1
|
Mao F, Hu Z, Shi R, Zhang H, Zhang Z, Li Y, Li X, Gao P, Li J, Liu M, Liu H, Chen J. Unravelling the prognostic and operative role of intratumoural microbiota in non-small cell lung cancer: Insights from 16S rRNA and RNA sequencing. Clin Transl Med 2025; 15:e70156. [PMID: 39754314 DOI: 10.1002/ctm2.70156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Complex interrelationships between the microbiota and cancer have been identified by several studies. However, despite delineating microbial composition in non-small cell lung cancer (NSCLC), key pathogenic microbiota and their underlying mechanisms remain unclear. METHODS We performed 16S rRNA V3-V4 amplicon and transcriptome sequencing on cancerous and adjacent normal tissue samples from 30 patients with NSCLC, from which clinical characteristics and prognosis outcomes were collected. We used 16S rRNA sequencing to dissect microbial composition and perform prognosis correlations, and in conjunction with transcriptome sequencing, we determined potential mechanisms underpinning significant microbiota actions. RESULTS In comparing different sample types, we identified more pronounced beta diversity disparity between NSCLC, lung squamous cell carcinoma (LUSC) and corresponding paired normal tissues. Concurrently, LUSC and lung adenocarcinoma exhibited distinct microbial composition traits at genus levels. Subsequently, four phyla, five classes, nine orders, 17 families and 36 genera were filtered out and were related to prognosis outcomes. Intriguingly, a protective microbial cluster was identified encompassing nine genera associated with delayed disease recurrence, with functional analyses suggested that these microbiota predominantly exerted metabolism-related functions. Additionally, a harmful microbial cluster (HMC) was identified, including three genera. In this HMC and subsequent prognosis model analyses, harmful intratumoural microbiota were potentially implicated in infection, inflammation and immune regulation. Crucially, we identified a microbial genus, Peptococcus, which was as an independent, detrimental NSCLC prognostic factor and potentially impacted prognosis outcomes via tumour necrosis factor (TNF) signalling. CONCLUSIONS We identified a substantial connection between intratumoural microbiota and NSCLC prognosis outcomes. Protective microbiota primarily exerted metabolic functions, whereas harmful microbiota were mainly implicated in infection, inflammation and immune modulation. Furthermore, Peptococcus may be significant in adverse NSCLC prognoses and serve as a potential biomarker for patient management and cancer screening. KEY POINTS Four phyla, five classes, nine orders, 17 families and 36 genera have been found associated with NSCLC prognosis. We identified a protective microbial cluster associated with delayed recurrence and a harmful microbial cluster related to shorter survival and earlier recurrence. We identified Peptococcus as an independent, detrimental prognostic factor for NSCLC, potentially impacting prognosis via TNF signalling.
Collapse
Affiliation(s)
- Fuling Mao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zixuan Hu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruifeng Shi
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zihe Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuanguang Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Penghu Gao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhui Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Minghui Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Martins FP, Andrade-Silva J, Teixeira BL, Ferrari A, Christoff AP, Cruz GNF, Paladino FV, de Oliveira LFV, Hernandes C. Oral microbiome test as an alternative diagnostic tool for gastric alterations: A prospective, bicentric cross-sectional study. PLoS One 2024; 19:e0314660. [PMID: 39621633 PMCID: PMC11611075 DOI: 10.1371/journal.pone.0314660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
The human microbiome plays a pivotal role in influencing various physiological processes and maintaining overall well-being, including the gastric system. Current diagnostic tests for gastric diseases often involve invasive procedures, sampling limitations, and medication effects, leading to potential diagnostic errors and discomfort to patients. Considering the connection between oral and gastric microbiomes, this cross-sectional study aimed to assess the diagnostic potential of the oral bacterial profile in patients undergoing upper digestive endoscopy. Oral samples from 266 participants across two Brazilian sites (Belterra and Sao Paulo) were sequenced and subjected to bioinformatic analysis to identify microbiome composition across endoscopy outcome groups, exploring alpha and beta-diversity, richness, and differential abundance and prevalence. Prevotella, Haemophilus, Fusobacterium, Neisseria, and Streptococcus were the most abundant genera observed. No significant associations were found between alpha diversity profiles and endoscopy outcomes; beta diversity analyses similarly showed no correlations. Overall, the study did not establish the oral microbiome as a reliable marker for gastric health, underscoring the necessity for broader studies in the development of non-invasive diagnostic tests.
Collapse
Affiliation(s)
| | | | | | - Angelo Ferrari
- Hospital Israelita Albert Einstein, Sao Paulo, São Paulo, Brazil
| | | | | | | | | | - Camila Hernandes
- Hospital Israelita Albert Einstein, Sao Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Khalili-Tanha G, Khalili-Tanha N, Rouzbahani AK, Mahdieh R, Jasemi K, Ghaderi R, Leylakoohi FK, Ghorbani E, Khazaei M, Hassanian SM, Gataa IS, Ferns GA, Nazari E, Avan A. Diagnostic, prognostic, and predictive biomarkers in gastric cancer: from conventional to novel biomarkers. Transl Res 2024; 274:35-48. [PMID: 39260559 DOI: 10.1016/j.trsl.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Gastric cancer is a major health concern worldwide. The survival rate of Gastric cancer greatly depends on the stage at which it is diagnosed. Early diagnosis is critical for improving survival outcomes. To improve the chances of early diagnosis, regular screening tests, such as an upper endoscopy or barium swallow, are recommended for individuals at a higher risk due to factors like family history or a previous diagnosis of gastric conditions. Biomarkers can be detected and measured using non-invasive methods such as blood tests, urine tests, breath analysis, or imaging techniques. These non-invasive approaches offer many advantages, including convenience, safety, and cost-effectiveness, making them valuable tools for disease diagnosis, monitoring, and research. Biomarker-based tests have emerged as a useful tool for identifying gastric cancer early, monitoring treatment response, assessing the recurrence risk, and personalizing treatment plans. In this current review, we have explored both classical and novel biomarkers for gastric cancer. We have centralized their potential clinical application and discussed the challenges in Gastric cancer research.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nima Khalili-Tanha
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK S7N 5B4, Canada
| | | | - Ramisa Mahdieh
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kimia Jasemi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rosa Ghaderi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Elnaz Ghorbani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Elham Nazari
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
4
|
Lu J, Tong Q. From pathogenesis to treatment: the impact of bacteria on cancer. Front Microbiol 2024; 15:1462749. [PMID: 39360320 PMCID: PMC11445166 DOI: 10.3389/fmicb.2024.1462749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
The intricate relationship between cancer and bacteria has garnered increasing attention in recent years. While traditional cancer research has primarily focused on tumor cells and genetic mutations, emerging evidence highlights the significant role of microbial communities within the tumor microenvironment in cancer development and progression. This review aims to provide a comprehensive overview of the current understanding of the complex interplay between cancer and bacteria. We explore the diverse ways in which bacteria influence tumorigenesis and tumor behavior, discussing direct interactions between bacteria and tumor cells, their impact on tumor immunity, and the potential modulation of the tumor microenvironment. Additionally, we delve into the mechanisms through which bacterial metabolites and extracellular products May affect cancer pathways. By conducting a thorough analysis of the existing literature, we underscore the multifaceted and intricate relationship between bacteria and cancer. Understanding this complex interplay could pave the way for novel therapeutic approaches and preventive strategies in cancer treatment.
Collapse
Affiliation(s)
| | - Qiang Tong
- Department of Gastrointestinal Surgery I Section, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Wang Y, Wang Y, Han W, Han M, Liu X, Dai J, Dong Y, Sun T, Xu J. Intratumoral and fecal microbiota reveals microbial markers associated with gastric carcinogenesis. Front Cell Infect Microbiol 2024; 14:1397466. [PMID: 39355268 PMCID: PMC11442432 DOI: 10.3389/fcimb.2024.1397466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
Background The relationship between dysbiosis of the gastrointestinal microbiota and gastric cancer (GC) has been extensively studied. However, microbiota alterations in GC patients vary widely across studies, and reproducible diagnostic biomarkers for early GC are still lacking in multiple populations. Thus, this study aimed to characterize the gastrointestinal microbial communities involved in gastric carcinogenesis through a meta-analysis of multiple published and open datasets. Methods We analyzed 16S rRNA sequencing data from 1,642 gastric biopsy samples and 394 stool samples across 11 independent studies. VSEARCH, QIIME and R packages such as vegan, phyloseq, cooccur, and random forest were used for data processing and analysis. PICRUSt software was employed to predict functions. Results The α-diversity results indicated significant differences in the intratumoral microbiota of cancer patients compared to non-cancer patients, while no significant differences were observed in the fecal microbiota. Network analysis showed that the positive correlation with GC-enriched bacteria increased, and the positive correlation with GC-depleted bacteria decreased compared to healthy individuals. Functional analyses indicated that pathways related to carbohydrate metabolism were significantly enriched in GC, while biosynthesis of unsaturated fatty acids was diminished. Additionally, we investigated non-Helicobacter pylori (HP) commensals, which are crucial in both HP-negative and HP-positive GC. Random forest models, constructed using specific taxa associated with GC identified from the LEfSe analysis, revealed that the combination of Lactobacillus and Streptococcus included alone could effectively discriminate between GC patients and healthy individuals in fecal samples (area under the curve (AUC) = 0.7949). This finding was also validated in an independent cohort (AUC = 0.7712). Conclusions This study examined the intratumoral and fecal microbiota of GC patients from a dual microecological perspective and identified Lactobacillus, Streptococcus, Roseburia, Faecalibacterium and Phascolarctobacterium as intratumoral and intestinal-specific co-differential bacteria. Furthermore, it confirmed the validity of the combination of Lactobacillus and Streptococcus as GC-specific microbial markers across multiple populations, which may aid in the early non-invasive diagnosis of GC.
Collapse
Affiliation(s)
- Yiwen Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Yue Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Wenjie Han
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Mengzhen Han
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Xiaolin Liu
- Department of Bioinformatics, Kanghui Biotechnology Co., Ltd., Shenyang, China
| | - Jianying Dai
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Yuesheng Dong
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Tao Sun
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Oncology Medicine, Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning, China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| | - Junnan Xu
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| |
Collapse
|
6
|
Ma D, Yang M, Sun C, Cui X, Xiong G, Wang Q, Jing W, Chen H, Lv X, Liu S, Li T, Zhao Y, Han L. cGAS suppresses hepatocellular carcinoma independent of its cGAMP synthase activity. Cell Death Differ 2024; 31:722-737. [PMID: 38594443 PMCID: PMC11164996 DOI: 10.1038/s41418-024-01291-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) is a key innate immune sensor that recognizes cytosolic DNA to induce immune responses against invading pathogens. The role of cGAS is conventionally recognized as a nucleotidyltransferase to catalyze the synthesis of cGAMP upon recognition of cytosolic DNA, which leads to the activation of STING and production of type I/III interferon to fight against the pathogen. However, given that hepatocytes are lack of functional STING expression, it is intriguing to define the role of cGAS in hepatocellular carcinoma (HCC), the liver parenchymal cells derived malignancy. In this study, we revealed that cGAS was significantly downregulated in clinical HCC tissues, and its dysregulation contributed to the progression of HCC. We further identified cGAS as an immune tyrosine inhibitory motif (ITIM) containing protein, and demonstrated that cGAS inhibited the progression of HCC and increased the response of HCC to sorafenib treatment by suppressing PI3K/AKT/mTORC1 pathway in cellular and animal models. Mechanistically, cGAS recruits SH2-containing tyrosine phosphatase 1 (SHP1) via ITIM, and dephosphorylates p85 in phosphatidylinositol 3-kinase (PI3K), which leads to the suppression of AKT-mTORC1 pathway. Thus, cGAS is identified as a novel tumor suppressor in HCC via its function independent of its conventional role as cGAMP synthase, which indicates a novel therapeutic strategy for advanced HCC by modulating cGAS signaling.
Collapse
Affiliation(s)
- Dapeng Ma
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Min Yang
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Caiyu Sun
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiuling Cui
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gaozhong Xiong
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiushi Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Haiqiang Chen
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoting Lv
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shili Liu
- Department of Microbiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Li
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yunxue Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lihui Han
- Shandong Provincial Key Laboratory of Infection & Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
7
|
Raoul P, De Gaetano V, Sciaraffia G, Ormea G, Cintoni M, Pozzo C, Strippoli A, Gasbarrini A, Mele MC, Rinninella E. Gastric Cancer, Immunotherapy, and Nutrition: The Role of Microbiota. Pathogens 2024; 13:357. [PMID: 38787209 PMCID: PMC11124250 DOI: 10.3390/pathogens13050357] [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: 03/15/2024] [Revised: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the treatment of gastric cancer (GC), which still represents the third leading cause of cancer-related death in Western countries. However, ICI treatment outcomes vary between individuals and need to be optimized. Recent studies have shown that gut microbiota could represent a key influencer of immunotherapy responses. At the same time, the nutritional status and diet of GC patients are also predictive of immunotherapy treatment response and survival outcomes. The objective of this narrative review is to gather recent findings about the complex relationships between the oral, gastric, and gut bacterial communities, dietary factors/nutritional parameters, and immunotherapy responses. Perigastric/gut microbiota compositions/functions and their metabolites could be predictive of response to immunotherapy in GC patients and even overall survival. At the same time, the strong influence of diet on the composition of the microbiota could have consequences on immunotherapy responses through the impact of muscle mass in GC patients during immunotherapy. Future studies are needed to define more precisely the dietary factors, such as adequate daily intake of prebiotics, that could counteract the dysbiosis of the GC microbiota and the impaired nutritional status, improving the clinical outcomes of GC patients during immunotherapy.
Collapse
Affiliation(s)
- Pauline Raoul
- Clinical Nutrition Unit, Department of Medical and Abdominal Surgery and Endocrine-Metabolic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy (M.C.); (M.C.M.)
| | - Valeria De Gaetano
- School of Specialization in Internal Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy; (V.D.G.); (G.S.)
| | - Gianmario Sciaraffia
- School of Specialization in Internal Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy; (V.D.G.); (G.S.)
| | - Ginevra Ormea
- Degree Course in Pharmacy, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Marco Cintoni
- Clinical Nutrition Unit, Department of Medical and Abdominal Surgery and Endocrine-Metabolic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy (M.C.); (M.C.M.)
- Research and Training Center in Human Nutrition, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Carmelo Pozzo
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.P.); (A.S.)
| | - Antonia Strippoli
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.P.); (A.S.)
| | - Antonio Gasbarrini
- Research and Training Center in Human Nutrition, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Digestive Disease Center (CEMAD), Department of Medical and Abdominal Surgery and Endocrine-Metabolic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Maria Cristina Mele
- Clinical Nutrition Unit, Department of Medical and Abdominal Surgery and Endocrine-Metabolic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy (M.C.); (M.C.M.)
- Research and Training Center in Human Nutrition, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Emanuele Rinninella
- Clinical Nutrition Unit, Department of Medical and Abdominal Surgery and Endocrine-Metabolic Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy (M.C.); (M.C.M.)
- Research and Training Center in Human Nutrition, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
8
|
Dicks LMT. Gut Bacteria Provide Genetic and Molecular Reporter Systems to Identify Specific Diseases. Int J Mol Sci 2024; 25:4431. [PMID: 38674014 PMCID: PMC11050607 DOI: 10.3390/ijms25084431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
With genetic information gained from next-generation sequencing (NGS) and genome-wide association studies (GWAS), it is now possible to select for genes that encode reporter molecules that may be used to detect abnormalities such as alcohol-related liver disease (ARLD), cancer, cognitive impairment, multiple sclerosis (MS), diabesity, and ischemic stroke (IS). This, however, requires a thorough understanding of the gut-brain axis (GBA), the effect diets have on the selection of gut microbiota, conditions that influence the expression of microbial genes, and human physiology. Bacterial metabolites such as short-chain fatty acids (SCFAs) play a major role in gut homeostasis, maintain intestinal epithelial cells (IECs), and regulate the immune system, neurological, and endocrine functions. Changes in butyrate levels may serve as an early warning of colon cancer. Other cancer-reporting molecules are colibactin, a genotoxin produced by polyketide synthetase-positive Escherichia coli strains, and spermine oxidase (SMO). Increased butyrate levels are also associated with inflammation and impaired cognition. Dysbiosis may lead to increased production of oxidized low-density lipoproteins (OX-LDLs), known to restrict blood vessels and cause hypertension. Sudden changes in SCFA levels may also serve as a warning of IS. Early signs of ARLD may be detected by an increase in regenerating islet-derived 3 gamma (REG3G), which is associated with changes in the secretion of mucin-2 (Muc2). Pro-inflammatory molecules such as cytokines, interferons, and TNF may serve as early reporters of MS. Other examples of microbial enzymes and metabolites that may be used as reporters in the early detection of life-threatening diseases are reviewed.
Collapse
Affiliation(s)
- Leon M T Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
9
|
Lei C, Xu Y, Zhang S, Huang C, Qin J. The role of microbiota in gastric cancer: A comprehensive review. Helicobacter 2024; 29:e13071. [PMID: 38643366 DOI: 10.1111/hel.13071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/19/2024] [Accepted: 03/25/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Gastric cancer (GC) continues to pose a significant global threat in terms of cancer-related fatalities. Despite notable advancements in medical research and therapies, further investigation is warranted to elucidate its underlying etiology and risk factors. Recent times have witnessed an escalated emphasis on comprehending the role of the microbiota in cancer development. METHODS This review briefly delves into recent developments in microbiome-related research pertaining to gastric cancer. RESULTS According to studies, the microbiota can influence GC growth by inciting inflammation, disrupting immunological processes, and generating harmful microbial metabolites. Furthermore, there is ongoing research into how the microbiome can impact a patient's response to chemotherapy and immunotherapy. CONCLUSION The utilization of the microbiome for detecting, preventing, and managing stomach cancer remains an active area of exploration.
Collapse
Affiliation(s)
- Changzhen Lei
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yitian Xu
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shaopeng Zhang
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Qin
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
10
|
Han HS, Lee KW. Liquid Biopsy: An Emerging Diagnostic, Prognostic, and Predictive Tool in Gastric Cancer. J Gastric Cancer 2024; 24:4-28. [PMID: 38225764 PMCID: PMC10774753 DOI: 10.5230/jgc.2024.24.e5] [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: 11/28/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024] Open
Abstract
Liquid biopsy, a minimally invasive procedure that causes minimal pain and complication risks to patients, has been extensively studied for cancer diagnosis and treatment. Moreover, it facilitates comprehensive quantification and serial assessment of the whole-body tumor burden. Several biosources obtained through liquid biopsy have been studied as important biomarkers for establishing early diagnosis, monitoring minimal residual disease, and predicting the prognosis and response to treatment in patients with cancer. Although the clinical application of liquid biopsy in gastric cancer is not as robust as that in other cancers, biomarker studies using liquid biopsy are being actively conducted in patients with gastric cancer. Herein, we aimed to review the role of various biosources that can be obtained from patients with gastric cancer through liquid biopsies, such as blood, saliva, gastric juice, urine, stool, peritoneal lavage fluid, and ascites, by dividing them into cellular and acellular components. In addition, we reviewed previous studies on the diagnostic, prognostic, and predictive biomarkers for gastric cancer using liquid biopsy and discussed the limitations of liquid biopsy and the challenges to overcome these limitations in patients with gastric cancer.
Collapse
Affiliation(s)
- Hye Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea.
| |
Collapse
|
11
|
Lee SY, Jhun J, Woo JS, Lee KH, Hwang SH, Moon J, Park G, Choi SS, Kim SJ, Jung YJ, Song KY, Cho ML. Gut microbiome-derived butyrate inhibits the immunosuppressive factors PD-L1 and IL-10 in tumor-associated macrophages in gastric cancer. Gut Microbes 2024; 16:2300846. [PMID: 38197259 PMCID: PMC10793689 DOI: 10.1080/19490976.2023.2300846] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024] Open
Abstract
Early detection and surgical treatment are essential to achieve a good outcome in gastric cancer (GC). Stage IV and recurrent GC have a poor prognosis. Therefore, new treatments for GC are needed. We investigated the intestinal microbiome of GC patients and attempted to reverse the immunosuppression of the immune and cancer cells of GC patients through the modulation of microbiome metabolites. We evaluated the levels of programmed death-ligand 1 (PD-L1) and interleukin (IL)-10 in the peripheral blood immunocytes of GC patients. Cancer tissues were obtained from patients who underwent surgical resection of GC, and stained sections of cancer tissues were visualized via confocal microscopy. The intestinal microbiome was analyzed using stool samples of healthy individuals and GC patients. Patient-derived avatar model was developed by injecting peripheral blood mononuclear cells (PBMCs) from advanced GC (AGC) patients into NSG mice, followed by injection of AGS cells. PD-L1 and IL-10 had higher expression levels in immune cells of GC patients than in those of healthy controls. The levels of immunosuppressive factors were increased in the immune and tumor cells of tumor tissues of GC patients. The abundances of Faecalibacterium and Bifidobacterium in the intestinal flora were lower in GC patients than in healthy individuals. Butyrate, a representative microbiome metabolite, suppressed the expression levels of PD-L1 and IL-10 in immune cells. In addition, the PBMCs of AGC patients showed increased levels of immunosuppressive factors in the avatar mouse model. Butyrate inhibited tumor growth in mice. Restoration of the intestinal microbiome and its metabolic functions inhibit tumor growth and reverse the immunosuppression due to increased PD-L1 and IL-10 levels in PBMCs and tumor cells of GC patients.
Collapse
Affiliation(s)
- Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - JooYeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sun-Hee Hwang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeonghyeon Moon
- Departments of Immunobiology and Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Goeun Park
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Korea
| | - Sun Shim Choi
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Korea
| | - So Jung Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoon Ju Jung
- Division of Gastrointestinal Surgery, Department of Surgery, Yeouido St. Mary’s Hospital, Seoul, Korea
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
12
|
Ciernikova S, Sevcikova A, Mladosievicova B, Mego M. Microbiome in Cancer Development and Treatment. Microorganisms 2023; 12:24. [PMID: 38257851 PMCID: PMC10819529 DOI: 10.3390/microorganisms12010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Targeting the microbiome, microbiota-derived metabolites, and related pathways represents a significant challenge in oncology. Microbiome analyses have confirmed the negative impact of cancer treatment on gut homeostasis, resulting in acute dysbiosis and severe complications, including massive inflammatory immune response, mucosal barrier disruption, and bacterial translocation across the gut epithelium. Moreover, recent studies revealed the relationship between an imbalance in the gut microbiome and treatment-related toxicity. In this review, we provide current insights into the role of the microbiome in tumor development and the impact of gut and tumor microbiomes on chemo- and immunotherapy efficacy, as well as treatment-induced late effects, including cognitive impairment and cardiotoxicity. As discussed, microbiota modulation via probiotic supplementation and fecal microbiota transplantation represents a new trend in cancer patient care, aiming to increase bacterial diversity, alleviate acute and long-term treatment-induced toxicity, and improve the response to various treatment modalities. However, a more detailed understanding of the complex relationship between the microbiome and host can significantly contribute to integrating a microbiome-based approach into clinical practice. Determination of causal correlations might lead to the identification of clinically relevant diagnostic and prognostic microbial biomarkers. Notably, restoration of intestinal homeostasis could contribute to optimizing treatment efficacy and improving cancer patient outcomes.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia;
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia;
| | - Beata Mladosievicova
- Institute of Pathological Physiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, 833 10 Bratislava, Slovakia;
| |
Collapse
|
13
|
Zhang YN, Cui ML, Zhang LM, Lu N, Quan X, Yin K, Li AN, Zhang MX. Gut microbiota in gastric cancer: A determinant of etiology or a therapeutic approach? Shijie Huaren Xiaohua Zazhi 2023; 31:933-939. [DOI: 10.11569/wcjd.v31.i22.933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/28/2023] Open
Abstract
The impact of the gut microbiota on the well-being and pathology of the host has garnered growing interest. In recent times, there has been a surge in understanding the mechanistic connections between the gut microbiota and cancer, particularly in relation to the genesis, progression, and therapeutic approaches for gastric cancer. The dysbiosis of the intestinal microbiome stands as a significant determinant in the etiology of gastric cancer. Currently, a preliminary consensus exists, although the precise mechanism remains incompletely understood. As research progresses, it becomes increasingly evident that intestinal flora significantly contributes to the therapeutic approach for gastric cancer. This paper gives a comprehensive review of the impact of intestinal flora on gastric cancer, examines the role of the intestinal microbiome in the management of gastric cancer, and elucidates the potential of utilizing the intestinal microbiome as an anti-tumor therapy, with an aim to furnish a point of reference and stimulate future research endeavors.
Collapse
Affiliation(s)
- Ya-Nan Zhang
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Man-Li Cui
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Ling-Min Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Ning Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| | - Xin Quan
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
| | - Kun Yin
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
| | - An-Na Li
- Xi'an Medical University, Xi'an 710000, Shaanxi Province, China
| | - Ming-Xin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, Shaanxi Province, China
| |
Collapse
|
14
|
Wang M, Yang G, Tian Y, Zhang Q, Liu Z, Xin Y. The role of the gut microbiota in gastric cancer: the immunoregulation and immunotherapy. Front Immunol 2023; 14:1183331. [PMID: 37457738 PMCID: PMC10348752 DOI: 10.3389/fimmu.2023.1183331] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/11/2023] [Indexed: 07/18/2023] Open
Abstract
Gastric cancer (GC) is one of the most common cancers, leading to the deaths of millions of people worldwide. Therefore, early detection and effective therapeutic strategies are of great value for decreasing the occurrence of advanced GC. The human microbiota is involved not only in the maintenance of physiological conditions, but also in human diseases such as obesity, diabetes, allergic and atopic diseases, and cancer. Currently, the composition of the bacteria in the host, their functions, and their influence on disease progression and treatment are being discussed. Previous studies on the gut microbiome have mostly focused on Helicobacter pylori (Hp) owing to its significant role in the development of GC. Nevertheless, the enrichment and diversity of other bacteria that can modulate the tumor microenvironment are involved in the progression of GC and the efficacy of immunotherapy. This review provides systematic insight into the components of the gut microbiota and their application in GC, including the specific bacteria of GC, their immunoregulatory effect, and their diagnostic value. Furthermore, we discuss the relationship between the metabolism of microbes and their potential applications, which may serve as a new approach for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Meiqi Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ge Yang
- College of Basic Medical Sciences and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yuan Tian
- College of Basic Medical Sciences and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Qihe Zhang
- College of Basic Medical Sciences and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Zhuo Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Xin
- College of Basic Medical Sciences and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| |
Collapse
|
15
|
Vadhwana B, Tarazi M, Boshier PR, Hanna GB. Evaluation of the Oesophagogastric Cancer-Associated Microbiome: A Systematic Review and Quality Assessment. Cancers (Basel) 2023; 15:2668. [PMID: 37345006 PMCID: PMC10216300 DOI: 10.3390/cancers15102668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 06/23/2023] Open
Abstract
OBJECTIVE Oesophagogastric cancer is the fifth most common cancer worldwide, with poor survival outcomes. The role of bacteria in the pathogenesis of oesophagogastric cancer remains poorly understood. DESIGN A systematic search identified studies assessing the oesophagogastric cancer microbiome. The primary outcome was to identify bacterial enrichment specific to oesophagogastric cancer. Secondary outcomes included appraisal of the methodology, diagnostic performance of cancer bacteria and the relationship between oral and tissue microbiome. RESULTS A total of 9295 articles were identified, and 87 studies were selected for analysis. Five genera were enriched in gastric cancer: Lactobacillus, Streptococcus, Prevotella, Fusobacterium and Veillonella. No clear trends were observed in oesophageal adenocarcinoma. Streptococcus, Prevotella and Fusobacterium were abundant in oesophageal squamous cell carcinoma. Functional analysis supports the role of immune cells, localised inflammation and cancer-specific pathways mediating carcinogenesis. STORMS reporting assessment identified experimental deficiencies, considering batch effects and sources of contamination prevalent in low-biomass samples. CONCLUSIONS Functional analysis of cancer pathways can infer tumorigenesis within the cancer-microbe-immune axis. There is evidence that study design, experimental protocols and analytical techniques could be improved to achieve more accurate and representative results. Whole-genome sequencing is recommended to identify key metabolic and functional capabilities of candidate bacteria biomarkers.
Collapse
Affiliation(s)
- Bhamini Vadhwana
- Department of Surgery and Cancer, Imperial College London, 7th floor Commonwealth building, Hammersmith Hospital, London W12 0HS, UK
| | - Munir Tarazi
- Department of Surgery and Cancer, Imperial College London, 7th floor Commonwealth building, Hammersmith Hospital, London W12 0HS, UK
| | - Piers R Boshier
- Department of Surgery and Cancer, Imperial College London, 7th floor Commonwealth building, Hammersmith Hospital, London W12 0HS, UK
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, 7th floor Commonwealth building, Hammersmith Hospital, London W12 0HS, UK
| |
Collapse
|
16
|
Xi J, Li Y, Zhang H, Bai Z. Dynamic variations of the gastric microbiota: Key therapeutic points in the reversal of Correa's cascade. Int J Cancer 2023; 152:1069-1084. [PMID: 36029278 DOI: 10.1002/ijc.34264] [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: 03/31/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023]
Abstract
Correa's cascade is a dynamic process in the development of intestinal-type gastric cancer (GC), and its pathological features, gastric microbiota and interactions between microorganisms and their hosts vary at different developmental stages. The characteristics of cells, tissues and gastric microbiota before or after key therapeutic points are critical for monitoring malignant transformation and early tumour reversal. This review summarises the pathological features of gastric mucosa, characteristics of gastric microbiota, specific microbial markers, microbe-microbe interactions and microbe-host interactions at different stages in Correa's cascade. The markers related to each Correa's cascade point were analysed in detail. We attempted to identify key therapeutic points for early cancer reversal and provide a novel approach to reduce the incidence of GC and improve precise treatment.
Collapse
Affiliation(s)
- Jiahui Xi
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, Lanzhou, China
| | - Yonghong Li
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumour, Gansu Provincial Hospital, Lanzhou, China
| | - Hui Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhongtian Bai
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Key Laboratory of Biotherapy and Regenerative Medicine, Gansu Province, Lanzhou, China.,General Surgery Department, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
17
|
Guo J, Wang P, Cui Y, Hu X, Chen F, Ma C. Protective Effects of Hydroxyphenyl Propionic Acids on Lipid Metabolism and Gut Microbiota in Mice Fed a High-Fat Diet. Nutrients 2023; 15:nu15041043. [PMID: 36839401 PMCID: PMC9959022 DOI: 10.3390/nu15041043] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Gut microbiota imbalances lead to the pathogenesis of non-alcoholic fatty liver disease (NAFLD), which is primarily accompanied by hepatic steatosis. Hydroxyphenyl propionic acids (HPP) have shown great potential in inhibiting lipid accumulation but their protective effects concerning NAFLD and intestinal microbiota have remained unclear. In this paper, we investigated the efficacies of 3-HPP and 4-HPP on hepatic steatosis and gut flora in mice fed a high-fat diet (HFD). We found that 3-HPP and 4-HPP administration decreased body weight and liver index, ameliorated dyslipidemia, and alleviated hepatic steatosis. Furthermore, 3-HPP and 4-HPP enhanced the multiformity of gut microbiota; improved the relative abundance of GCA-900066575, unidentified_Lachnospiraceae, and Lachnospiraceae_UCG-006 at genus level; increased concentration of acetic acid, propionic acid and butanoic acid in faeces; and reduced systemic endotoxin levels in NAFLD mice. Moreover, 4-HPP upregulated the relative abundance of genera Rikenella and downregulated the relative abundance of Faecalibaculum. Furthermore, 3-HPP and 4-HPP regulated lipid metabolism and ameliorated gut dysbiosis in NAFLD mice and 4-HPP was more effective than 3-HPP.
Collapse
Affiliation(s)
- Jingling Guo
- Key Laboratory of Fruits and Vegetable Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, National Engineering Research Center for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Pan Wang
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Yifan Cui
- Key Laboratory of Fruits and Vegetable Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, National Engineering Research Center for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xiaosong Hu
- Key Laboratory of Fruits and Vegetable Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, National Engineering Research Center for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Fang Chen
- Key Laboratory of Fruits and Vegetable Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, National Engineering Research Center for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chen Ma
- Key Laboratory of Fruits and Vegetable Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, National Engineering Research Center for Fruit and Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Correspondence: ; Tel.: +86-158-4777-3782
| |
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
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
|
Gut Microbiota and Enteral Nutrition Tolerance in Non-Abdominal Infection Septic ICU Patients: An Observational Study. Nutrients 2022; 14:nu14245342. [PMID: 36558501 PMCID: PMC9783285 DOI: 10.3390/nu14245342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background: The effect of gut microbiota on enteral nutrition tolerance in critically ill patients is unclear. Methods: Non-abdominal sepsis patients in an ICU, sorted by whether they reached 20 Kcal/kg/day on the 3rd day of EN, were divided into tolerance and intolerance groups. Their feces on day 1 and day 3 of EN initiation were collected for 16s rDNA and short-chain fatty acid (SCFA) testing. Results: There were 14 patients included in the tolerance group and 10 in the intolerance group. On EN day 1, the OTUs and microbiota diversity were higher in the tolerance group than in the intolerance group. The ratio of Firmicutes to Bacteroidetes was higher in the intolerance group on EN day 1. The genus Parabacteroides were the most significantly elevated in the tolerance group. On EN day 3, the genus Escherichia-Shigella was the most significantly elevated in the tolerance group. On EN day 3, the levels of SCFA decreased more significantly in the intolerance group. Conclusion: Enteral nutrition tolerance is associated with microbiota features and short-chain fatty acid levels. A higher ratio of Firmicutes to Bacteroidetes and microbiota diversity on EN day 1 may help in the early prediction of EN tolerance.
Collapse
|
20
|
Lopes C, Chaves J, Ortigão R, Dinis‐Ribeiro M, Pereira C. Gastric cancer detection by non-blood-based liquid biopsies: A systematic review looking into the last decade of research. United European Gastroenterol J 2022; 11:114-130. [PMID: 36461757 PMCID: PMC9892482 DOI: 10.1002/ueg2.12328] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/21/2022] [Indexed: 12/04/2022] Open
Abstract
Gastric cancer (GC) screening is arguable in most Western countries. Liquid biopsies are a great promise to answer the unmet need for less invasive diagnostic biomarkers in GC. Thus, we aimed at systematically reviewing the current knowledge on liquid biopsy-based biomarkers in GC screening. A systematic search on PubMed/MEDLINE and Scopus databases was performed on published articles reporting the use of non-blood specimen (saliva, gastric juice [GJ], urine and stool) on GC diagnosis. 3208 records were retrieved by June 2022. After removal of duplicate records, 2379 abstracts were screened, and 84 full texts included in this systematic review. More than 90% of studies were reported on Asian populations. Overall, 9 studies explored stool-, 12 saliva-, and 29 urine-derived biomarkers for GC detection. Additionally, 37 studies, representing the majority, analyzed GJ, focusing on nucleic acid molecules. Several miRNAs and lncRNA molecules have been associated with GC risk, particularly miR-21 (area under the curve [AUC] = 0.97, 95% CI: 0.94-1.00). Considering salivary biomarkers, the best described model in validation sets included the soybean agglutinin and Vicia villosa agglutinin lectins (AUC = 0.89, 95% CI: 0.80-0.99). Most studies in urine carried out metabolomic approaches, with two discriminatory models presenting AUC values superior to 0.97. This systematic review emphasizes the potential role of non-blood-based biomarkers, although further validation, particularly in Western countries, is mandatory, namely for non-invasive screening and/or monitoring, as well as the use of GJ as a tool to enhance upper gastrointestinal endoscopy accuracy.
Collapse
Affiliation(s)
- Catarina Lopes
- Precancerous Lesions and Early Cancer Management GroupResearch Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group)Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC)PortoPortugal,CINTESIS – Center for Health Technology and Services ResearchUniversity of PortoPortoPortugal,ICBAS‐UP – Institute of Biomedical Sciences Abel SalazarUniversity of PortoPortoPortugal
| | - Jéssica Chaves
- Precancerous Lesions and Early Cancer Management GroupResearch Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group)Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC)PortoPortugal,Department of GastroenterologyPortuguese Oncology Institute of PortoPortoPortugal
| | - Raquel Ortigão
- Precancerous Lesions and Early Cancer Management GroupResearch Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group)Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC)PortoPortugal,Department of GastroenterologyPortuguese Oncology Institute of PortoPortoPortugal
| | - Mário Dinis‐Ribeiro
- Precancerous Lesions and Early Cancer Management GroupResearch Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group)Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC)PortoPortugal,Department of GastroenterologyPortuguese Oncology Institute of PortoPortoPortugal
| | - Carina Pereira
- Precancerous Lesions and Early Cancer Management GroupResearch Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group)Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC)PortoPortugal,CINTESIS – Center for Health Technology and Services ResearchUniversity of PortoPortoPortugal
| |
Collapse
|
21
|
Meta-analysis of the gut microbiota alterations in patients with gastric cancer in China. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
22
|
Mathebela P, Damane BP, Mulaudzi TV, Mkhize-Khwitshana ZL, Gaudji GR, Dlamini Z. Influence of the Microbiome Metagenomics and Epigenomics on Gastric Cancer. Int J Mol Sci 2022; 23:13750. [PMID: 36430229 PMCID: PMC9693604 DOI: 10.3390/ijms232213750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Gastric cancer (GC) is one of the major causes of cancer deaths worldwide. The disease is seldomly detected early and this limits treatment options. Because of its heterogeneous and complex nature, the disease remains poorly understood. The literature supports the contribution of the gut microbiome in the carcinogenesis and chemoresistance of GC. Drug resistance is the major challenge in GC therapy, occurring as a result of rewired metabolism. Metabolic rewiring stems from recurring genetic and epigenetic factors affecting cell development. The gut microbiome consists of pathogens such as H. pylori, which can foster both epigenetic alterations and mutagenesis on the host genome. Most of the bacteria implicated in GC development are Gram-negative, which makes it challenging to eradicate the disease. Gram-negative bacterium co-infections with viruses such as EBV are known as risk factors for GC. In this review, we discuss the role of microbiome-induced GC carcinogenesis. The disease risk factors associated with the presence of microorganisms and microbial dysbiosis are also discussed. In doing so, we aim to emphasize the critical role of the microbiome on cancer pathological phenotypes, and how microbiomics could serve as a potential breakthrough in determining effective GC therapeutic targets. Additionally, consideration of microbial dysbiosis in the GC classification system might aid in diagnosis and treatment decision-making, taking the specific pathogen/s involved into account.
Collapse
Affiliation(s)
- Precious Mathebela
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| | - Botle Precious Damane
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| | - Zilungile Lynette Mkhize-Khwitshana
- School of Medicine, University of Kwa-Zulu Natal, Durban, KwaZulu-Natal 4013, South Africa
- SAMRC Research Capacity Development Division, South African Medical Research Council, Tygerberg, Cape Town 7501, South Africa
| | - Guy Roger Gaudji
- Department of Urology, Level 7, Bridge C, Steve Biko Academic Hospital, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
| |
Collapse
|
23
|
Kashyap S, Pal S, Chandan G, Saini V, Chakrabarti S, Saini NK, Mittal A, Thakur VK, Saini AK, Saini RV. Understanding the cross-talk between human microbiota and gastrointestinal cancer for developing potential diagnostic and prognostic biomarkers. Semin Cancer Biol 2022; 86:643-651. [PMID: 33971261 DOI: 10.1016/j.semcancer.2021.04.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/19/2021] [Accepted: 04/29/2021] [Indexed: 02/07/2023]
Abstract
The interaction between gut microbes and gastrointestinal (GI) tract carcinogenesis has always attracted researchers' attention to identify therapeutic targets or potential prognostic biomarkers. Various studies have suggested that the microbiota do show inflammation and immune dysregulation, which led to carcinogenesis in GI tract. In this review, we have focused on the role of microbes present in the gut, intestine, or faeces in GI tract cancers, including esophageal cancer, gastric cancer, and colorectal cancer. Herein, we have discussed the importance of the microbes and their metabolites, which could serve as diagnostic biomarkers for cancer detection, especially in the early stage, and prognostic markers. To maximize the effect of the treatment strategies, an accurate evaluation of the prognosis is imperative for clinicians. There is a vast difference in the microbiota profiles within a population and across the populations depending upon age, diet, lifestyle, genetic makeup, use of antibiotics, and environmental factors. Therefore, the diagnostic efficiency of the microbial markers needs to be further validated. A deeper understanding of the GI cancer and the host microbiota is needed to acquire pivotal information about disease status.
Collapse
Affiliation(s)
- Sheetal Kashyap
- Department of Biotechnology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India
| | - Soumya Pal
- Department of Biotechnology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India
| | - Gourav Chandan
- Central Research Cell, MM Institute of Medical Sciences & Research, Maharishi Markandeshwar (Deemed to be University), Mullana, 133207, Haryana, India
| | - Vipin Saini
- Maharishi Markandeshwar University, Solan, 173229, Himachal Pradesh, India
| | - Sasanka Chakrabarti
- Central Research Cell, MM Institute of Medical Sciences & Research, Maharishi Markandeshwar (Deemed to be University), Mullana, 133207, Haryana, India
| | - Neeraj K Saini
- Department of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amit Mittal
- Central Research Cell, MM Institute of Medical Sciences & Research, Maharishi Markandeshwar (Deemed to be University), Mullana, 133207, Haryana, India
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, Edinburgh, EH9 3JG, UK
| | - Adesh K Saini
- Department of Biotechnology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India.
| | - Reena V Saini
- Department of Biotechnology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133207, Haryana, India.
| |
Collapse
|
24
|
Zhao Y, Li Z, Zhao L, Wang J, Wang F, Zhang Q, Wang X, Sang Y, Zhan J, He J, Li N, Kang X, Chen J, Wang R. Two novel lactic acid bacteria, Limosilactobacillus fermentum MN-LF23 and Lactobacillus gasseri MN-LG80, inhibited Helicobacter pylori infection in C57BL/6 mice. Food Funct 2022; 13:11061-11069. [PMID: 36197065 DOI: 10.1039/d2fo02034c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Helicobacter pylori (H. pylori) is one of the most prevalent pathogens globally, and long-term infection causes various gastrointestinal diseases such as gastritis and even cancer. In the present study, we screened dozens of lactic acid bacteria for the efficacy to inhibit H. pylori growth in vitro, and tested the therapeutic effects of candidate strains in vivo. The results showed that Limosilactobacillus fermentum MN-LF23 (LF23) and Lactobacillus gasseri MN-LG80 (LG80) significantly reduced the abundance of Helicobacter by 90% and 83% in the infected mice, respectively, and decreased the levels of serum urease and H. pylori-specific IgG. Both bacterial strains tended to ameliorate H. pylori infection-induced gastric mucosa damage and lymphocyte infiltration, and reduced levels of serum inflammatory cytokines such as TNF-α, IL-1β, and IL-6. In addition, their culture supernatants also showed a therapeutic effect, as efficient as the bacterial cells. Furthermore, both strains significantly regulated gastric microbiota profile, and their supernatants restored the diversity of gastric microbiota. LF23 increased the abundance of Lactobacillus murinus and reduced the abundance of Desulfovibrio, whereas LG80 increased the abundance of Lactobacillus reuteri and reduced the abundance of Bilophila. Both LF23 and LG80 enriched beneficial commensals such as Faecalibaculum rodentium, and reduced detrimental bacteria such as H. pylori and Lachnoclostridium. In conclusion, we identified two novel lactic acid bacteria L. fermentum MN-LF23 and L. gasseri MN-LG80 that can remarkably inhibit H. pylori infection.
Collapse
Affiliation(s)
- Yuyang Zhao
- College of Food Science and Nutritional Engineering, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100083, China
| | - Zhipeng Li
- R&D Center, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Huhhot, 011500, Inner Mongolia, China.
- Shanghai Mengniu Biotechnology R&D Co., Ltd., 201210, Shanghai, China
| | - Liang Zhao
- College of Food Science and Nutritional Engineering, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100083, China
| | - Jian Wang
- College of Food Science and Nutritional Engineering, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100083, China
| | - Fan Wang
- R&D Center, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Huhhot, 011500, Inner Mongolia, China.
- Shanghai Mengniu Biotechnology R&D Co., Ltd., 201210, Shanghai, China
| | - Qi Zhang
- College of Food Science and Nutritional Engineering, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100083, China
| | - Xiaoyu Wang
- College of Food Science and Nutritional Engineering, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing 100083, China
| | - Yue Sang
- Hebei Engineering Research Center of Animal Product, Sanhe 065200, China
| | - Jing Zhan
- Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Jingjing He
- Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Ning Li
- R&D Center, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Huhhot, 011500, Inner Mongolia, China.
| | - Xiaohong Kang
- R&D Center, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Huhhot, 011500, Inner Mongolia, China.
- Shanghai Mengniu Biotechnology R&D Co., Ltd., 201210, Shanghai, China
| | - Jianguo Chen
- R&D Center, Inner Mongolia Mengniu Dairy (Group) Co. Ltd., Huhhot, 011500, Inner Mongolia, China.
- Shanghai Mengniu Biotechnology R&D Co., Ltd., 201210, Shanghai, China
| | - Ran Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| |
Collapse
|
25
|
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
|
26
|
Zhang C, Hu A, Li J, Zhang F, Zhong P, Li Y, Li Y. Combined Non-Invasive Prediction and New Biomarkers of Oral and Fecal Microbiota in Patients With Gastric and Colorectal Cancer. Front Cell Infect Microbiol 2022; 12:830684. [PMID: 35663463 PMCID: PMC9161364 DOI: 10.3389/fcimb.2022.830684] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background There is no information on the commonality and specificity of oral and fecal microbiota in patients with gastric cancer (GC) and colorectal cancer (CRC). Methods The high-throughput 16S rRNA gene V4 region sequencing was used to perform bioinformatics analysis of oral, fecal, and tissue microbiota in GC (76 subjects), CRC (53), and healthy controls (HC, 70). Furthermore, we determined the microbial characteristics of each part, constructed and verified three classifiers for GC and CRC, and evaluated curves of receiver operating characteristic and precision–recall with probability of disease. Results Compared to HC, the microbial richness and diversity of GC and CRC decreased in oral cavity and increased in stool; additionally, these indexes in GC tissue were higher than those in CRC tissue. In GC and CRC patients, Haemophilus, Neisseria, Faecalibacterium, and Romboutsia were significantly reduced compared to the relative abundance value of oral or fecal bacterial genera in the HC group, while the Streptococcus, Gemella, Escherichia-Shigella, and Fusobacterium were significantly increased. The oral and tissue microbiota have similar and abundant shared bacterial networks. The single and combined microbial detection have good AUC values based on POD indices for predicting GC, CRC, and gastrointestinal (GI) cancers (GC and CRC). Conclusion This study is the first to examine the characteristics of oral, fecal, and tumor microbiota in GC and CRC patients, and the similarities and differences in their microbial changes are reported. These oral or fecal bacteria (Haemophilus, Neisseria, Faecalibacterium, Romboutsia, Streptococcus, Gemella, Escherichia-Shigella, and Fusobacterium) may be involved in tumor evolution as potentially characteristic genera. In addition, both oral and fecal microbial detection may provide a solid theoretical foundation for the non-invasive prediction of these cancers.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Asheng Hu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jingxing Li
- Department of Computer Science, Faculty of Science, University of Western Ontario, London, ON, Canada
| | - Fangfang Zhang
- Department of Anesthesiology, Hefei BOE Hospital, Hefei, China
| | - Pei Zhong
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yaxian Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Determining the protective effects of Ma-Mu-Ran Antidiarrheal Capsules against acute DSS-induced enteritis using 16S rRNA gene sequencing and fecal metabolomics. Chin J Nat Med 2022; 20:364-377. [DOI: 10.1016/s1875-5364(22)60158-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Indexed: 11/20/2022]
|
28
|
Roles of Microbiota in Cancer: From Tumor Development to Treatment. JOURNAL OF ONCOLOGY 2022; 2022:3845104. [PMID: 35342407 PMCID: PMC8941494 DOI: 10.1155/2022/3845104] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 12/11/2022]
Abstract
Cancer as a second leading cause of death arises from multifactorial pathology. The association of microbiota and their products with various pathologic conditions including cancer is receiving significant attention over the past few years. Mounting evidence showed that human microbiota is an emerging target in tumor onset, progression, prevention, and even diagnosis. Accordingly, modulating this composition might influence the response to tumor therapy and therapeutic resistance as well. Through this review, one could conceive of complex interaction between the microbiome and cancer in either positive or negative manner by which may hold potential for finding novel preventive and therapeutic strategies against cancer.
Collapse
|
29
|
Herrera-Pariente C, Montori S, Llach J, Bofill A, Albeniz E, Moreira L. Biomarkers for Gastric Cancer Screening and Early Diagnosis. Biomedicines 2021; 9:biomedicines9101448. [PMID: 34680565 PMCID: PMC8533304 DOI: 10.3390/biomedicines9101448] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the most common cancers worldwide, with a bad prognosis associated with late-stage diagnosis, significantly decreasing the overall survival. This highlights the importance of early detection to improve the clinical course of these patients. Although screening programs, based on endoscopic or radiologic approaches, have been useful in countries with high incidence, they are not cost-effective in low-incidence populations as a massive screening strategy. Additionally, current biomarkers used in daily routine are not specific and sensitive enough, and most of them are obtained invasively. Thus, it is imperative to discover new noninvasive biomarkers able to diagnose early-stage gastric cancer. In this context, liquid biopsy is a promising strategy. In this review, we briefly discuss some of the potential biomarkers for gastric cancer screening and diagnosis identified in blood, saliva, urine, stool, and gastric juice.
Collapse
Affiliation(s)
- Cristina Herrera-Pariente
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (J.L.); (A.B.)
| | - Sheyla Montori
- UPNA, IdiSNA, Navarrabiomed Biomedical Research Center, Gastrointestinal Endoscopy Research Unit, 31008 Pamplona, Spain; (S.M.); (E.A.)
| | - Joan Llach
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (J.L.); (A.B.)
| | - Alex Bofill
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (J.L.); (A.B.)
| | - Eduardo Albeniz
- UPNA, IdiSNA, Navarrabiomed Biomedical Research Center, Gastrointestinal Endoscopy Research Unit, 31008 Pamplona, Spain; (S.M.); (E.A.)
- Endoscopy Unit, Gastroenterology Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
| | - Leticia Moreira
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (C.H.-P.); (J.L.); (A.B.)
- Correspondence:
| |
Collapse
|
30
|
Gut Microbiota as Potential Biomarker and/or Therapeutic Target to Improve the Management of Cancer: Focus on Colibactin-Producing Escherichia coli in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13092215. [PMID: 34063108 PMCID: PMC8124679 DOI: 10.3390/cancers13092215] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Gut microbiota is emerging as new diagnostic and prognostic marker and/or therapeutic target to improve the management of cancer. This review aims to summarize microbial signatures that have been associated with digestive and other cancers. We report the clinical relevance of these microbial markers to predict the response to cancer therapy. Among these biomarkers, colibactin-producing E. coli are prevalent in the colonic mucosa of patients with colorectal cancer and they promote colorectal carcinogenesis in several pre-clinical models. Here we discuss the promising use of colibactin-producing E. coli as a new predictive factor and a therapeutic target in colon cancer management. Abstract The gut microbiota is crucial for physiological development and immunological homeostasis. Alterations of this microbial community called dysbiosis, have been associated with cancers such colorectal cancers (CRC). The pro-carcinogenic potential of this dysbiotic microbiota has been demonstrated in the colon. Recently the role of the microbiota in the efficacy of anti-tumor therapeutic strategies has been described in digestive cancers and in other cancers (e.g., melanoma and sarcoma). Different bacterial species seem to be implicated in these mechanisms: F. nucleatum, B. fragilis, and colibactin-associated E. coli (CoPEC). CoPEC bacteria are prevalent in the colonic mucosa of patients with CRC and they promote colorectal carcinogenesis in susceptible mouse models of CRC. In this review, we report preclinical and clinical data that suggest that CoPEC could be a new factor predictive of poor outcomes that could be used to improve cancer management. Moreover, we describe the possibility of using these bacteria as new therapeutic targets.
Collapse
|
31
|
Ding Q, Zhang Z, Li Y, Liu H, Hao Q, Yang Y, Ringø E, Olsen RE, Clarke JL, Ran C, Zhou Z. Propionate induces intestinal oxidative stress via Sod2 propionylation in zebrafish. iScience 2021; 24:102515. [PMID: 34142031 PMCID: PMC8188496 DOI: 10.1016/j.isci.2021.102515] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/17/2021] [Accepted: 05/03/2021] [Indexed: 12/21/2022] Open
Abstract
Propionate and propionyl-CoA accumulation have been associated with the development of mitochondrial dysfunction. In this study, we show that propionate induces intestinal damage in zebrafish when fed a high-fat diet (HFD). The intestinal damage was associated with oxidative stress owing to compromised superoxide dismutase 2 (Sod2) activity. Global lysine propionylation analysis of the intestinal samples showed that Sod2 was propionylated at lysine 132 (K132), and further biochemical assays demonstrated that K132 propionylation suppressed Sod2 activity. In addition, sirtuin 3 (Sirt3) played an important role in regulating Sod2 activity via modulating de-propionylation. Finally, we revealed that intestinal oxidative stress resulting from Sod2 propionylation contributed to compositional change of gut microbiota. Collectively, our results in this study show that there is a link between Sod2 propionylation and oxidative stress in zebrafish intestines and highlight the potential mechanism of intestinal problems associated with high propionate levels. Propionate supplementation in high-fat diet induces intestinal damage Propionate induces oxidative stress via Sod2 propionylation at 132 lysine site Increased Sod2 propionylation is associated with reduced expression of Sirt3 Intestinal oxidative stress alters gut microbiota composition
Collapse
Affiliation(s)
- Qianwen Ding
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China.,Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yu Li
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Hongliang Liu
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Qiang Hao
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Einar Ringø
- Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Rolf Erik Olsen
- Norway-China Joint Lab on Fish Gastrointestinal Microbiota, Institute of Biology, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | | | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zhigang Zhou
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|