1
|
Yao H, Liu T, Chen Y, She L, Wu T, Liu D, Deng Y, Han Y, Chen K, Deng J, Zhang J, Chen J, Liu F. Dysregulated gastric microbial communities and functional shifts in chronic atrophic versus non-atrophic gastritis: a Helicobacter pylori-Negative observational study. BMC Gastroenterol 2025; 25:304. [PMID: 40301773 PMCID: PMC12039101 DOI: 10.1186/s12876-025-03900-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) infection was identified as a substantial risk factor for gastric cancer development, but the eradication of H. pylori did not necessarily lead to a reduction in the incidence of gastric cancer. Non-Helicobacter pylori (non-H. pylori) bacteria in the stomach are involved in the transformation of gastritis carcinoma. The aim of this study was to characterize the microbiome composition of the gastric mucosa and its functions in non-H. pylori (H. pylori-negative) patients with chronic atrophic gastritis (CAG) and chronic non-atrophic gastritis (CNAG). METHODS Fourteen CNAG samples and twenty-three CAG samples were collected. The composition of the gastric microbiome was analyzed using 16 S rDNA gene sequencing. The bioinformatic analysis was performed using alpha and beta diversity analyses, PICRUSt2, and linear discriminant analysis effect size (LEfSe). RESULTS The two groups shared the same most abundant bacterial phyla (Pseudomonadota, Bacillota, Actinomycetota, and Bacteroidota). The top 5 most abundant bacterial genera in the CAG group were Sphingomonas, Ralstonia, Brevundimonas, Methyloversatilis, and Pseudomonas. In the CNAG group, the top genera were Brevundimonas, Ralstonia, Sphingomonas, Methyloversatilis, and Acinetobacter. Differential analysis revealed distinct genera between groups: the CAG group showed enrichment in Sphingomonas, Ralstonia, Bradyrhizobium, Roseateles, and Acidithiobacillus, while the CNAG group was enriched in Brevundimonas, Rhodococcus, Hydrogenophaga, Bacteroides, and Leifsonia (p < 0.05). Sphingomonas exhibited a positive correlation with Acidithiobacillus but negative correlations with Brevundimonas, Hydrogenophaga, and Leifsonia. Pathways related to xenobiotic biodegradation, metabolism, signal transduction, cofactor/vitamin metabolism, cancer, infectious diseases, and digestive system were enriched in the CAG group. In contrast, the CNAG group showed enrichment in amino acid metabolism, translation, replication/repair, and terpenoid/polyketide metabolism. CONCLUSION Gastric mucosal microbiota dysbiosis and functional shifts are significantly associated with chronic atrophic gastritis. Taxa such as Sphingomonas and Ralstonia, enriched in CAG patients, may indicate microbial signatures associated with early atrophic transition and provide candidates for further mechanistic validation.
Collapse
Affiliation(s)
- Hong Yao
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Tingting Liu
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Yiming Chen
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Ling She
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Tingfeng Wu
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Dongsong Liu
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Yuhong Deng
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Yubin Han
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Kai Chen
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Jianmin Deng
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Jue Zhang
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China
| | - Jinfeng Chen
- The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China.
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, Guangdong, People's Republic of China.
| | - Fengbin Liu
- Lingnan Institute of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China.
- Baiyun Hospital of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
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
|
3
|
Zhou C, Bisseling TM, van der Post RS, Boleij A. The influence of Helicobacter pylori, proton pump inhibitor, and obesity on the gastric microbiome in relation to gastric cancer development. Comput Struct Biotechnol J 2024; 23:186-198. [PMID: 38075398 PMCID: PMC10704269 DOI: 10.1016/j.csbj.2023.11.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 05/11/2025] Open
Abstract
Helicobacter pylori infection is still the main risk factor for the development of gastric cancer (GC). We explore the scientific evidence for the role of the gastric microbiome beyond Helicobacter pylori (H. pylori) in gastric carcinogenesis. The composition of the gastric microbiome in healthy individuals, in presence and absence of H. pylori infection, in proton pump inhibitor (PPI)-users, obese individuals, and GC patients was investigated. Possible mechanisms for microbial involvement, limitations of available research and options for future studies are provided. A common finding amongst studies was increased levels of Streptococcus, Prevotella, Neisseria, and Actinomyces in healthy individuals or those with H. pylori-negative gastritis. In PPI-users the risk for GC increases with the treatment duration, and the gastric microbiome shifts, with the most consistent increase in the genus Streptococcus. Similarly, in obese individuals, Streptococcus was the most abundant genus, with an increased risk for cardia GC. The genera Streptococcus, Lactobacillus and Prevotella were found to be more prominent in GC patients in multiple studies. Potential mechanisms of non-H. pylori microbiota contributing to GC are linked to lipopolysaccharide production, contribution to inflammatory pathways, and the formation of N-nitroso compounds and reactive oxygen species. In conclusion, the knowledge of the gastric microbiome in GC is mainly descriptive and based on sequencing of gastric mucosal samples. For a better mechanistic understanding of microbes in GC development, longitudinal cohorts including precancerous lesions, different regions in the stomach, and subtypes of GC, and gastric organoid models for diffuse and intestinal type GC should be employed.
Collapse
Affiliation(s)
- Chengliang Zhou
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Pathology, P.O. box 9101, 6500 HB Nijmegen, the Netherlands
| | - Tanya M. Bisseling
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Gastroenterology and Hepatology, P.O. box 9101, 6500 HB Nijmegen, the Netherlands
| | - Rachel S. van der Post
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Pathology, P.O. box 9101, 6500 HB Nijmegen, the Netherlands
| | - Annemarie Boleij
- Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Pathology, P.O. box 9101, 6500 HB Nijmegen, the Netherlands
| |
Collapse
|
4
|
Huma Arya P, Vadhwana B, Tarazi M. Microbial dysbiosis in gastric cancer: Association or causation? Best Pract Res Clin Gastroenterol 2024; 72:101961. [PMID: 39645283 DOI: 10.1016/j.bpg.2024.101961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/26/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024]
Affiliation(s)
- Pallavi Huma Arya
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, White City, W12 0HS, UK.
| | - Bhamini Vadhwana
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, White City, W12 0HS, UK.
| | - Munir Tarazi
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, White City, W12 0HS, UK.
| |
Collapse
|
5
|
Onishi T. Incidence of Postoperative Diabetes Mellitus After Roux-en-Y Reconstruction for Gastric Cancer: Retrospective Single-Center Cohort Study. JMIRX MED 2024; 5:e56405. [PMID: 39149857 PMCID: PMC11337235 DOI: 10.2196/56405] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 08/17/2024]
Abstract
Background Sleeve gastrectomy is an effective surgical option for morbid obesity, and it improves glucose homeostasis. In patients with gastric cancer and type 2 diabetes mellitus (DM), gastrectomy, including total gastrectomy, is beneficial for glycemic control. Objective This study aims to clarify the effects of gastrectomy and different reconstructive techniques on the incidence of postoperative DM in patients with gastric cancer. Methods This retrospective, single-center, cohort study included 715 patients without DM who underwent total gastrectomy at the Tokyo Metropolitan Bokutoh Hospital between August 2005 and March 2019. Patients underwent reconstruction by Roux-en-Y (RY) gastric bypass or other surgical techniques (OT), with DM onset determined by hemoglobin A1c levels or medical records. Analyses included 2-sample, 2-tailed t tests; χ2 tests; and the Kaplan-Meier method with log-rank tests to compare the onset curves between the RY and OT groups, along with additional curves stratified by sex. A Swimmer plot for censoring and new-onset DM was implemented. Results Stratified data analysis compared the RY and OT reconstruction methods. The hazard ratio was 1.52 (95% CI 1.06-2.18; P=.02), which indicated a statistically significant difference in the incidence of new-onset diabetes between the RY and OT groups in patients with gastric cancer. The hazard ratio after propensity score matching was 1.42 (95% CI 1.09-1.86; P=.009). Conclusions This first-of-its-kind study provides insight into how different methods of gastric reconstruction affect postoperative diabetes. The results suggest significant differences in new-onset DM after surgery based on the reconstruction method. This research highlights the need for careful surgical planning to consider potential postoperative DM, particularly in patients with a family history of DM. Future studies should investigate the role of gut microbiota and other reconstructive techniques, such as laparoscopic jejunal interposition, in developing postoperative DM.
Collapse
Affiliation(s)
- Tatsuki Onishi
- Data Science and AI Innovation Research Promotion Center, Shiga University, 1 Chome-1-1 Bamba, Hikone, Shiga, 522-0069, Japan, 81 749 27 1030
- Department of Anesthesia, Juntendo University Shizuoka Hospital, Izunokuni, Shizuoka, Japan
- Department of Anesthesia, Kyowa Hospital, Kyoto, Japan
| |
Collapse
|
6
|
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
|
7
|
Shaheen MMA, Hroub M, Talahmeh L. Factors associated with irritable bowel syndrome and Helicobacter pylori infection: public knowledge and awareness of signs and symptoms. J Int Med Res 2024; 52:3000605241248041. [PMID: 38775336 PMCID: PMC11113039 DOI: 10.1177/03000605241248041] [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: 09/17/2023] [Accepted: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVE To investigate factors related to the risk of developing irritable bowel syndrome (IBS) or Helicobacter pylori infection. METHODS This cross-sectional, questionnaire-based study analysed the responses from participants that completed an online questionnaire, which asked about their knowledge of the causes and risk factors associated with IBS and H. pylori infection. RESULTS The study analysed responses from 230 participants: 181 females (of 227 participants; 79.7%) and 190 aged 18-40 years (of 228; 83.3%). Of the 230 participants, 40 (17.4%) had been diagnosed by a physician with IBS and 57 (24.8%) had been diagnosed with H. pylori infection. Of 226 participants, 93 (41.2%) had self-medicated with antibiotics in the past 6 months for various reasons. The overall mean ± SD knowledge score about IBS and H. pylori infection for the study cohort (n = 230) was 35.8 ± 19.2%. Wald χ2-test analysis demonstrated that chronic diseases, antibiotic use and having an endoscopy were significantly associated with developing IBS. Male sex and chronic diseases were significantly associated with H. pylori infection. Logistic regression analysis showed no relationship between IBS and H. Pylori infection. CONCLUSION Chronic diseases was the only risk factor common for IBS and H. pylori infection.
Collapse
Affiliation(s)
- Muamar M. A. Shaheen
- Department of Clinical Pharmacy and Practice, Faculty of Pharmacy and Medical Sciences, Hebron University, Hebron, West Bank, Palestine
| | - Maysaa Hroub
- Department of Clinical Pharmacy and Practice, Faculty of Pharmacy and Medical Sciences, Hebron University, Hebron, West Bank, Palestine
| | - Lana Talahmeh
- Department of Clinical Pharmacy and Practice, Faculty of Pharmacy and Medical Sciences, Hebron University, Hebron, West Bank, Palestine
| |
Collapse
|
8
|
Park CH. Unveiling the Gastrointestinal Microbiome Symphony: Insights Into Post-Gastric Cancer Treatment Microbial Patterns and Potential Therapeutic Avenues. J Gastric Cancer 2024; 24:89-98. [PMID: 38225768 PMCID: PMC10774752 DOI: 10.5230/jgc.2024.24.e4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/17/2024] Open
Abstract
This review delved into the intricate relationship between the gastrointestinal microbiome and gastric cancer, particularly focusing on post-treatment alterations, notably following gastrectomy, and the effects of anticancer therapies. Following gastrectomy, analysis of fecal samples revealed an increased presence of oral cavity aerotolerant and bile acid-transforming bacteria in the intestine. Similar changes were observed in the gastric microbiome, highlighting significant alterations in taxon abundance and emphasizing the reciprocal interaction between the oral and gastric microbiomes. In contrast, the impact of chemotherapy and immunotherapy on the gut microbiome was subtle, although discernible differences were noted between treatment responders and non-responders. Certain bacterial taxa showed promise as potential prognostic markers. Notably, probiotics emerged as a promising approach for postgastrectomy recovery, displaying the capacity to alleviate inflammation, bolster immune responses, and maintain a healthy gut microbiome. Several strains, including Bifidobacterium, Lactobacillus, and Clostridium butyricum, exhibited favorable outcomes in postoperative patients, suggesting their potential roles in comprehensive patient care. In conclusion, understanding the intricate interplay between the gastrointestinal microbiome and gastric cancer treatment offers prospects for predicting responses and enhancing postoperative recovery. Probiotics, with their positive impact on inflammation and immunity, have emerged as potential adjuncts in patient care. Continued research is imperative to fully harness the potential of microbiome-based interventions in the management of gastric cancer.
Collapse
Affiliation(s)
- Chan Hyuk Park
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea.
| |
Collapse
|
9
|
Li XJ, Gao MG, Chen XX, Rong YM, Huang LL, Huang JS. Genetically Predicted Causal Effects of Gut Microbiota and Gut Metabolites on Digestive Tract Cancer: A Two-Sample Mendelian Randomization Analysis. World J Oncol 2023; 14:558-569. [PMID: 38022400 PMCID: PMC10681779 DOI: 10.14740/wjon1737] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Background Evidence from numerous observational studies and clinical trials has linked gut microbiota and metabolites to digestive tract cancer. However, the causal effect between these factors remains uncertain. Methods Data for this study were obtained from the MiBioGen, TwinsUK Registry, and FinnGen (version R8). Two-sample Mendelian randomization analysis with inverse variance weighting method was primarily used, and the results were validated by heterogeneity analysis, pleiotropy test, and sensitivity analysis. Results At P < 5 × 10-8, our analysis identified four gut microbiotas as risk factors for digestive tract cancer and six as risk factors for colorectal cancer. Conversely, one gut microbiota exhibited protection against bile duct cancer, and two showed protective effects against stomach cancer. At P < 1 × 10-5, our investigation revealed five, six, three, eight, eight, and eight gut microbiotas as risk factors for esophageal, stomach, bile duct, liver, pancreatic, and colorectal cancers, respectively. In contrast, four, two, eight, two, two, and five gut microbiotas exhibited protective effects against these cancers. Additionally, GABA, a metabolite of gut microbiota, displayed a significant protective effect against colorectal cancer. Conclusion In conclusion, specific gut microbiota and metabolites play roles as risk factors or protective factors for digestive tract cancer, and a causal relationship between them has been established, offering novel insights into gut microbiota-mediated cancer development.
Collapse
Affiliation(s)
- Xu Jia Li
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- These authors contributed equally to this work
| | - Meng Ge Gao
- Department of Clinical Nutrition, Huadu District People’s Hospital, Southern Medical University, Guangzhou 510800, China
- These authors contributed equally to this work
| | - Xu Xian Chen
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yu Ming Rong
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ling Li Huang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jin Sheng Huang
- VIP Department, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
10
|
Chattopadhyay I, Gundamaraju R, Rajeev A. Diversification and deleterious role of microbiome in gastric cancer. Cancer Rep (Hoboken) 2023; 6:e1878. [PMID: 37530125 PMCID: PMC10644335 DOI: 10.1002/cnr2.1878] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/20/2023] [Accepted: 07/22/2023] [Indexed: 08/03/2023] Open
Abstract
Gut microbiota dictates the fate of several diseases, including cancer. Most gastric cancers (GC) belong to gastric adenocarcinomas (GAC). Helicobacter pylori colonizes the gastric epithelium and is the causative agent of 75% of all stomach malignancies globally. This bacterium has several virulence factors, including cytotoxin-associated gene A (CagA), vacuolating cytotoxin (VacA), and outer membrane proteins (OMPs), all of which have been linked to the development of gastric cancer. In addition, bacteria such as Escherichia coli, Streptococcus, Clostridium, Haemophilus, Veillonella, Staphylococcus, and Lactobacillus play an important role in the development of gastric cancer. Besides, lactic acid bacteria (LAB) such as Bifidobacterium, Lactobacillus, Lactococcus, and Streptococcus were found in greater abundance in GAC patients. To identify potential diagnostic and therapeutic interventions for GC, it is essential to understand the mechanistic role of H. pylori and other bacteria that contribute to gastric carcinogenesis. Furthermore, understanding bacteria-host interactions and bacteria-induced inflammatory pathways in the host is critical for developing treatment targets for gastric cancer.
Collapse
Affiliation(s)
| | - Rohit Gundamaraju
- ER stress and Mucosal Immunology TeamSchool of Health Sciences, University of TasmaniaLauncestonTasmaniaAustralia
| | - Ashwin Rajeev
- Department of BiotechnologyCentral University of Tamil NaduThiruvarurIndia
| |
Collapse
|
11
|
Chen J, Nie S, Qiu X, Zheng S, Ni C, Yuan Y, Gong Y. Leveraging existing 16S rRNA microbial data to identify diagnostic biomarker in Chinese patients with gastric cancer: a systematic meta-analysis. mSystems 2023; 8:e0074723. [PMID: 37787561 PMCID: PMC10654077 DOI: 10.1128/msystems.00747-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/11/2023] [Indexed: 10/04/2023] Open
Abstract
IMPORTANCE Gastric cancer is a significant and growing health problem in China. Studies have revealed significant differences in gastric microbiota between patients with gastric cancer and non-cancerous patients, suggesting that microbiota may play a role in tumorigenesis. In this meta-analysis, existing 16S rRNA microbial data were analyzed to find combinations consisting of five genera, which had good efficacy in distinguishing gastric cancer from non-cancerous patients in multiple types of samples. These results lend support to the use of microbial markers in detecting gastric cancer. Moreover, these biomarkers are plausible candidates for further mechanistic research into the role of the microbiota in tumorigenesis.
Collapse
Affiliation(s)
- Jijun Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of GI Cancer Etiology and Prevention, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Siru Nie
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of GI Cancer Etiology and Prevention, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xunan Qiu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of GI Cancer Etiology and Prevention, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuwen Zheng
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of GI Cancer Etiology and Prevention, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chuxuan Ni
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of GI Cancer Etiology and Prevention, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of GI Cancer Etiology and Prevention, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuehua Gong
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention, Liaoning Education Department, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of GI Cancer Etiology and Prevention, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
12
|
Shin WS, Xie F, Chen B, Yu J, Lo KW, Tse GMK, To KF, Kang W. Exploring the Microbiome in Gastric Cancer: Assessing Potential Implications and Contextualizing Microorganisms beyond H. pylori and Epstein-Barr Virus. Cancers (Basel) 2023; 15:4993. [PMID: 37894360 PMCID: PMC10605912 DOI: 10.3390/cancers15204993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
While previous research has primarily focused on the impact of H. pylori and Epstein-Barr virus (EBV), emerging evidence suggests that other microbial influences, including viral and fungal infections, may also contribute to gastric cancer (GC) development. The intricate interactions between these microbes and the host's immune response provide a more comprehensive understanding of gastric cancer pathogenesis, diagnosis, and treatment. The review highlights the roles of established players such as H. pylori and EBV and the potential impacts of gut bacteria, mainly Lactobacillus, Streptococcus, hepatitis B virus, hepatitis C virus, and fungi such as Candida albicans. Advanced sequencing technologies offer unprecedented insights into the complexities of the gastric microbiome, from microbial diversity to potential diagnostic applications. Furthermore, the review highlights the potential for advanced GC diagnosis and therapies through a better understanding of the gut microbiome.
Collapse
Affiliation(s)
- Wing Sum Shin
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.S.); (F.X.); (B.C.); (K.W.L.); (G.M.K.T.); (K.F.T.)
| | - Fuda Xie
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.S.); (F.X.); (B.C.); (K.W.L.); (G.M.K.T.); (K.F.T.)
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China;
- CUHK—Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China
| | - Bonan Chen
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.S.); (F.X.); (B.C.); (K.W.L.); (G.M.K.T.); (K.F.T.)
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China;
- CUHK—Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China;
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Kwok Wai Lo
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.S.); (F.X.); (B.C.); (K.W.L.); (G.M.K.T.); (K.F.T.)
| | - Gary M. K. Tse
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.S.); (F.X.); (B.C.); (K.W.L.); (G.M.K.T.); (K.F.T.)
| | - Ka Fai To
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.S.); (F.X.); (B.C.); (K.W.L.); (G.M.K.T.); (K.F.T.)
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Wei Kang
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China; (W.S.S.); (F.X.); (B.C.); (K.W.L.); (G.M.K.T.); (K.F.T.)
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China;
- CUHK—Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China
| |
Collapse
|
13
|
Dadgar N, Edlukudige Keshava V, Raj MS, Wagner PL. The Influence of the Microbiome on Immunotherapy for Gastroesophageal Cancer. Cancers (Basel) 2023; 15:4426. [PMID: 37760397 PMCID: PMC10526145 DOI: 10.3390/cancers15184426] [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: 08/08/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Immunotherapy has shown promise as a treatment option for gastroesophageal cancer, but its effectiveness is limited in many patients due to the immunosuppressive tumor microenvironment (TME) commonly found in gastrointestinal tumors. This paper explores the impact of the microbiome on the TME and immunotherapy outcomes in gastroesophageal cancer. The microbiome, comprising microorganisms within the gastrointestinal tract, as well as within malignant tissue, plays a crucial role in modulating immune responses and tumor development. Dysbiosis and reduced microbial diversity are associated with poor response rates and treatment resistance, while specific microbial profiles correlate with improved outcomes. Understanding the complex interactions between the microbiome, tumor biology, and immunotherapy is crucial for developing targeted interventions. Microbiome-based biomarkers may enable personalized treatment approaches and prediction of patient response. Interventions targeting the microbiome, such as microbiota-based therapeutics and dietary modifications, offer the potential for reshaping the gut microbiota and creating a favorable TME that enhances immunotherapy efficacy. Further research is needed to reveal the underlying mechanisms, and large-scale clinical trials will be required to validate the efficacy of microbiome-targeted interventions.
Collapse
Affiliation(s)
- Neda Dadgar
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44106, USA;
| | | | - Moses S. Raj
- Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA; (V.E.K.); (M.S.R.)
| | - Patrick L. Wagner
- Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA; (V.E.K.); (M.S.R.)
| |
Collapse
|
14
|
Li J, Wu G, Yang J, Yan J, Li D, Wang Q, Xia Y, Zhu J, Guo B, Cheng F, Sun J, Cao H, Zhang F. Pulmonary microbiota signatures adjacent to adenocarcinoma, squamous cell carcinoma and benign lesion. Front Oncol 2023; 13:1163359. [PMID: 37361591 PMCID: PMC10288182 DOI: 10.3389/fonc.2023.1163359] [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: 02/10/2023] [Accepted: 05/10/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction The occurrence and progression of lung cancer are influenced by pulmonary microbiota, yet the relationship between changes in the pulmonary microbiota and lung cancer remains unclear. Methods To investigate the correlation between pulmonary microbiota and the signature of lung lesions, we analyzed the microbial composition at sites adjacent to the stage 1 adenocarcinoma, squamous carcinoma and benign lesion tissues in 49 patients by using 16S ribosomal RNA gene sequencing. We then conducted Linear discriminant analysis, receiver operating characteristic (ROC) curve analysis and PICRUSt prediction based on 16S sequencing results. Results Overall, the microbiota composition at sites close to lung lesions showed significant differences between different lesion types. Based on the results of LEfSe analysis, Ralstonia, Acinetobacter and Microbacterium are the dominant genera of lung adenocarcinoma (LUAD), lung squamous carcinoma (LUSC) and benign lesions (BENL), respectively. Furthermore, we determined the diagnostic value of the abundance ratio of Ralstonia to Acinetobacter in adenocarcinoma patients through ROC curve analysis. The PICRUSt analysis revealed 15 remarkably different metabolic pathways in these lesion types. In LUAD patients, the increase of the pathway associated with xenobiotic biodegradation may be due to the continuous proliferation of microbe with degradation ability of xenobiotics, which implied that LUAD patients are often exposed to harmful environment. Discussion The abundance of Ralstonia was related to the development of lung cancer. By measuring the abundance of microbiota in diseased tissues, we can distinguish between different types of lesions. The differences in pulmonary microbiota between lesion types are significant in understanding the occurrence and development of lung lesions.
Collapse
Affiliation(s)
- Jinyou Li
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Gang Wu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Ju Yang
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Lifestyle-Medicine Strategy to Improve Outcome for Cancer patients (LIOC) Group, Chinese Society of Nutritional Oncology, Beijing, China
| | - Jiai Yan
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Lifestyle-Medicine Strategy to Improve Outcome for Cancer patients (LIOC) Group, Chinese Society of Nutritional Oncology, Beijing, China
| | - Dan Li
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Lifestyle-Medicine Strategy to Improve Outcome for Cancer patients (LIOC) Group, Chinese Society of Nutritional Oncology, Beijing, China
| | - Qinyue Wang
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Lifestyle-Medicine Strategy to Improve Outcome for Cancer patients (LIOC) Group, Chinese Society of Nutritional Oncology, Beijing, China
| | - Yanping Xia
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Lifestyle-Medicine Strategy to Improve Outcome for Cancer patients (LIOC) Group, Chinese Society of Nutritional Oncology, Beijing, China
| | - Jie Zhu
- Department of Infection Control, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Baoliang Guo
- School of Bioengineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Fengyue Cheng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Jing Sun
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Lifestyle-Medicine Strategy to Improve Outcome for Cancer patients (LIOC) Group, Chinese Society of Nutritional Oncology, Beijing, China
| | - Hong Cao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Lifestyle-Medicine Strategy to Improve Outcome for Cancer patients (LIOC) Group, Chinese Society of Nutritional Oncology, Beijing, China
- School of Bioengineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Feng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Nutrition, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- School of Bioengineering, Jiangnan University, Wuxi, Jiangsu, China
- School of Environment and Civil Engineering, Jiangnan University, Wuxi, Jiangsu, China
- Chinese Society of Nutritional Oncology, Beijing, China
| |
Collapse
|
15
|
Zhou S, Li C, Liu L, Yuan Q, Miao J, Wang H, Ding C, Guan W. Gastric microbiota: an emerging player in gastric cancer. Front Microbiol 2023; 14:1130001. [PMID: 37180252 PMCID: PMC10172576 DOI: 10.3389/fmicb.2023.1130001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Gastric cancer (GC) is a common cancer worldwide with a high mortality rate. Many microbial factors influence GC, of which the most widely accepted one is Helicobacter pylori (H. pylori) infection. H. pylori causes inflammation, immune reactions and activation of multiple signaling pathways, leading to acid deficiency, epithelial atrophy, dysplasia and ultimately GC. It has been proved that complex microbial populations exist in the human stomach. H. pylori can affect the abundance and diversity of other bacteria. The interactions among gastric microbiota are collectively implicated in the onset of GC. Certain intervention strategies may regulate gastric homeostasis and mitigate gastric disorders. Probiotics, dietary fiber, and microbiota transplantation can potentially restore healthy microbiota. In this review, we elucidate the specific role of the gastric microbiota in GC and hope these data can facilitate the development of effective prevention and therapeutic approaches for GC.
Collapse
Affiliation(s)
- Shizhen Zhou
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chenxi Li
- Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lixiang Liu
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qinggang Yuan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - Ji Miao
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Hao Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chao Ding
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
16
|
Peng X, Yao S, Huang J, Zhao Y, Chen H, Chen L, Yu Z. Alterations in bacterial community dynamics from noncancerous to Gastric cancer. Front Microbiol 2023; 14:1138928. [PMID: 36970687 PMCID: PMC10034189 DOI: 10.3389/fmicb.2023.1138928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
Gastric microbiome has been shown to contribute to gastric carcinogenesis, understanding how alterations in gastric microbiome is helpful to the prevention and treatment of gastric cancer (GC). However, few studies have focused on the change of microbiome during the gastric carcinogenesis. In this study, the microbiome of gastric juice samples from healthy control (HC), gastric precancerous lesions (GPL) and gastric cancer (GC) was investigated by 16S rRNA gene sequencing. Our results showed that the alpha diversity of patients with GC was significantly lower than other groups. Compared to other groups, some genera in GC group were shown to be up-regulated (e.g., Lautropia and Lactobacillus) and down-regulated (e.g., Peptostreptococcus and Parvimonas). More importantly, the emergence of Lactobacillus was closely related to the occurrence and development of GC. Moreover, the microbial interactions and networks in GPL exhibited higher connectivity, complexity and lower clustering property, while GC showed the opposite trend. Taken together, we suggest that changes in the gastric microbiome are associated with GC and perform a key function in maintaining the tumor microenvironment. Therefore, our findings will provide new ideas and references for the treatment of GC.
Collapse
Affiliation(s)
- Xuan Peng
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Siqi Yao
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jing Huang
- Department of Medical Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Yiming Zhao
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Hao Chen
- Department of Medical Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Liyu Chen
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
- Liyu Chen,
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
- *Correspondence: Zheng Yu,
| |
Collapse
|
17
|
Nikitina D, Lehr K, Vilchez-Vargas R, Jonaitis LV, Urba M, Kupcinskas J, Skieceviciene J, Link A. Comparison of genomic and transcriptional microbiome analysis in gastric cancer patients and healthy individuals. World J Gastroenterol 2023; 29:1202-1218. [PMID: 36926663 PMCID: PMC10011954 DOI: 10.3748/wjg.v29.i7.1202] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/19/2022] [Accepted: 12/21/2022] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Helicobacter pylori and the stomach microbiome play a crucial role in gastric carcinogenesis, and detailed characterization of the microbiome is necessary for a better understanding of the pathophysiology of the disease. There are two common modalities for microbiome analysis: DNA (16S rRNA gene) and RNA (16S rRNA transcript) sequencing. The implications from the use of one or another sequencing approach on the characterization and comparability of the mucosal microbiome in gastric cancer (GC) are poorly studied. AIM To characterize the microbiota of GC using 16S rRNA gene and its transcript and determine difference in the bacterial composition. METHODS In this study, 316 DNA and RNA samples extracted from 105 individual stomach biopsies were included. The study cohort consisted of 29 healthy control individuals and 76 patients with GC. Gastric tissue biopsy samples were collected from damaged mucosa and healthy mucosa at least 5 cm from the tumor tissue. From the controls, healthy stomach mucosa biopsies were collected. From all biopsies RNA and DNA were extracted. RNA was reverse transcribed into cDNA. V1-V2 region of bacterial 16S rRNA gene from all samples were amplified and sequenced on an Illumina MiSeq platform. Bray-Curtis algorithm was used to construct sample-similarity matrices abundances of taxonomic ranks in each sample type. For significant differences between groups permutational multivariate analysis of variance and Mann-Whitney test followed by false-discovery rate test were used. RESULTS Microbial analysis revealed that only a portion of phylotypes (18%-30%) overlapped between microbial profiles obtained from DNA and RNA samples. Detailed analysis revealed differences between GC and controls depending on the chosen modality, identifying 17 genera at the DNA level and 27 genera at the RNA level. Ten of those bacteria were found to be different from the control group at both levels. The key taxa showed congruent results in various tests used; however, differences in 7 bacteria taxa were found uniquely only at the DNA level, and 17 uniquely only at the RNA level. Furthermore, RNA sequencing was more sensitive for detecting differences in bacterial richness, as well as differences in the relative abundance of Reyranella and Sediminibacterium according to the type of GC. In each study group (control, tumor, and tumor adjacent) were found differences between DNA and RNA bacterial profiles. CONCLUSION Comprehensive microbial study provides evidence for the effect of choice of sequencing modality on the microbiota profile, as well as on the identified differences between case and control.
Collapse
Affiliation(s)
- Darja Nikitina
- Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas 44307, Lithuania
| | - Konrad Lehr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg 39120, Germany
| | - Ramiro Vilchez-Vargas
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg 39120, Germany
| | | | - Mindaugas Urba
- Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas 44307, Lithuania
| | - Juozas Kupcinskas
- Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas 44307, Lithuania
- Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas 44307, Lithuania
| | - Jurgita Skieceviciene
- Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas 44307, Lithuania
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg 39120, Germany
| |
Collapse
|
18
|
Mendes-Rocha M, Pereira-Marques J, Ferreira RM, Figueiredo C. Gastric Cancer: The Microbiome Beyond Helicobacter pylori. Curr Top Microbiol Immunol 2023; 444:157-184. [PMID: 38231218 DOI: 10.1007/978-3-031-47331-9_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Gastric cancer remains an important global health burden. Helicobacter pylori is the major etiological factor in gastric cancer, infecting the stomach of almost half of the population worldwide. Recent progress in microbiome research offered a new perspective on the complexity of the microbial communities of the stomach. Still, the role of the microbiome of the stomach beyond H. pylori in gastric carcinogenesis is not well understood and requires deeper investigation. The gastric bacterial communities of gastric cancer patients are distinct from those of patients without cancer, but the microbial alterations that occur along the process of gastric carcinogenesis, and the mechanisms through which microorganisms influence cancer progression still need to be clarified. Except for Epstein-Barr virus, the potential significance of the virome and of the mycobiome in gastric cancer have received less attention. This chapter updates the current knowledge regarding the gastric microbiome, including bacteria, viruses, and fungi, within the context of H. pylori-mediated carcinogenesis. It also reviews the possible roles of the local gastric microbiota, as well as the microbial communities of the oral and gut ecosystems, as biomarkers for gastric cancer detection. Finally, it discusses future perspectives and acknowledges limitations in the area of microbiome research in the gastric cancer setting, to which further research efforts should be directed. These will be fundamental not only to increase our current understanding of host-microbial interactions but also to facilitate translation of the findings into innovative preventive, diagnostic, and therapeutic strategies to decrease the global burden of gastric cancer.
Collapse
Affiliation(s)
- Melissa Mendes-Rocha
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Joana Pereira-Marques
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal
| | - Rui M Ferreira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal
| | - Ceu Figueiredo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal.
- Department of Pathology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
19
|
Zi M, Zhang Y, Hu C, Zhang S, Chen J, Yuan L, Cheng X. A literature review on the potential clinical implications of streptococci in gastric cancer. Front Microbiol 2022; 13:1010465. [PMID: 36386672 PMCID: PMC9643750 DOI: 10.3389/fmicb.2022.1010465] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/03/2022] [Indexed: 10/29/2023] Open
Abstract
Streptococcus is widely found in nature and the human body, and most species are not pathogenic. In recent years, studies have found that Streptococcus is associated with gastric cancer. Streptococcus was found to be enriched in the oral cavity, stomach and intestine of gastric cancer patients and found to be increased in gastric cancer tissues, suggesting that Streptococcus may be the pathogenic bacteria underlying gastric cancer. This review discusses the discovery of Streptococcus, the relationship between Streptococcus and gastric cancer, and the possible carcinogenic mechanism of Streptococcus and summarizes the progress of the research on the role of Streptococcus in gastric cancer to provide new ideas for the early detection, diagnosis and treatment of gastric cancer.
Collapse
Affiliation(s)
- Mengli Zi
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yanqiang Zhang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Can Hu
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shengjie Zhang
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jinxia Chen
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li Yuan
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
20
|
Bacterial Involvement in Progression and Metastasis of Adenocarcinoma of the Stomach. Cancers (Basel) 2022; 14:cancers14194886. [PMID: 36230809 PMCID: PMC9562638 DOI: 10.3390/cancers14194886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Infectious bacteria influence primary gastric carcinogenesis, organotropism, and metastatic progression by altering the microenvironment at the primary and secondary tumors. Key species include Helicobacter pylori (H. pylori) and Mycoplasma hyorhinis (M. hyorhinis). Inflammation caused by H. pylori virulence factors, such as CagA, VacA, and oipA, disrupt epithelial integrity, which allows the primary tumor to progress through the metastatic process. Evidence supports the activation of aquaporin-5 by CagA-positive H. pylori infection, promoting epithelial–mesenchymal transition via the extracellular signal-regulated kinase/mitogen-activated protein kinase (MEK/ERK) pathway, thus laying the foundation for metastatic disease. M. hyorhinis has also been implicated in gastric neoplasia via β-catenin stabilization and subsequent activation of the WNT-signaling pathway, promoting gastric cancer cell motility and inciting cancer progression. Abstract Gastric cancer metastasis is a process in which the tumor microenvironment may carry significant influence. Helicobacter pylori (H. pylori) infection is well-established as a contributor to gastric carcinoma. However, the role that these bacteria and others may play in gastric carcinoma metastasis is a current focus of study. A review of the literature was conducted to elucidate the process by which gastric adenocarcinoma metastasizes, including its ability to utilize both the lymphatic system and the venous system to disseminate. Studies that investigate the tumor microenvironment at both the primary and secondary sites were assessed in detail. H. pylori and Mycoplasma hyorhinis (M. hyorhinis) were found to be important drivers of the pathogenesis of gastric adenocarcinoma by modifying various steps in cell metastasis, including epithelial–mesenchymal transition, cell migration, and cell invasion. H. pylori is also a known driver of MALT lymphoma, which is often reversible simply with the eradication of infection. M. hyorhinis has been implicated in gastric neoplasia via β-catenin stabilization and subsequent activation of the WNT-signaling pathway, promoting gastric cancer cell motility and inciting cancer progression. Fusobacterium nucleatum (F. nucleatum) and its association with worse prognosis in diffuse-type gastric adenocarcinoma are also reviewed. Recognition of the roles that bacteria play within the metastatic cascade is vital in gastrointestinal adenocarcinoma treatment and potential reoccurrence. Further investigation is needed to establish potential treatment for metastatic gastric carcinoma by targeting the tumor microenvironment.
Collapse
|
21
|
Li Y, Huang X, Tong D, Jiang C, Zhu X, Wei Z, Gong T, Jin C. Relationships among microbiota, gastric cancer, and immunotherapy. Front Microbiol 2022; 13:987763. [PMID: 36171746 PMCID: PMC9511979 DOI: 10.3389/fmicb.2022.987763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/03/2022] [Indexed: 12/07/2022] Open
Abstract
Currently, conventional neoadjuvant therapy or postoperative adjuvant therapy, such as chemotherapy and radiation therapy, can only bring limited survival benefits to gastric cancer (GC). Median survival after palliative chemotherapy is also low, at about 8-10 months. Immunotargeting is a new option for the treatment of GC, but has not been widely replicated. The highly immunosuppressed tumor microenvironment (TME) discounts the efficacy of immunotherapy for GC. Therefore, new strategies are needed to enhance the immune response of the TME. This paper reviewed the relationship between microorganisms and GC, potential links between microorganisms and immunotherapy and research of microorganisms combined immunotherapy.
Collapse
Affiliation(s)
- Yuzhen Li
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Xiaona Huang
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Desheng Tong
- Key Laboratory of National Health and Family Planning Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Chenyu Jiang
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Xiaodan Zhu
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Zhipeng Wei
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Tingjie Gong
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Chunhui Jin
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
22
|
Comparison of the gastric microbiome in Billroth I and Roux-en-Y reconstructions after distal gastrectomy. Sci Rep 2022; 12:10594. [PMID: 35732881 PMCID: PMC9217802 DOI: 10.1038/s41598-022-14886-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022] Open
Abstract
The changes in gastric microbiota following reconstruction after gastrectomy have not been reported. This study aimed to compare the gastric microbiota following Billroth I and Roux-en-Y reconstructions after distal gastrectomy. We enrolled 71 gastrectomized patients with gastric cancer; 31 and 40 underwent Billroth I and Roux-en-Y reconstructions, respectively. During upper gastrointestinal endoscopy, gastric fluid was collected immediately before and 6 months after distal gastrectomy. Deoxyribonucleic acid isolated from each sample was evaluated using 16S ribosomal ribonucleic acid metagenomic analysis. Analysis revealed that the gastric microbiota’s species richness (expressed as the alpha diversity) was significantly lower after than before distal gastrectomy (operational taxonomic units, p = 0.001; Shannon index, p = 0.03). The interindividual diversity (beta diversity) was significantly different before and after distal gastrectomy (unweighted UniFrac distances, p = 0.04; weighted UniFrac distances, p = 0.001; Bray–Curtis, p = 0.001). Alpha and beta diversity were not significantly different between Billroth I and Roux-en-Y reconstructions (observed operational taxonomic units, p = 0.58; Shannon index, p = 0.95; unweighted UniFrac distances, p = 0.65; weighted UniFrac distances, p = 0.67; Bray–Curtis, p = 0.63). Our study demonstrated significant differences in gastric microbiota diversity, composition, and community before and after distal gastrectomy but no difference between Billroth I and Roux-en-Y reconstruction after distal gastrectomy.
Collapse
|
23
|
Chen S, Xue X, Zhang Y, Zhang H, Huang X, Chen X, Deng G, Luo S, Gao J. Vaginal Atopobium is Associated with Spontaneous Abortion in the First Trimester: a Prospective Cohort Study in China. Microbiol Spectr 2022; 10:e0203921. [PMID: 35311570 PMCID: PMC9045190 DOI: 10.1128/spectrum.02039-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/14/2022] [Indexed: 11/20/2022] Open
Abstract
Spontaneous abortion (SA) has received more and more attention in light of its increasing incidence. However, the causes and pathogenesis of SA remain largely unknown, especially for those without any pathological features. In this study, we characterized the vaginal microbiota diversity and composition of pregnant women in their first trimester and evaluated the association between the vaginal microbiota and SA before 12 weeks of gestation. Participants' bacterial profiles were analyzed by 16S rRNA gene sequencing in the V3-V4 regions at 5-8 weeks of gestation. A total of 48 patients with SA at 12 weeks of gestation were included as the study group, while 116 women with normal pregnancies (NPs) were included as a control group. The results indicated that the richness of the vaginal microbiome in SA patients was higher (Chao1, P < 0.05) and different in composition relative to that of women with NPs (unweighted UniFrac, R = 0.15, P < 0.01; binary Jaccard, R = 0.15, P < 0.01). Furthermore, the genus Apotobium was significantly enriched in SA patients. An extreme gradient-boosting (XGBoost) analysis was able to classify Atopobium-induced SA more reliably (area under the receiver operating characteristic curve, 0.69; threshold, 0.01%). Moreover, after adjusting for potential confounders, the results showed a robust association between Apotobium and SA (as a categorical variable [<0.01%]; adjusted odds ratio, 2.9; 95% confidence interval, 1.3 to 6.5; P = 0.01). In conclusion, higher vaginal Apotobium levels were associated with SA in the first trimester. IMPORTANCE Spontaneous abortion (SA) is the most common adverse pregnancy outcome in the first trimester. The causal drivers of SA have become a substantial challenge to reveal and overcome. We hypothesize that vaginal microbial dysbiosis is associated with SA, as it was related to several female reproductive disorders in previous studies. In our study, we characterized the vaginal microbiota of patients with SA at 12 weeks of gestation as the study group, and women with normal pregnancies were enrolled as a control group. Generally, significant differences were discovered in the vaginal microbiota between the two groups. Our study also revealed that Apotobium may play an important role in the pathogenesis of SA. To our knowledge, this study is the first detailed elaboration of the vaginal microbiota composition and vaginal Apotobium in association with SA. We believe that our findings will inspire more researchers to consider dynamic changes in the vaginal microbiota as critical features for further studies of nosogenesis not only for SA but also other reproductive diseases.
Collapse
Affiliation(s)
- Si Chen
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaomeng Xue
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yingxuan Zhang
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huimin Zhang
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xuge Huang
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaofeng Chen
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Gaopi Deng
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese, Guangzhou, Guangdong China
| | - Songping Luo
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese, Guangzhou, Guangdong China
| | - Jie Gao
- Department of Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese, Guangzhou, Guangdong China
| |
Collapse
|
24
|
The Implication of Gastric Microbiome in the Treatment of Gastric Cancer. Cancers (Basel) 2022; 14:cancers14082039. [PMID: 35454944 PMCID: PMC9028069 DOI: 10.3390/cancers14082039] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Gastric cancer (GC) represents the fifth most common cancer worldwide. Recent developments in PCR and metagenomics clarify that the stomach contains a powerful microbiota. Conventional treatments for GC that include surgery, chemotherapy, and radiotherapy are not very effective. That’s why new therapeutic strategies are needed. The intestinal microbiota is involved in oncogenesis and cancer prevention, and the effectiveness of chemotherapy. Recent studies have shown that certain bacteria may enhance the effect of some traditional antineoplastic drugs and immunotherapies. Abstract Gastric cancer (GC) is one of the most common and deadly malignancies worldwide. Helicobacter pylori have been documented as a risk factor for GC. The development of sequencing technology has broadened the knowledge of the gastric microbiome, which is essential in maintaining homeostasis. Recent studies have demonstrated the involvement of the gastric microbiome in the development of GC. Therefore, the elucidation of the mechanism by which the gastric microbiome contributes to the development and progression of GC may improve GC’s prevention, diagnosis, and treatment. In this review, we discuss the current knowledge about changes in gastric microbial composition in GC patients, their role in carcinogenesis, the possible therapeutic role of the gastric microbiome, and its implications for current GC therapy.
Collapse
|
25
|
Wang H, Jiang Y, Liang Y, Wei L, Zhang W, Li L. Observation of the cervical microbiome in the progression of cervical intraepithelial neoplasia. BMC Cancer 2022; 22:362. [PMID: 35379200 PMCID: PMC8981842 DOI: 10.1186/s12885-022-09452-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Cervical microbial community in the cervical intraepithelial neoplasia and cervical cancer patients was analysed to study its composition, diversity and signalling pathways by high-throughput 16S rDNA sequencing,and the candidate genes associated with occurrence and progression of cervical intraepithelial neoplasia were screened out and the model was established to predict the evolution of cervical intraepithelial neoplasia malignant transformation from the cervical microbial genes aspect. METHODS Cervical tissues of normal, cervical intraepithelial neoplasia and cervical cancer patients without receiving any treatment were collected. The correlation between candidate genes and cervical intraepithelial neoplasia progression was initially determined by analyzing the microbial flora. Real-time fluorescence quantitative PCR was used to detect the expression of candidate genes in different cervical tissues, ROC curve and logistic regression was used to analyse and predict the risk factors related to the occurrence and progression of cervical intraepithelial neoplasia. Finally, the early warning model of cervical intraepithelial neoplasia occurrence and progression is established. RESULTS Cervical tissues from normal, cervical intraepithelial neoplasia and cervical cancer patients were collected for microbial community high-throughput 16S rDNA sequencing. The analysis revealed five different pathways related to cervical intraepithelial neoplasia. 10 candidate genes were selected by further bioinformatics analysis and preliminary screening. Real time PCR, ROC curve and Logistic regression analysis showed that human papillomavirus infection, TCT severity, ABCG2, TDG, PCNA were independent risk factors for cervical intraepithelial neoplasia. We used these indicators to establish a random forest model. Seven models were built through different combinations. The model 4 (ABCG2 + PCNA + TDG) was the best early warning model for the occurrence and progression of CIN. CONCLUSIONS A total of 5 differential pathways and 10 candidate genes related to occurrence and progression of cervical intraepithelial neoplasia were found in cervical microbial community. This study firstly identified the genes from cervical microbial community that play an important role in the occurrence and progression of cervical intraepithelial neoplasia. At the same time, the early warning model including ABCG2 + PCNA+TDG genes provided a new idea and target for clinical prediction and blocking the evolution of cervical intraepithelial neoplasia malignant transformation from the aspect of cervical microbiological related genes.
Collapse
Affiliation(s)
- He Wang
- Department of gynecologic oncology, Guangxi Medical University Cancer Hospital, 71 He Di Road, Nanning, 530021, Guangxi, China
| | - Yanming Jiang
- Department of Obstetrics and Gynecology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yuejuan Liang
- Department of Obstetrics and Gynecology, Liuzhou People's Hospital, Liuzhou, China
| | - Lingjia Wei
- Department of Obstetrics and Gynecology, Guangxi Medical University, Nanning, China
| | - Wei Zhang
- Department of gynecologic oncology, Guangxi Medical University Cancer Hospital, 71 He Di Road, Nanning, 530021, Guangxi, China
| | - Li Li
- Department of gynecologic oncology, Guangxi Medical University Cancer Hospital, 71 He Di Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
26
|
Sun QH, Zhang J, Shi YY, Zhang J, Fu WW, Ding SG. Microbiome changes in the gastric mucosa and gastric juice in different histological stages of Helicobacter pylori-negative gastric cancers. World J Gastroenterol 2022; 28:365-380. [PMID: 35110955 PMCID: PMC8771614 DOI: 10.3748/wjg.v28.i3.365] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The gastric microbiota in patients with gastric cancer (GC) has received increasing attention, but the profiling of the gastric microbiome through the histological stages of gastric tumorigenesis remains poorly understood, especially for patients with Helicobacter pylori-negative GC (HPNGC).
AIM To characterize microbial profiles of gastric mucosa and juice for HPNGC carcinogenesis and identify distinct taxa in precancerous lesions.
METHODS The 16S rRNA gene analysis was performed on gastric mucosa from 134 Helicobacter pylori-negative cases, including 56 superficial gastritis (SG), 9 atrophic gastritis (AG), 27 intestinal metaplasia (IM), 29 dysplasia (Dys), and 13 GC cases, to investigate differences in gastric microbial diversity and composition across the disease stages. In addition, paired gastric mucosa and juice samples from 18 SG, 18 IM, and 18 Dys samples were analyzed. α-Diversity was measured by Shannon and Chao1 indexes, and β-diversity was calculated using partial least squares discrimination analysis (PLS-DA). Differences in the microbial composition across disease stages in different sample types were assessed using the linear discriminant analysis effect size.
RESULTS The diversity and composition of the bacterial microbiota in the gastric mucosa changed progressively across stages of gastric carcinogenesis. The diversity of the gastric mucosa microbiota was found to be significantly lower in the IM and Dys groups than in the SG group, and the patients with GC had the lowest bacterial community richness (P < 0.05). Patients with IM and those with Dys had similar gastric mucosa microbiota profiles with Ralstonia and Rhodococcus as the predominant genera. Microbial network analysis showed that there was increasing correlation strength between IM and Dys (|correlation threshold|≥ 0.5, P < 0.05). GC and its precancerous lesions have distinguishable bacterial taxa; our results identified HPNGC-associated bacteria Streptococcaceae and Lactobacillaceae (P < 0.05). Additionally, across precancerous lesion stages from AG to Dys in Helicobacter pylori-negative patients, Burkholderiaceae abundance continuously increased, while Streptococcaceae and Prevotellaceae abundance presented a continuous downward trend. Furthermore, the microbial diversity was higher in gastric juice (P < 0.001) than in the mucosa, while PLS-DA revealed a statistically significant difference between the two groups (ANOSIM, P = 0.001). A significant difference in the microbial structure was identified, with Proteobacteria being more prevalent in the gastric mucosa and Firmicutes being more abundant in gastric juice.
CONCLUSION Our results provide insights into potential taxonomic biomarkers for HPNGC and its precancerous stages and assist in predicting the prognosis of IM and Dys based on the mucosal microbiota profile.
Collapse
Affiliation(s)
- Qing-Hua Sun
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Yan-Yan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 10019, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Wei-Wei Fu
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Shi-Gang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
27
|
Yang Y, Ji R, Zhao X, Cao X, Wang Q, Jiang Q, Zhang Y, Zheng W, Wu X, Yang A. Alterations in Gastric Mucosal Microbiota in Gastric Carcinogenesis: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2021; 8:754959. [PMID: 34926502 PMCID: PMC8678046 DOI: 10.3389/fmed.2021.754959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Abstract
Background: The gastric microbiota profile alters during gastric carcinogenesis. We aimed to identify the alterations in the alpha diversity and relative abundance of bacterial phyla and genera of gastric microbiota in the development of gastric cancer (GC). Methods: The systematic review was performed based on a published protocol with the registration number CRD42020206973. We searched through PubMed, EMBASE and Cochrane databases, as well as conference proceedings and references of review articles (May 2021) for observational studies reporting either the relative abundance of bacterial phyla or genera, or alpha diversity indexes in both GC and non-cancer groups. Selection of studies and data extraction were performed independently by two researchers, with disagreements resolved through discussion. Risk of bias was assessed using the self-modified Newcastle-Ottawa Scale. Results of random-effects meta-analyses were presented as mean differences (MD). Results: Our systematic review included 751 GC patients and 792 non-cancer patients from 14 case-control studies. Gastric cancer group had fewer operational taxonomic units (OTUs) (MD = -68.52, 95%CI: -126.65 to -10.39) and a lower Simpson index (MD = -0.13, 95%CI: -0.20 to -0.07) compared with non-cancer group. At the phylum level, gastric cancer group had a higher abundance of Firmicutes (MD = 7.11, 95%CI: 1.76 to 12.46). At the genus level, Streptococcus (MD = 3.03, 95%CI: 0.07 to 6.00) and Lactobacillus (MD = 5.15, 95%CI: 1.27 to 9.04) were found to be enriched in GCgroup. The relative abundance of the rest bacterial phyla or genera analyzed in our study did not significantly differ between two groups. Subgroup analyses indicated that the source of samples was the major source of interstudy heterogeneity. Conclusion: This systematic review suggested that gastric microbiota dysbiosis occurred in gastric carcinogenesis, with alpha diversity declined and microbiota composition altered.
Collapse
Affiliation(s)
- Yingyun Yang
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Ruoyu Ji
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xinyu Zhao
- National Clinical Research Center for Digestive Diseases, Department of Clinical Epidemiology and Evidence-based Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinyuan Cao
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Qiang Wang
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Qingwei Jiang
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yizhen Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Weiyang Zheng
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xi Wu
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Aiming Yang
- Department of Gastroenterology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| |
Collapse
|
28
|
Tarazi M, Jamel S, Mullish BH, Markar SR, Hanna GB. Impact of gastrointestinal surgery upon the gut microbiome: A systematic review. Surgery 2021; 171:1331-1340. [PMID: 34809971 DOI: 10.1016/j.surg.2021.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND There is evidence from preclinical models that the gut microbiome may impact outcomes from gastrointestinal surgery, and that surgery may alter the gut microbiome. However, the extent to which gastrointestinal surgery modulates the gut microbiome in clinical practice is currently poorly defined. This systematic review aims to evaluate the changes observed in the gut microbiome after gastrointestinal surgery. METHODS A systematic review and meta-analysis were conducted according to the PRISMA guidelines by screening EMBASE, MEDLINE/PubMed, Web of Science, and CENTRAL for comparative studies meeting the predetermined inclusion criteria. The primary outcome was the difference between pre and postoperative bacterial taxonomic composition and diversity metrics among patients receiving gastrointestinal surgery. RESULTS In total, 33 studies were identified including 6 randomized controlled trials and 27 prospective cohort studies reporting a total of 968 patients. Gastrointestinal surgery was associated with an increase in α diversity and a shift in β diversity postoperatively. Multiple bacterial taxa were identified to consistently trend toward an increase or decrease postoperatively. A difference in microbiota across geographic provenance was also observed. There was a distinct lack of studies showing correlation with clinical outcomes or performing microbiome functional analysis. Furthermore, there was a lack of standardization in sampling, analytical methodology, and reporting. CONCLUSION This review highlights changes in bacterial taxa associated with gastrointestinal surgery. There is a need for standardization of microbial analysis methods and reporting of results to allow interstudy comparison. Further adequately powered multicenter studies are required to better assess variation in microbial changes and its potential associations with clinical outcomes.
Collapse
Affiliation(s)
- Munir Tarazi
- Department of Surgery and Cancer, Imperial College London, UK. https://www.twitter.com/TaraziMunir
| | - Sara Jamel
- Department of Surgery and Cancer, Imperial College London, UK
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, UK. https://www.twitter.com/bhmullish
| | - Sheraz R Markar
- Department of Surgery and Cancer, Imperial College London, UK; Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden. https://www.twitter.com/MarkarSheraz
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, UK.
| |
Collapse
|
29
|
Shen G, Wu L, Jia H, Liu J. The feasibility of modified pancreatogastrostomy in vivo and its effect on intestinal microecology. Am J Transl Res 2021; 13:10288-10297. [PMID: 34650697 PMCID: PMC8507024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
PURPOSE To evaluate the feasibility of modified binding pancreatogastrostomy (MBPA) by comparing it with traditional pancreatogastrostomy (TPA) and to determine the surgical effects on the intestinal microecology. METHODS The surgical effects on the intestinal microecology of female Bama minipigs (n = 20) were determined by measuring the expressions of the intestinal microbial proteins in the gastric juice, gastric mucosa, and feces before and after MBPA and TPA. We then constructed an integrated interaction network based on the metabolomics and 16S amplicon data, the microbiota, the metabolites, and the associated pathways. RESULTS The average time required for anastomosis was significantly lower after MBPA than after TPA, but the breaking force did not significantly differ between them. We identified 25 and 51 differentially expressed metabolites and microbiota, respectively. An interaction network was constructed using 16 metabolites (including pyruvic and lactic acids), 27 microbiota (including Ruminococcaceae_UCG-00) and six pathways (including pyruvate metabolism). CONCLUSION Anastomosis might be achieved sooner and with less pancreatic leakage using MBPA compared with TPA. Pancreatogastrostomy inhibits Ruminococcaceae activity, leading to increased expressions of pyruvic and lactic acids in the gut.
Collapse
Affiliation(s)
- Guoliang Shen
- General Surgery, Department of Hepatobiliary and Pancreatic Surgery and Minimal Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical CollegeHangzhou 310014, Zhejiang, China
| | - Luning Wu
- Thyroid Gland Breast Surgery, Dongyang People’s HospitalJinhua 322100, Zhejiang, China
| | - Hangdong Jia
- Hepatobiliary Pancreatic Surgery, Zhejiang Chinese Medical UniversityHangzhou 310000, Zhejiang, China
| | - Junwei Liu
- General Surgery, Department of Hepatobiliary and Pancreatic Surgery and Minimal Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical CollegeHangzhou 310014, Zhejiang, China
| |
Collapse
|
30
|
Comment on: "Long‑term persistence of gastric dysbiosis after eradication of Helicobacter pylori in patients who underwent endoscopic submucosal dissection for early gastric cancer. Gastric Cancer, 2020 Nov 17" by Watanabe et al. Gastric Cancer 2021; 24:978-979. [PMID: 33411059 DOI: 10.1007/s10120-020-01152-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
|
31
|
Jiao X, Zhang L, Du D, Wang L, Song Q, Liu S. Alteration of vaginal microbiota in patients with recurrent miscarriage. J OBSTET GYNAECOL 2021; 42:248-255. [PMID: 34020581 DOI: 10.1080/01443615.2021.1904851] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The aim of this study was to characterise the structure of vaginal microbiota in unexplained recurrent miscarriage (RM). The vaginal bacterial communities of 16 patients with RM and 20 healthy volunteers were sampled. Then, the microbiomes of bacterial profiles of RM patients and healthy volunteers were compared by sequencing the V3-V4 regions of the bacterial 16S ribosomal RNA gene using the Illumina MiSeq platform (Illumina, San Diego, CA). Taxonomic analysis demonstrated that abundance of Lactobacillus and Gardnerella were significantly different between the RM and control groups. Furthermore, at the genus level, Lactobacillus was the most dominant genus in the two groups. Statistically significant differences were observed in three genera between RM and control groups. In the control group, two bacterial taxa were significantly more abundant (Lactobacillus and Gardnerella), while only one taxon was overrepresented in the RM group (Atopobium). These present findings provide experimental evidence supporting vaginal microbiota dysbiosis in women with RM.Impact statementWhat is already known on this subject? Currently, bacterial vaginosis is thought to be mainly due to the vaginal dysbacteriosis, which can induce unexplained recurrent miscarriage, premature rupture of membranes, low birth weight premature birth, premature birth, chorioamnionitis and series of diseases.What do the results of this study add? The current study demonstrated that Lactobacillus and Gardnerella were significantly decreased in RM patients compared to healthy control, while Atopobium was overrepresented in the RM group.What are the implications of these findings for clinical practice and/or further research? Clinically, women with RM might benefit from vaginal microbiota treatment, adjuvant therapy with Lactobacillus-based live biotherapeutics.
Collapse
Affiliation(s)
- Xuejuan Jiao
- Gynaecology and obstetrics, First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Lanling Zhang
- Gynaecology and obstetrics, First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Danli Du
- Gynaecology and obstetrics, First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Lingling Wang
- Gynaecology and obstetrics, First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Qianqian Song
- Gynaecology and obstetrics, First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Shuyu Liu
- Gynaecology and obstetrics, First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| |
Collapse
|
32
|
Palrasu M, Zaika E, El-Rifai W, Que J, Zaika AI. Role of Bacterial and Viral Pathogens in Gastric Carcinogenesis. Cancers (Basel) 2021; 13:1878. [PMID: 33919876 PMCID: PMC8070847 DOI: 10.3390/cancers13081878] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/02/2021] [Accepted: 04/11/2021] [Indexed: 01/10/2023] Open
Abstract
Gastric cancer (GC) is one of the deadliest malignancies worldwide. In contrast to many other tumor types, gastric carcinogenesis is tightly linked to infectious events. Infections with Helicobacter pylori (H. pylori) bacterium and Epstein-Barr virus (EBV) are the two most investigated risk factors for GC. These pathogens infect more than half of the world's population. Fortunately, only a small fraction of infected individuals develops GC, suggesting high complexity of tumorigenic processes in the human stomach. Recent studies suggest that the multifaceted interplay between microbial, environmental, and host genetic factors underlies gastric tumorigenesis. Many aspects of these interactions still remain unclear. In this review, we update on recent discoveries, focusing on the roles of various gastric pathogens and gastric microbiome in tumorigenesis.
Collapse
Affiliation(s)
- Manikandan Palrasu
- Department of Surgery, University of Miami, Miami, FL 33136, USA; (M.P.); (E.Z.); (W.E.-R.)
| | - Elena Zaika
- Department of Surgery, University of Miami, Miami, FL 33136, USA; (M.P.); (E.Z.); (W.E.-R.)
| | - Wael El-Rifai
- Department of Surgery, University of Miami, Miami, FL 33136, USA; (M.P.); (E.Z.); (W.E.-R.)
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33136, USA
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA;
| | - Alexander I. Zaika
- Department of Surgery, University of Miami, Miami, FL 33136, USA; (M.P.); (E.Z.); (W.E.-R.)
- Department of Veterans Affairs, Miami VA Healthcare System, Miami, FL 33136, USA
| |
Collapse
|
33
|
Yang J, Zhou X, Liu X, Ling Z, Ji F. Role of the Gastric Microbiome in Gastric Cancer: From Carcinogenesis to Treatment. Front Microbiol 2021; 12:641322. [PMID: 33790881 PMCID: PMC8005548 DOI: 10.3389/fmicb.2021.641322] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/22/2021] [Indexed: 01/10/2023] Open
Abstract
The development of sequencing technology has expanded our knowledge of the human gastric microbiome, which is now known to play a critical role in the maintenance of homeostasis, while alterations in microbial community composition can promote the development of gastric diseases. Recently, carcinogenic effects of gastric microbiome have received increased attention. Gastric cancer (GC) is one of the most common malignancies worldwide with a high mortality rate. Helicobacter pylori is a well-recognized risk factor for GC. More than half of the global population is infected with H. pylori, which can modulate the acidity of the stomach to alter the gastric microbiome profile, leading to H. pylori-associated diseases. Moreover, there is increasing evidence that bacteria other than H. pylori and their metabolites also contribute to gastric carcinogenesis. Therefore, clarifying the contribution of the gastric microbiome to the development and progression of GC can lead to improvements in prevention, diagnosis, and treatment. In this review, we discuss the current state of knowledge regarding changes in the microbial composition of the stomach caused by H. pylori infection, the carcinogenic effects of H. pylori and non-H. pylori bacteria in GC, as well as the potential therapeutic role of gastric microbiome in H. pylori infection and GC.
Collapse
Affiliation(s)
- Jinpu Yang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinxin Zhou
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaosun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Ji
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Pereira-Marques J, Ferreira RM, Machado JC, Figueiredo C. The influence of the gastric microbiota in gastric cancer development. Best Pract Res Clin Gastroenterol 2021; 50-51:101734. [PMID: 33975676 DOI: 10.1016/j.bpg.2021.101734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 01/31/2023]
Abstract
Colonization of the stomach by Helicobacter pylori is the trigger for a series of gastric mucosal changes that culminate in gastric cancer. Infection with this bacterium is considered the major risk factor for this malignancy. The introduction of high-throughput sequencing technologies coupled to advanced computational pipelines offered an improved understanding of the microbiome, and it is now currently accepted that, besides H. pylori, the stomach harbours a complex microbial community. While it is well established that H. pylori plays a central role in gastric carcinogenesis, the significance of the non-H. pylori microbiota is yet to be clarified. This review will address the state of the art on the relationship between the gastric microbiota and gastric cancer development, and identify areas where additional research is needed before translating microbiome research into preventive and therapeutic strategies to reduce gastric cancer burden.
Collapse
Affiliation(s)
- Joana Pereira-Marques
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal.
| | - Rui M Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal.
| | - Jose C Machado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal; Department of Pathology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200 - 319, Porto, Portugal.
| | - Ceu Figueiredo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal; Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135, Porto, Portugal; Department of Pathology, Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200 - 319, Porto, Portugal.
| |
Collapse
|
35
|
Barra WF, Sarquis DP, Khayat AS, Khayat BCM, Demachki S, Anaissi AKM, Ishak G, Santos NPC, Dos Santos SEB, Burbano RR, Moreira FC, de Assumpção PP. Gastric Cancer Microbiome. Pathobiology 2021; 88:156-169. [PMID: 33588422 DOI: 10.1159/000512833] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Identifying a microbiome pattern in gastric cancer (GC) is hugely debatable due to the variation resulting from the diversity of the studied populations, clinical scenarios, and metagenomic approach. H. pylori remains the main microorganism impacting gastric carcinogenesis and seems necessary for the initial steps of the process. Nevertheless, an additional non-H. pylori microbiome pattern is also described, mainly at the final steps of the carcinogenesis. Unfortunately, most of the presented results are not reproducible, and there are no consensual candidates to share the H. pylori protagonists. Limitations to reach a consistent interpretation of metagenomic data include contamination along every step of the process, which might cause relevant misinterpretations. In addition, the functional consequences of an altered microbiome might be addressed. Aiming to minimize methodological bias and limitations due to small sample size and the lack of standardization of bioinformatics assessment and interpretation, we carried out a comprehensive analysis of the publicly available metagenomic data from various conditions relevant to gastric carcinogenesis. Mainly, instead of just analyzing the results of each available publication, a new approach was launched, allowing the comprehensive analysis of the total sample amount, aiming to produce a reliable interpretation due to using a significant number of samples, from different origins, in a standard protocol. Among the main results, Helicobacter and Prevotella figured in the "top 6" genera of every group. Helicobacter was the first one in chronic gastritis (CG), gastric cancer (GC), and adjacent (ADJ) groups, while Prevotella was the leader among healthy control (HC) samples. Groups of bacteria are differently abundant in each clinical situation, and bacterial metabolic pathways also diverge along the carcinogenesis cascade. This information may support future microbiome interventions aiming to face the carcinogenesis process and/or reduce GC risk.
Collapse
Affiliation(s)
| | | | - André Salim Khayat
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Brazil.,Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | | | - Samia Demachki
- Unidade Laboratorial de Anatomia Patológica, Universidade Federal do Pará, Belém, Brazil
| | - Ana Karyssa Mendes Anaissi
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Brazil.,Unidade Laboratorial de Anatomia Patológica, Universidade Federal do Pará, Belém, Brazil
| | - Geraldo Ishak
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Brazil.,Serviço de Cirurgia Geral e do Aparelho Digestivo, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | | | | | - Rommel Rodriguez Burbano
- Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil.,Hospital Ophir Loyola, Belém, Brazil
| | | | - Paulo Pimentel de Assumpção
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém, Brazil, .,Serviço de Cirurgia Geral e do Aparelho Digestivo, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil,
| |
Collapse
|
36
|
Horvath A, Bausys A, Sabaliauskaite R, Stratilatovas E, Jarmalaite S, Schuetz B, Stiegler P, Bausys R, Stadlbauer V, Strupas K. Distal Gastrectomy with Billroth II Reconstruction is Associated with Oralization of Gut Microbiome and Intestinal Inflammation: A Proof-of-Concept Study. Ann Surg Oncol 2021; 28:1198-1208. [PMID: 32504369 PMCID: PMC7801296 DOI: 10.1245/s10434-020-08678-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Subtotal gastrectomy with Billroth II reconstruction (SGB2) results in increased gastric pH and diminished gastric barrier. Increased gastric pH following PPI therapy has an impact on the gut microbiome, intestinal inflammation, and possibly patient health. If similar changes are present after SGB2, these can be relevant for patient health and long-term outcomes after surgery. The aim of the study is to investigate whether SGB2 is associated with specific changes in gut microbiome composition and intestinal inflammation. PATIENTS AND METHODS This cross-sectional proof-of-concept study includes patients after SGB2 (n = 14) for early gastric cancer and their nongastrectomized in-house relatives as controls (n = 8). Fecal microbiome composition, intestinal inflammation (fecal calprotectin), gut permeability (DAO, LBP, sCD14), systemic inflammation (CRP) markers, and gastrointestinal symptoms are investigated. This study is registered at ClinicalTrials.gov (NCT03418428). RESULTS Microbiome oralization following SGB2 was defined by an increase in Escherichia-Shigella, Enterococcus, Streptococcus, and other typical oral cavity bacteria (Veillonella, Oribacterium, and Mogibacterium) abundance. The fecal calprotectin was increased in the SGB2 group [100.9 (52.1; 292) vs. 25.8 (17; 66.5); p = 0.014], and calprotectin levels positively correlated with the abundance of Streptococcus (rs = 0.639; padj = 0.023). Gastrointestinal symptoms in SGB2 patients were associated with distinct taxonomic changes of the gut microbiome. CONCLUSIONS SGB2 is associated with oralization of the gut microbiome; intestinal inflammation and microbiome changes were associated with gastrointestinal symptoms. These novel findings may open gut microbiome as a new target for therapy to improve quality of life and general patient health in long-term survivors after SGB2.
Collapse
Affiliation(s)
- Angela Horvath
- Department of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria
| | - Augustinas Bausys
- Department of Abdominal Surgery and Oncology, National Cancer Institute, Vilnius, Lithuania.
- Clinic of Gastroenterology, Nephrourology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania.
- Department of Transplantation Surgery, Medical University of Graz, Graz, Austria.
| | | | | | | | | | - Philipp Stiegler
- Department of Transplantation Surgery, Medical University of Graz, Graz, Austria
| | - Rimantas Bausys
- Department of Abdominal Surgery and Oncology, National Cancer Institute, Vilnius, Lithuania
- Clinic of Gastroenterology, Nephrourology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Vanessa Stadlbauer
- Department of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria
| | - Kestutis Strupas
- Clinic of Gastroenterology, Nephrourology and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
37
|
Watanabe T, Nadatani Y, Suda W, Higashimori A, Otani K, Fukunaga S, Hosomi S, Tanaka F, Nagami Y, Taira K, Tanigawa T, Nakatsu G, Hattori M, Fujiwara Y. Long-term persistence of gastric dysbiosis after eradication of Helicobacter pylori in patients who underwent endoscopic submucosal dissection for early gastric cancer. Gastric Cancer 2021; 24:710-720. [PMID: 33201352 PMCID: PMC8065006 DOI: 10.1007/s10120-020-01141-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastric microbiome, other than Helicobacter pylori, plays a role in the tumorigenesis of gastric cancer (GC). Patients who undergo endoscopic submucosal dissection for early GC have a high risk of developing metachronous GC even after successful eradication of H. pylori. Thus, we investigated the microbial profiles and associated changes in such patients after the eradication of H. pylori. METHODS A total of 19 H. pylori-infected patients with early GC who were or to be treated by endoscopic resection, with paired biopsy samples at pre- and post-eradication therapy, were retrospectively enrolled. Ten H. pylori-negative patients were enrolled as controls. Biopsy samples were analyzed using 16S rRNA sequencing. RESULTS H. pylori-positive patients exhibited low richness and evenness of bacteria with the deletion of several genera, including Blautia, Ralstonia, Faecalibacterium, Methylobacterium, and Megamonas. H. pylori eradication partially restored microbial diversity, as assessed during a median follow-up at 13 months after eradication therapy. However, post-eradication patients had less diversity than that in the controls and possessed a lower abundance of the five genera mentioned above. The eradication of H. pylori also altered the bacterial composition, but not to the same extent as that in controls. The microbial communities could be clustered into three separate groups: H. pylori-negative, pre-eradication, and post-eradication. CONCLUSION Changes in dysbiosis may persist long after the eradication of H. pylori in patients with a history of GC. Dysbiosis may be involved in the development of both primary and metachronous GC after the eradication of H. pylori in such patients.
Collapse
Affiliation(s)
- Toshio Watanabe
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Yuji Nadatani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Sciences Laboratory for Microbiome Sciences, Yokohama, Kanagawa Japan
| | - Akira Higashimori
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Koji Otani
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Shusei Fukunaga
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Shuhei Hosomi
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Fumio Tanaka
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Yasuaki Nagami
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Koichi Taira
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| | - Tetsuya Tanigawa
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan ,Department of Gastroenterology, Osaka City Juso Hospital, Osaka, Japan
| | - Geicho Nakatsu
- Department of Immunology and Infectious Diseases/Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA USA
| | - Masahira Hattori
- RIKEN Center for Integrative Medical Sciences Laboratory for Microbiome Sciences, Yokohama, Kanagawa Japan ,Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yasuhiro Fujiwara
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi Abeno-ku, Osaka, Japan
| |
Collapse
|
38
|
Throat Microbial Community Structure and Functional Changes in Postsurgery Laryngeal Carcinoma Patients. Appl Environ Microbiol 2020; 86:AEM.01849-20. [PMID: 33008819 DOI: 10.1128/aem.01849-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
The microbial community structure in the throat and its shift after laryngectomy in laryngeal squamous cell carcinoma (LSCC) patients were investigated. Thirty swab samples taken prior to laryngectomy (SLC), 18 samples 1 week after laryngectomy (SLCA1w), and 30 samples 24 weeks after laryngectomy (SLCA24w) from 30 LSCC patients were examined. Microbial diversity was profiled through sequencing the V3-V4 variable region of the 16S rRNA gene. Quantitative real-time PCR (qPCR) was used to validate the 16S rRNA sequence data for the V3-V4 region. The community structure and function of throat microbiota were assessed by PICRUSt (phylogenetic investigation of communities by reconstruction of unobserved states) analysis. Both alpha and beta diversity results showed significant differences in the throat microbiota of LSCC patients before and after laryngectomy (P < 0.05). The drinking index of the SLC group was positively associated with the genus abundance of Prevotella (P < 0.05). The SLCA1w group had lower abundances of Fusobacterium, Leptotrichia, Lachnoanaerobaculum, and Veillonella than the SLC group (P < 0.05). The SLCA24w group had higher abundances of Streptococcus and Leptotrichia as well as lower abundances of Fusobacterium and Alloprevotella than the SLC group (P < 0.05). The throat microbiomes of the SLC group could be implicated in human cancer signaling pathways, as evidenced by PICRUSt analysis (P < 0.05). Our study clarifies alterations in throat microbial community structure and function in LSCC patients during the perioperative period and postoperative recovery period.IMPORTANCE Laryngeal squamous cell carcinoma greatly impacts patients' lives, and noninvasive means of prognostic assessment are valuable in determining the effectiveness of laryngectomy. We set out to study the microbial structure changes in the throat before and after laryngectomy and found the gene functions of several throat bacteria to be associated with human cancer signaling pathways. Our findings may offer insights into the disease management of patients with laryngeal squamous cell carcinoma. We hope to provide a means of using molecular mechanisms to improve the prognosis of laryngeal cancer treatment and to facilitate relevant research.
Collapse
|
39
|
Severe preeclampsia is associated with a higher relative abundance of Prevotella bivia in the vaginal microbiota. Sci Rep 2020; 10:18249. [PMID: 33106556 PMCID: PMC7588441 DOI: 10.1038/s41598-020-75534-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
We sought to compare the vaginal microbiota profiles of Taiwanese women with severe preeclampsia (SPE) and normotensive control pregnancies. In a discovery cohort, vaginal swab samples and paired blood specimens were simultaneously obtained at the time of caesarean delivery from 30 women with SPE and 30 controls. The composition of vaginal microbiota was characterised by 16S ribosomal RNA gene sequencing of the V3–V4 region. Results were subsequently validated by real-time qPCR. We sought confirmation of our findings in an expanded cohort consisting of 58 women with SPE and 55 controls. In both the discovery and confirmation cohorts, women with SPE had higher relative abundance of Prevotella bivia in their vaginal microbial community (P = 0.006 and 0.011, respectively). Plasma levels of tumour necrosis factor alpha (TNF-α) were higher when compared with controls (P = 0.031) in the confirmation cohort. Three variables (vaginal Prevotella bivia, plasma TNF-α, and body mass index [BMI]) were included in a prediction panel for SPE. Of these, BMI was the most predictive variable. The area under the curve (AUC) of predicted probability values for the three-variable panel revealed that it can discriminate between SPE and normotensive pregnancies with good accuracy (AUC = 0.797, P < 0.001). We conclude that enrichment of Prevotella bivia in vaginal microbiota, which is tightly regulated by BMI, may be involved in the pathogenesis of SPE.
Collapse
|
40
|
Nasr R, Shamseddine A, Mukherji D, Nassar F, Temraz S. The Crosstalk between Microbiome and Immune Response in Gastric Cancer. Int J Mol Sci 2020; 21:ijms21186586. [PMID: 32916853 PMCID: PMC7556019 DOI: 10.3390/ijms21186586] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/04/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the end result of a complex interplay between host genetics, environmental factors, and microbial factors. The link between gut microbiome and gastric cancer has been attributed to persistent activation of the host's immune system by gut microbiota. The end result of this dysregulated interaction between host epithelium and microbes is a state of chronic inflammation. Gut bacteria can promote anti-tumor immune responses through several mechanisms. These include triggering T-cell responses to bacterial antigens that can cross-react with tumor antigens or cause tumor-specific antigen recognition; engagement of pattern recognition receptors that mediate pro-immune or anti-inflammatory effects or via small metabolites that mediate systemic effects on the host. Here we review the role of the gut microbiome including H. pylori and non-H. pylori gastric bacteria, the immune response, and immunotherapy using checkpoint inhibitors. We also review the evidence for cross talk between the gut microbiome and immune response in gastric cancer.
Collapse
Affiliation(s)
- Rihab Nasr
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut Medical Center, Riad El Solh, Beirut 1107, Lebanon;
| | - Ali Shamseddine
- Department of Internal Medicine, Hematology/Oncology Division, American University of Beirut Medical Center, Riad El Solh, Beirut 1107, Lebanon; (A.S.); (D.M.); (F.N.)
| | - Deborah Mukherji
- Department of Internal Medicine, Hematology/Oncology Division, American University of Beirut Medical Center, Riad El Solh, Beirut 1107, Lebanon; (A.S.); (D.M.); (F.N.)
| | - Farah Nassar
- Department of Internal Medicine, Hematology/Oncology Division, American University of Beirut Medical Center, Riad El Solh, Beirut 1107, Lebanon; (A.S.); (D.M.); (F.N.)
| | - Sally Temraz
- Department of Internal Medicine, Hematology/Oncology Division, American University of Beirut Medical Center, Riad El Solh, Beirut 1107, Lebanon; (A.S.); (D.M.); (F.N.)
- Correspondence: ; Tel.: +961-137-4374
| |
Collapse
|
41
|
Sung JJY, Coker OO, Chu E, Szeto CH, Luk STY, Lau HCH, Yu J. Gastric microbes associated with gastric inflammation, atrophy and intestinal metaplasia 1 year after Helicobacter pylori eradication. Gut 2020; 69:1572-1580. [PMID: 31974133 PMCID: PMC7456733 DOI: 10.1136/gutjnl-2019-319826] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/03/2019] [Accepted: 12/14/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Helicobacter pylori is associated with gastric inflammation, precancerous gastric atrophy (GA) and intestinal metaplasia (IM). We aimed to identify microbes that are associated with progressive inflammation, GA and IM 1 year after H. pylori eradication. DESIGN A total of 587 H. pylori-positive patients were randomised to receive H. pylori eradication therapy (295 patients) or placebo (292 patients). Bacterial taxonomy was analysed on 404 gastric biopsy samples comprising 102 pairs before and after 1 year H. pylori eradication and 100 pairs before and after 1 year placebo by 16S rRNA sequencing. RESULTS Analysis of microbial sequences confirmed the eradication of H. pylori in treated group after 1 year. Principal component analysis revealed distinct microbial clusters reflected by increase in bacterial diversity (p<0.00001) after H. pylori eradication. While microbial interactions remained largely unchanged after placebo treatment, microbial co-occurrence was less in treated group. Acinetobacter lwoffii, Streptococcus anginosus and Ralstonia were enriched while Roseburia and Sphingomonas were depleted in patients with persistent inflammation 1 year after H. pylori eradication. A distinct cluster of oral bacteria comprising Peptostreptococcus, Streptococcus, Parvimonas, Prevotella, Rothia and Granulicatella were associated with emergence and persistence of GA and IM. Probiotic Faecalibacterium praustznii was depleted in subjects who developed GA following H. pylori eradication. Functional pathways including amino acid metabolism and inositol phosphate metabolism were enriched while folate biosynthesis and NOD-like receptor signalling decreased in atrophy/IM-associated gastric microbiota. CONCLUSION This study identified that gastric microbes contribute to the progression of gastric carcinogenesis after H. pylori eradication.
Collapse
Affiliation(s)
- Joseph J Y Sung
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Olabisi Oluwabukola Coker
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Eagle Chu
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Chun Ho Szeto
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Simson Tsz Yat Luk
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Harry Cheuk Hay Lau
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Jun Yu
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| |
Collapse
|
42
|
Erawijantari PP, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T, Saito Y, Fukuda S, Yachida S, Yamada T. Influence of gastrectomy for gastric cancer treatment on faecal microbiome and metabolome profiles. Gut 2020; 69:1404-1415. [PMID: 31953253 PMCID: PMC7398469 DOI: 10.1136/gutjnl-2019-319188] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/26/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Recent evidence points to the gut microbiome's involvement in postoperative outcomes, including after gastrectomy. Here, we investigated the influence of gastrectomy for gastric cancer on the gut microbiome and metabolome, and how it related to postgastrectomy conditions. DESIGN We performed shotgun metagenomics sequencing and capillary electrophoresis time-of-flight mass spectrometry-based metabolomics analyses on faecal samples collected from participants with a history of gastrectomy for gastric cancer (n=50) and compared them with control participants (n=56). RESULTS The gut microbiota in the gastrectomy group showed higher species diversity and richness (p<0.05), together with greater abundance of aerobes, facultative anaerobes and oral microbes. Moreover, bile acids such as genotoxic deoxycholic acid and branched-chain amino acids were differentially abundant between the two groups (linear discriminant analysis (LDA) effect size (LEfSe): p<0.05, q<0.1, LDA>2.0), as were also Kyoto Encyclopedia of Genes and Genomes modules involved in nutrient transport and organic compounds biosynthesis (LEfSe: p<0.05, q<0.1, LDA>2.0). CONCLUSION Our results reveal alterations of gut microbiota after gastrectomy, suggesting its association with postoperative comorbidities. The multi-omic approach applied in this study could complement the follow-up of patients after gastrectomy.
Collapse
Affiliation(s)
- Pande Putu Erawijantari
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan
| | - Sayaka Mizutani
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Hirotsugu Shiroma
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan
| | - Satoshi Shiba
- Division of Cancer Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Takeshi Nakajima
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Tokyo, Japan
| | - Taku Sakamoto
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Tokyo, Japan
| | - Yutaka Saito
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Tokyo, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Ebina, Kanagawa, Japan,Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | - Shinichi Yachida
- Division of Cancer Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan .,Department of Cancer Genome Informatics, Graduate School of Medicine/Faculty of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takuji Yamada
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan
| |
Collapse
|
43
|
Wang L, Xin Y, Zhou J, Tian Z, Liu C, Yu X, Meng X, Jiang W, Zhao S, Dong Q. Gastric Mucosa-Associated Microbial Signatures of Early Gastric Cancer. Front Microbiol 2020; 11:1548. [PMID: 32733423 PMCID: PMC7358557 DOI: 10.3389/fmicb.2020.01548] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
Alterations in the microbiome are associated with the development of gastric cancer. Our study aimed to identify dysbiotic features in early gastric cancer (EC). The gastric microbiome was assessed in EC (n = 30), advanced gastric cancer (AC) (n = 30), and chronic gastritis (CG) (n = 60). The results demonstrated significant differences in the microbial profile and composition between EC and AC, suggesting alterations associated with gastric cancer progression. Linear discriminant analysis (LDA) effect size (LEfSe) analyses identified 32 bacterial genera that were associated with EC. Functional analyses of the gastric microbiome showed that the production of urease and synthesis of bacterial flagella were weakened in EC, while the glycolysis of fructose and hydrolysis of glycosides were enhanced. A classifier based on a random forest (RF) machine learning algorithm identified a microbial signature that distinguished EC from CG or AC with high accuracy. The correct identification of the signature was further validated in independent cohorts. This signature enriched of bacteria with varied abundance, high degree of bacterial interactions and carcinogenic potentials. Constrained principal coordinate analyses revealed that the presence of Helicobacter pylori and the cagA and vacA virulence genotypes influenced the structure of the gastric microbiome. To determine the impacts of host genetic variations on the gastric microbiome, six previously reported single nucleotide polymorphisms (SNPs) were examined. The minor allele of MUC1 rs4072037 was associated with an increased abundance of Ochrobactrum. The gastric microbiome altered in EC, which might be attributed in part to host genetic variations, H. pylori infection, bacterial virulence and environmental adaptations. The identified microbial signature could serve as biomarkers for clinical assessment of gastric cancer risk in high-risk patients.
Collapse
Affiliation(s)
- Lili Wang
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yongning Xin
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jianhua Zhou
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenguang Liu
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Xinjuan Yu
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xinying Meng
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Weina Jiang
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Shoufeng Zhao
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Quanjiang Dong
- Central Laboratories, Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
44
|
Li D, He R, Hou G, Ming W, Fan T, Chen L, Zhang L, Jiang W, Wang W, Lu Z, Feng H, Geng Q. Characterization of the Esophageal Microbiota and Prediction of the Metabolic Pathways Involved in Esophageal Cancer. Front Cell Infect Microbiol 2020; 10:268. [PMID: 32676460 PMCID: PMC7333312 DOI: 10.3389/fcimb.2020.00268] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/05/2020] [Indexed: 01/02/2023] Open
Abstract
Esophageal microbiota plays important roles in esophageal cancer. Esophagectomy, as the most important therapeutic way, contributes to changes of esophageal microbiome. However, there are few studies examining the esophageal microbiome and the metabolic changes before and after esophagectomy. The present study characterized the esophageal microbiome of 17 patients with esophageal squamous cell carcinoma (ESCC), 11 patients with esophagogastric junction (EGJ) cancer, 15 patients at 9–12 months after radical esophagectomy and 16 healthy controls (HC). 16S ribosomal RNA gene sequencing was used to evaluate the microbiome and predict the metabolic pathways. Our results showed that the microbial diversity was significantly lower in ESCC, EGJ and post-ESCC groups than that in the HC group. The abundance of Fusobacteria was higher (7.01 vs. 1.12%, P = 0.039) and the abundance of Actinobacteria (1.61 vs. 4.04%) was lower in the ESCC group than that in the HC group. We found significant differences in the abundance of Bacteroidetes (20.45 vs. 9.86%, P = 0.026), Fusobacteria (7.01 vs. 1.66%, P = 0.030) between ESCC and post-ESCC groups. The results of microbial composition analysis and PICRUSt demonstrated significant differences between ESCC and HC groups. The β diversity and PICRUSt suggested that the microbial composition and metabolic pathways were similar to HC group after esophagectomy. The monitoring of the esophagus microbiota may be an essential method to predict the recurrence of tumor.
Collapse
Affiliation(s)
- Donghang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guoqiang Hou
- Department of Thoracic Surgery, Yangxin County People's Hospital, Yangxin, China
| | - Wei Ming
- Department of Thoracic Surgery, Yangxin County People's Hospital, Yangxin, China
| | - Tao Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Chen
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenyang Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haojie Feng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
45
|
Zhang X, Pan Z. Influence of microbiota on immunity and immunotherapy for gastric and esophageal cancers. Gastroenterol Rep (Oxf) 2020; 8:206-214. [PMID: 32665852 PMCID: PMC7333930 DOI: 10.1093/gastro/goaa014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
Gastric and esophageal cancers are multifactorial and multistage-involved malignancy. While the impact of gut microbiota on overall human health and diseases has been well documented, the influence of gastric and esophageal microbiota on gastric and esophageal cancers remains unclear. This review will discuss the reported alteration in the composition of gastric and esophageal microbiota in normal and disease conditions, and the potential role of dysbiosis in carcinogenesis and tumorigenesis. This review will also discuss how dysbiosis stimulates local and systemic immunity, which may impact on the immunotherapy for cancer.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Zui Pan
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
46
|
Rajilic-Stojanovic M, Figueiredo C, Smet A, Hansen R, Kupcinskas J, Rokkas T, Andersen L, Machado JC, Ianiro G, Gasbarrini A, Leja M, Gisbert JP, Hold GL. Systematic review: gastric microbiota in health and disease. Aliment Pharmacol Ther 2020; 51:582-602. [PMID: 32056247 DOI: 10.1111/apt.15650] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/09/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Helicobacter pylori is the most infamous constituent of the gastric microbiota and its presence is the strongest risk factor for gastric cancer and other gastroduodenal diseases. Although historically the healthy stomach was considered a sterile organ, we now know it is colonised with a complex microbiota. However, its role in health and disease is not well understood. AIM To systematically explore the literature on the gastric microbiota in health and disease as well as the gut microbiota after bariatric surgery. METHODS A systematic search of online bibliographic databases MEDLINE/EMBASE was performed between 1966 and February 2019 with screening in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Randomised controlled trials, cohort studies and observational studies were included if they reported next-generation sequencing derived microbiota analysis on gastric aspirate/tissue or stool samples (bariatric surgical outcomes). RESULTS Sixty-five papers were eligible for inclusion. With the exception of H pylori-induced conditions, overarching gastric microbiota signatures of health or disease could not be determined. Gastric carcinogenesis induces a progressively altered microbiota with an enrichment of oral and intestinal taxa as well as significant changes in host gastric mucin expression. Proton pump inhibitors usage increases gastric microbiota richness. Bariatric surgery is associated with an increase in potentially pathogenic proteobacterial species in patient stool samples. CONCLUSION While H pylori remains the single most important risk factor for gastric disease, its capacity to shape the collective gastric microbiota remains to be fully elucidated. Further studies are needed to explore the intricate host/microbial and microbial/microbial interplay.
Collapse
|
47
|
Achamrah N, Grigioni S, Coëffier M, Ainseba N, Déchelotte P. Gastric Necrosis After Binge Eating in Bulimia: Recovery From Eating Disorder After Total Gastrectomy. Front Psychiatry 2020; 11:741. [PMID: 32903630 PMCID: PMC7438759 DOI: 10.3389/fpsyt.2020.00741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/15/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Gastric necrosis following acute gastric dilatation is rare but more common in females with eating disorders, such as anorexia nervosa or bulimia, during which patients often alternate restriction and binge eating behaviors. CASE PRESENTATION A 37-year old female patient with a history of 15 years of bulimia nervosa was admitted to the emergency department 24 h after binge eating. Abdominal Computed Tomography imaging showed major gastric distension reaching the pelvis and compressing the digestive organs. Total gastrectomy was required because of gastric necrosis. The patient reported significant reduction in bulimic symptoms after gastrectomy. CONCLUSION We discuss here the possible mechanisms underlying this recovery, including changes in gut-derived factors that could mediate eating behavior changes.
Collapse
Affiliation(s)
- Najate Achamrah
- Nutrition Department, Rouen University Hospital Center, Rouen, France.,Normandie Univ, UNIROUEN, INSERM UMR 1073, Nutrition, Inflammation et dysfonction de l'axe Intestin-Cerveau, IRIB, Rouen, France.,Clinical Investigation Centre CIC 1404, INSERM and Rouen University Hospital, Rouen, France
| | - Sébastien Grigioni
- Nutrition Department, Rouen University Hospital Center, Rouen, France.,Normandie Univ, UNIROUEN, INSERM UMR 1073, Nutrition, Inflammation et dysfonction de l'axe Intestin-Cerveau, IRIB, Rouen, France
| | - Moïse Coëffier
- Nutrition Department, Rouen University Hospital Center, Rouen, France.,Normandie Univ, UNIROUEN, INSERM UMR 1073, Nutrition, Inflammation et dysfonction de l'axe Intestin-Cerveau, IRIB, Rouen, France.,Clinical Investigation Centre CIC 1404, INSERM and Rouen University Hospital, Rouen, France
| | - Nadjib Ainseba
- Digestive Surgery, Beauvais Hospital Center, Beauvais, France
| | - Pierre Déchelotte
- Nutrition Department, Rouen University Hospital Center, Rouen, France.,Normandie Univ, UNIROUEN, INSERM UMR 1073, Nutrition, Inflammation et dysfonction de l'axe Intestin-Cerveau, IRIB, Rouen, France.,Clinical Investigation Centre CIC 1404, INSERM and Rouen University Hospital, Rouen, France
| |
Collapse
|
48
|
Wu CY. Initiatives for a Healthy Stomach. ACTA ACUST UNITED AC 2019; 17:628-635. [PMID: 31776804 DOI: 10.1007/s11938-019-00266-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The stomach is not only a major digestive organ, but also a sophisticated endocrine organ and an essential microbial defense organ. Stomach diseases, such as peptic ulcer diseases, stomach cancer, and functional dyspepsia, are important health burden worldwide. However, the public awareness about the stomach health and diseases are relatively low. RECENT FINDINGS To promote the awareness of medical professionals and also the general population on stomach health and diseases, the healthy stomach initiatives (HSI) was established in 2012. The HSI is very active in promoting international collaboration studies and public awareness events focused on stomach health and diseases. The HSI chose October 2 as the "World Stomach Day" to promote public awareness, to invite physician involvement, to increase scientific research, to apply government support, and to get industrial sponsorship. The first World Stomach Day was celebrated in Taipei in 2018 with very successful public educational activities. To promote the study of stomach diseases and the public awareness of stomach health is an important work. The HSI and the World Stomach Day will play essential roles in this work.
Collapse
Affiliation(s)
- Chun-Ying Wu
- Institute of Biomedical Informatics, National Yang-Ming University, Taipei, Taiwan.
- Division of Translational Research and Center of Excellence for Cancer Research, Taipei Veterans General Hospital, No. 322, Section 2, Shipai Rd., Beitou District, 11267, Taipei, Taiwan.
- Department of Public Health, China Medical University, Taichung, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.
- Taiwan Microbiota Consortium, Taipei, Taiwan.
| |
Collapse
|
49
|
Microbial carcinogenesis: Lactic acid bacteria in gastric cancer. Biochim Biophys Acta Rev Cancer 2019; 1872:188309. [PMID: 31394110 DOI: 10.1016/j.bbcan.2019.07.004] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 02/08/2023]
Abstract
While Helicobacter pylori is a fundamental risk factor, gastric cancer (GC) aetiology involves combined effects of microbial (both H. pylori and non-H. pylori), host and environmental factors. Significant differences exist between the gastric microbiome of those with gastritis, intestinal metaplasia and GC, suggesting that dysbiosis in the stomach is dynamic and correlates with progression to GC. Most notably, a consistent increase in abundance of lactic acid bacteria (LAB) has been observed in GC patients including Streptococcus, Lactobacillus, Bifidobacterium and Lactococcus. This review summarises how LAB can influence GC by a number of mechanisms that include supply of exogenous lactate -a fuel source for cancer cells that promotes inflammation, angiogenesis, metastasis, epithelial-mesenchymal transition and immune evasion-, production of reactive oxygen species and N-nitroso compounds, as well as anti-H. pylori properties that enable colonization by other non-H. pylori carcinogenic pathobionts.
Collapse
|
50
|
Liang W, Yang Y, Wang H, Wang H, Yu X, Lu Y, Shen S, Teng L. Gut microbiota shifts in patients with gastric cancer in perioperative period. Medicine (Baltimore) 2019; 98:e16626. [PMID: 31464899 PMCID: PMC6736490 DOI: 10.1097/md.0000000000016626] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is one of the common malignant tumors in China, with a high morbidity and mortality. With the development and application of high-throughput sequencing technologies and metagenomics, a great quantity of studies have shown that gastrointestinal microbiota is closely related to digestive system diseases. Although some studies have reported the effect of long-term follow-up after subtotal gastrectomy on intestinal flora changes in patients with GC. However, the features of gut microbiota and their shifts in patients with GC in perioperative period remain unclear.This study was designed to characterize fecal microbiota shifts of the patients with GC before and after the radical distal gastrectomy (RDG) during their hospital staying periods. Furthermore, fecal microbiota was also compared between the GC patients and healthy individuals.Patients who were diagnosed with advanced gastric adenocarcinoma at distal stomach were enrolled in the study. The bacterial burden within fecal samples was determined using quantitative polymerase chain reaction. To analyze the diversity and composition of gut microbiota from fecal DNA of 20 GC patients and 22 healthy controls, amplicons of the 16S rRNA gene from all subjects were pyrosequenced. To study gut microbiota shifts, the fecal microbiota from 6 GC patients before and after RDG was detected and subsequently analyzed. Short-chain fatty acids were also detected by chromatography spectrometer in these 6 GC patients.RDG had a moderate effect on bacterial richness and evenness, but had pronounced effects on the composition of postoperative gut microbiota compared with preoperative group. The relative abundances of genera Akkermansia, Esherichia/Shigella, Lactobacillus, and Dialister were significant changed in perioperative period. Remarkably, higher abundances of Escherichia/Shigella, Veillonella, and Clostridium XVIII and lower abundances of Bacteroides were observed in gut microbiota of overall GC patients compared to healthy controls.This study is the first study to characterize the altered gut microbiota within fecal samples from GC patients during perioperative period, and provide a new insights on such microbial perturbations as a potential effector of perioperative period phenotype. Further research must validate these discoveries and may evaluate targeted microbiota shifts to improve outcomes in GC patients.
Collapse
Affiliation(s)
| | - Yan Yang
- Department of Laboratory, First Affiliated Hospital, School of Medicine
| | | | | | | | | | - Shengrong Shen
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | | |
Collapse
|