic diseases (NCDs), and the risk of developing cancer. Intracellular NODs play key coordinated actions with innate immune 'Toll-like' receptors leading to a diverse array of gene expressions that initiate inflammatory and immune responses. There has been an improvement in the understanding of the molecular and genetic implications of these receptors in, among others, such aspects as resting energy expenditure, insulin resistance, and cell proliferation. Genetic factors and polymorphisms of the receptors are determinants of the risk and severity of NCDs and cancer, and it is conceivable that dietary factors may have significant differential consequences depending on them. Host factors are difficult to influence, while environmental factors are predominant and approachable with a preventive and/or therapeutic intention in obesity, T2D, and cancer. However, beyond the recognition of the activation of NODs by peptidoglycan as its prototypical agonist, the underlying molecular response(s) and its consequences on these diseases remain ill-defined. Metabolic (re)programming is a hallmark of NCDs and cancer in which nutritional strategies might play a key role in preventing the unprecedented expansion of these diseases. A better understanding of the participation and effects of immunonutritional dietary ingredients can boost integrative knowledge fostering interdisciplinary science between nutritional precision and personalized medicine against cancer. This review summarizes the current evidence concerning the relationship(s) and consequences of NODs on immune and metabolic health.
Collapse
Affiliation(s)
- César Jeri Apaza
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| | - Marisol Días
- Center of Biological Enginneering (CEB), Iberian Nantotechnology Laboratory (INL), University of Minho, 4715-330 Braga, Portugal;
| | - Aurora García Tejedor
- Bioactivity and Nutritional Immunology Group (BIOINUT), Faculty of Health Sciences, Universidad Internacional de Valencia (VIU), Pintor Sorolla 21, 46002 Valencia, Spain;
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols-Morreale (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Melchor Fernández Almagro 6, 28029 Madrid, Spain
| | - José Moisés Laparra Llopis
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA Food), Ctra Cantoblanco, 8, 28049 Madrid, Spain;
| |
Collapse
|
49
|
Yarahmadi A, Afkhami H. The role of microbiomes in gastrointestinal cancers: new insights. Front Oncol 2024; 13:1344328. [PMID: 38361500 PMCID: PMC10867565 DOI: 10.3389/fonc.2023.1344328] [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: 12/01/2023] [Accepted: 12/20/2023] [Indexed: 02/17/2024] Open
Abstract
Gastrointestinal (GI) cancers constitute more than 33% of new cancer cases worldwide and pose a considerable burden on public health. There exists a growing body of evidence that has systematically recorded an upward trajectory in GI malignancies within the last 5 to 10 years, thus presenting a formidable menace to the health of the human population. The perturbations in GI microbiota may have a noteworthy influence on the advancement of GI cancers; however, the precise mechanisms behind this association are still not comprehensively understood. Some bacteria have been observed to support cancer development, while others seem to provide a safeguard against it. Recent studies have indicated that alterations in the composition and abundance of microbiomes could be associated with the progression of various GI cancers, such as colorectal, gastric, hepatic, and esophageal cancers. Within this comprehensive analysis, we examine the significance of microbiomes, particularly those located in the intestines, in GI cancers. Furthermore, we explore the impact of microbiomes on various treatment modalities for GI cancer, including chemotherapy, immunotherapy, and radiotherapy. Additionally, we delve into the intricate mechanisms through which intestinal microbes influence the efficacy of GI cancer treatments.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
50
|
Martin A, Jauvain M, Bergsten E, Demontant V, Lehours P, Barau C, Levy M, Rodriguez C, Sobhani I, Amiot A. Gastric microbiota in patients with gastric MALT lymphoma according to Helicobacter pylori infection. Clin Res Hepatol Gastroenterol 2024; 48:102247. [PMID: 37981222 DOI: 10.1016/j.clinre.2023.102247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Gastric Mucosa Associated Lymphoid Tissue lymphoma (GML) development is triggered by Helicobacter pylori (H. pylori) infection. Little is known about the impact of H. pylori infection on gastric microbiota. METHODS The gastric microbiota was retrospectively investigated using 16S rRNA gene sequencing in 32 patients with untreated GML (10 H. pylori-positive and 22 H. pylori-negative), 23 with remitted and 18 refractory GML and 35 controls. Differences in microbial diversity, bacterial composition and taxonomic repartition were assessed. RESULTS There was no change in diversity and bacterial composition between GML and control patients taking into account H. pylori status. Differential taxa analysis identified specific changes associated with H. pylori-negative GML: the abundances of Actinobacillus, Lactobacillus and Chryseobacterium were increased while the abundances of Veillonella, Atopobium, Leptotrichia, Catonella, Filifactor and Escherichia_Shigella were increased in control patients. In patients with remitted GML, the genera Haemophilus and Moraxella were significantly more abundant than in refractory patients, while Atopobium and Actinomyces were significantly more abundant in refractory patients. CONCLUSION Detailed analysis of the gastric microbiota revealed significant changes in the bacterial composition of the gastric mucosa in patients with GML that may have a role in gastric lymphomagenesis but not any new pathobionts.
Collapse
Affiliation(s)
- Antoine Martin
- Department of Gastroenterology, Henri-Mondor University Hospital, Universite Paris Est Creteil, AP-HP, EA7375, 51, Avenue du Marechal de Lattre de Tassigny CRETEIL, Creteil F-94010, France
| | - Marine Jauvain
- UMR1312 Bordeaux Institute of Cancer, BRIC, Université de Bordeaux, Bordeaux 33076, France; French National Reference Center for Campylobacters and Helicobacters, Bordeaux Hospital University Center, Bordeaux, France
| | - Emma Bergsten
- Department of Gastroenterology, Henri-Mondor University Hospital, Universite Paris Est Creteil, AP-HP, EA7375, 51, Avenue du Marechal de Lattre de Tassigny CRETEIL, Creteil F-94010, France
| | - Vanessa Demontant
- Genomics Platform and Virology Unit, Henri-Mondor University Hospital, AP-HP, Institut Mondor de Recherche Biomédicale, Universite Paris Est Creteil, INSERM U955, Créteil F-94010 France
| | - Philippe Lehours
- UMR1312 Bordeaux Institute of Cancer, BRIC, Université de Bordeaux, Bordeaux 33076, France; French National Reference Center for Campylobacters and Helicobacters, Bordeaux Hospital University Center, Bordeaux, France
| | - Caroline Barau
- Plateforme de Ressources Biologique, Henri-Mondor University Hospital, AP-HP, University Paris Est Creteil, F-94010, France
| | - Michael Levy
- Department of Gastroenterology, Henri-Mondor University Hospital, Universite Paris Est Creteil, AP-HP, EA7375, 51, Avenue du Marechal de Lattre de Tassigny CRETEIL, Creteil F-94010, France
| | - Christophe Rodriguez
- Genomics Platform and Virology Unit, Henri-Mondor University Hospital, AP-HP, Institut Mondor de Recherche Biomédicale, Universite Paris Est Creteil, INSERM U955, Créteil F-94010 France
| | - Iradj Sobhani
- Department of Gastroenterology, Henri-Mondor University Hospital, Universite Paris Est Creteil, AP-HP, EA7375, 51, Avenue du Marechal de Lattre de Tassigny CRETEIL, Creteil F-94010, France
| | - Aurelien Amiot
- Department of Gastroenterology, Henri-Mondor University Hospital, Universite Paris Est Creteil, AP-HP, EA7375, 51, Avenue du Marechal de Lattre de Tassigny CRETEIL, Creteil F-94010, France.
| |
Collapse
|