1
|
Katona BW, Shukla A, Hu W, Nyul T, Dudzik C, Arvanitis A, Clay D, Dungan M, Weber M, Tu V, Hao F, Gan S, Chau L, Buchner AM, Falk GW, Jaffe DL, Ginsberg G, Palmer SN, Zhan X, Patterson AD, Bittinger K, Ni J. Microbiota and metabolite-based prediction tool for colonic polyposis with and without a known genetic driver. Gut Microbes 2025; 17:2474141. [PMID: 40069167 PMCID: PMC11913376 DOI: 10.1080/19490976.2025.2474141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Despite extensive investigations into the microbiome and metabolome changes associated with colon polyps and colorectal cancer (CRC), the microbiome and metabolome profiles of individuals with colonic polyposis, including those with (Gene-pos) and without (Gene-neg) a known genetic driver, remain comparatively unexplored. Using colon biopsies, polyps, and stool from patients with Gene-pos adenomatous polyposis (N = 9), Gene-neg adenomatous polyposis (N = 18), and serrated polyposis syndrome (SPS, N = 11), we demonstrated through 16S rRNA sequencing that the mucosa-associated microbiota in individuals with colonic polyposis is representative of the microbiota associated with small polyps, and that both Gene-pos and SPS cohorts exhibit differential microbiota populations relative to Gene-neg polyposis cohorts. Furthermore, we used these differential microbiota taxa to perform linear discriminant analysis to differentiate Gene-neg subjects from Gene-pos and from SPS subjects with an accuracy of 89% and 93% respectively. Stool metabolites were quantified via 1H NMR, revealing an increase in alanine in SPS subjects relative to non-polyposis subjects, and Partial Least Squares Discriminant Analysis (PLS-DA) analysis indicated that the proportion of leucine to tyrosine in fecal samples may be predictive of SPS. Use of these microbial and metabolomic signatures may allow for better diagnostric and risk-stratification tools for colonic polyposis patients and their families as well as promote development of microbiome-targeted approaches for polyp prevention.
Collapse
Affiliation(s)
- Bryson W. Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ashutosh Shukla
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiming Hu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Thomas Nyul
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina Dudzik
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Alex Arvanitis
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Clay
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michaela Dungan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marina Weber
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Vincent Tu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Shuheng Gan
- Peter O’Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lillian Chau
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anna M. Buchner
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gary W. Falk
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David L. Jaffe
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gregory Ginsberg
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Suzette N. Palmer
- Peter O’Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaowei Zhan
- Peter O’Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Josephine Ni
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Wang X, Chen H, Yang M, Huang M, Zhang D, Li M, Wang H, Zhou Q, Lu L, Li Y, Yu J, Ma L. Influence of gut microbiota and immune markers in different stages of colorectal adenomas. Front Microbiol 2025; 16:1556056. [PMID: 40309115 PMCID: PMC12040870 DOI: 10.3389/fmicb.2025.1556056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Objective Colorectal adenomas (CRA) are the primary precancerous lesions leading to colorectal cancer (CRC). Early detection and intervention of CRA can significantly reduce the incidence of CRC. We investigated the relationships between the gut microbiome and the expression levels of PD-L1, IL-6, and IFN-γ at different CRA stages. Methods Participants were divided into normal, non-advanced adenoma (NAA), and advanced adenoma (AA) groups. PD-L1 expression in collected tissues was analyzed via immunohistochemistry (IHC) and Western blotting. Serum IL-6 and IFN-γ levels were measured using Enzyme-Linked Immunosorbent Assay (ELISA). 16S rRNA gene sequencing was used to examine gut microbiota changes, with correlation analysis to assess microbial influences on CRA progression. Results The main differences in bacterial composition among the three groups were found within the Firmicutes and Bacteroidetes phyla. In the normal vs. NAA comparison, Clostridium sensu stricto, Faecalimonas, Gemmiger, and Ruminococcus were more abundant in the normal group, while Solobacterium was enriched in the NAA group. For the normal vs. AA comparison, the normal group was enriched with Anaerostipes, Blautia, Clostridium sensu stricto, Intestinibacter, Phocaeicola, and Turicibacter, whereas Solobacterium was more abundant in the AA group. In the NAA vs. AA comparison, the NAA group exhibited higher levels of Blautia, Faecalimonas, and Turicibacter relative to the AA group. Anaerostipes and Blautia are positively correlated with taurine and hypotaurine metabolism, propanoate metabolism, and zeatin biosynthesis. PD-L1 protein levels progressively increase with CRA advancement. Additionally, Faecalimonas, and Solobacterium were negatively associated with IFN-γ, while Gemmiger, and Anaerostipes were positively associated with IL-6. Conclusion This study highlights the dynamic alterations in gut microbiota composition and their potential influence on the regulation of inflammatory cytokines and PD-L1 expression during CRA progression. The enrichment of protective taxa, such as Anaerostipes and Blautia, in the normal group emphasizes their potential role in mitigating adenoma progression. Dietary modulation to promote the proliferation of these beneficial bacteria could serve as a promising strategy to improve colorectal health. Future research should further explore the specific relationships between dietary components, gut microbiota, and metabolic pathways, and assess the effects of dietary interventions on gut health.
Collapse
Affiliation(s)
- Xianmei Wang
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hang Chen
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Meng Yang
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Minshan Huang
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dan Zhang
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mingke Li
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hui Wang
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qingqing Zhou
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihong Lu
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu Li
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiangkun Yu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Lanqing Ma
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
3
|
Bakir-Gungor B, Temiz M, Canakcimaksutoglu B, Yousef M. Prediction of colorectal cancer based on taxonomic levels of microorganisms and discovery of taxonomic biomarkers using the Grouping-Scoring-Modeling (G-S-M) approach. Comput Biol Med 2025; 187:109813. [PMID: 39929003 DOI: 10.1016/j.compbiomed.2025.109813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/09/2025] [Accepted: 02/05/2025] [Indexed: 02/12/2025]
Abstract
Colorectal cancer (CRC) is one of the most prevalent forms of cancer globally. The human gut microbiome plays an important role in the development of CRC and serves as a biomarker for early detection and treatment. This research effort focuses on the identification of potential taxonomic biomarkers of CRC using a grouping-based feature selection method. Additionally, this study investigates the effect of incorporating biological domain knowledge into the feature selection process while identifying CRC-associated microorganisms. Conventional feature selection techniques often fail to leverage existing biological knowledge during metagenomic data analysis. To address this gap, we propose taxonomy-based Grouping Scoring Modeling (G-S-M) method that integrates biological domain knowledge into feature grouping and selection. In this study, using metagenomic data related to CRC, classification is performed at three taxonomic levels (genus, family and order). The MetaPhlAn tool is employed to determine the relative abundance values of species in each sample. Comparative performance analyses involve six feature selection methods and four classification algorithms. When experimented on two CRC associated metagenomics datasets, the highest performance metric, yielding an AUC of 0.90, is observed at the genus taxonomic level. At this level, 7 out of top 10 groups (Parvimonas, Peptostreptococcus, Fusobacterium, Gemella, Streptococcus, Porphyromonas and Solobacterium) were commonly identified for both datasets. Moreover, the identified microorganisms at genus, family, and order levels are thoroughly discussed via refering to CRC-related metagenomic literature. This study not only contributes to our understanding of CRC development, but also highlights the applicability of taxonomy-based G-S-M method in tackling various diseases.
Collapse
Affiliation(s)
- Burcu Bakir-Gungor
- Department of Computer Engineering, Faculty of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey
| | - Mustafa Temiz
- Department of Electrical and Computer Engineering, Faculty of Engineering, Abdullah Gul University, Kayseri, 38080, Turkey.
| | - Beyza Canakcimaksutoglu
- Department of Bioengineering, Faculty of Life and Natural Science, Abdullah Gul University, Kayseri, 38080, Turkey
| | - Malik Yousef
- Department of Information Systems, Zefat Academic College, Zefat, 13206, Israel; Galilee Digital Health Research Center (GDH), Zefat Academic College, Israel
| |
Collapse
|
4
|
Reichmann R, Nimptsch K, Pischon T, Gunter MJ, Jenab M, Eriksen AK, Tjonneland A, Janke J, Katzke V, Kaaks R, Schulze MB, Eichelmann F, Masala G, Sieri S, Pasanisi F, Tumino R, Giraudo MT, Rothwell J, Severi G, Jakszyn P, Sanchez-Perez MJ, Amiano P, Colorado-Yohar SM, Guevara M, van Guelpen B, Aglago EK, Heath AK, Smith-Byrne K, Weiderpass E, Aleksandrova K. Sex- and site-specific associations of circulating lipocalin 2 and incident colorectal cancer: Results from the EPIC cohort. Int J Cancer 2025; 156:930-942. [PMID: 39511728 DOI: 10.1002/ijc.35205] [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: 03/23/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 11/15/2024]
Abstract
Experimental research has uncovered lipocalin 2 (LCN2) as a novel biomarker implicated in the modulation of intestinal inflammation, metabolic homeostasis, and colon carcinogenesis. However, evidence from human research has been scant. We, therefore, explored the association of pre-diagnostic circulating LCN2 concentrations with incident colorectal cancer (CRC) in a nested case-control study within the in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. LCN2 was measured in 1267 incident CRC cases matched to 1267 controls using incidence density sampling. Conditional logistic regression was used to estimate incidence rate ratios (IRRs) and 95% confidence intervals (95% CIs) according to tumor subsite and sex. Weighted Cox proportional hazard regression was used to explore associations by adiposity status. In multivariable-adjusted analyses, the IRR [95% CI] per doubling in LCN2 concentration was 1.16 [0.98-1.37] for CRC overall, 1.26 [1.00-1.59] for colon cancer, and 1.08 [0.85-1.38] for rectal cancer. The association for colon cancer was more pronounced in women (IRR [95% CI], 1.66 [1.20-2.30]) and for proximal colon cancer (IRR [95% CI], 1.96 [1.15-3.34]), whereas no association was seen in men and distal colon cancer. The association for colon cancer was positive in individuals with high waist circumference (hazard ratio [95% CI], 1.69 [1.52-1.88]) and inverse in individuals with low waist circumference (hazard ratio [95% CI], 0.86 [0.76-0.98], P interaction<0.01). Overall, these data suggest that pre-diagnostic LCN2 concentrations were positively associated with colon cancer, particularly occurring in the proximal colon, in women and among individuals with abdominal adiposity.
Collapse
Grants
- Dutch Ministry of Public Health, Welfare and Sports (VWS), Netherlands Cancer Registry (NKR), LK Research Funds, Dutch Prevention Funds, Dutch ZON (Zorg Onderzoek Nederland), World Cancer Research Fund (WCRF), Statistics Netherlands (The Netherlands)
- German Cancer Aid, German Cancer Research Center (DKFZ), German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Federal Ministry of Education and Research (BMBF) (Germany)
- Danish Cancer Society (Denmark)
- Ligue Contre le Cancer, Institut Gustave Roussy, Mutuelle Générale de l'Education Nationale, Institut National de la Santé et de la Recherche Médicale (INSERM) (France)
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London
- 001 World Health Organization
- Associazione Italiana per la Ricerca sul Cancro-AIRC-Italy, Compagnia di SanPaolo and National Research Council (Italy)
- International Agency for Research on Cancer (IARC)
- Cancer Research UK (14,136 to EPIC-Norfolk; C8221/A29017 to EPIC-Oxford), Medical Research Council (1,000,143 to EPIC-Norfolk; MR/M012190/1 to EPIC-Oxford) (United Kingdom).
- Health Research Fund (FIS)-Instituto de Salud Carlos III (ISCIII), Regional Governments of Andalucía, Asturias, Basque Country, Murcia and Navarra, and the Catalan Institute of Oncology-ICO (Spain)
- Swedish Cancer Society, Swedish Research Council, Region Skåne and Region Västerbotten (Sweden)
Collapse
Affiliation(s)
- Robin Reichmann
- Biomarkers and Metabolism Research Group, Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany
| | - Katharina Nimptsch
- Molecular Epidemiology Research Group, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tobias Pischon
- Molecular Epidemiology Research Group, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Core Facility Biobank, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marc J Gunter
- International Agency for Research on Cancer, World Health Organization, Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Mazda Jenab
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Anne Kirstine Eriksen
- Diet, Cancer and Health Research Group, Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
| | - Anne Tjonneland
- Diet, Cancer and Health Research Group, Danish Cancer Institute, Danish Cancer Society, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Jürgen Janke
- Molecular Epidemiology Research Group, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Verena Katzke
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Department of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Giovanna Masala
- Prevention and Clinical Network, Institute for the Study and Prevention of Cancer (ISPRO), Florenz, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy
| | - Fabrizio Pasanisi
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Rosario Tumino
- Hyblean Association for Epidemiological Research, Associazione Iblea per la Ricerca Epidemiologica (A.I.R.E.-ONLUS), Ragusa, Italy
| | - Maria Teresa Giraudo
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Joseph Rothwell
- CESP-Univ. Paris-Saclay, UVSQ, Inserm-"Exposome, heredity, cancer and health" Team, The Centre for Research in Epidemiology and Population Health, Villejuif, France
| | - Gianluca Severi
- CESP-Univ. Paris-Saclay, UVSQ, Inserm-"Exposome, heredity, cancer and health" Team, The Centre for Research in Epidemiology and Population Health, Villejuif, France
- Department of Statistics, Computer Science, Applications "G. Parenti" (DISIA), University of Florence, Florence, Italy
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Maria Jose Sanchez-Perez
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Public Health Research and Health Services Research Group, Andalusian School of Public Health (EASP), Granada, Andalucía, Spain
- Epidemiology, Prevention and Control of Cancer Research Group, Biosanitary Research Institute of Granada (ibs.Granada), Granada, Spain
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain
| | - Pilar Amiano
- Sub Directorate for Public Health and Addictions of Gipuzkoa, Ministry of Health of the Basque Government, San Sebastian, Spain
- Epidemiology of Chronic and Communicable Diseases Group, Biodonostia Health Research Institute, San Sebastian, Spain
- Instituto de Salud Carlos III, CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Sandra M Colorado-Yohar
- Department of Epidemiology, Murcia Regional Health Council, Instituto Murciano de Investigación Biosanitaria, Murcia, Spain
- Research Group on Demography and Health, National Faculty of Public Health, University of Antioquia, Medellin, Colombia
| | - Marcela Guevara
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Epidemiology and Health Prevention Service, Institute of Public Health and Labor of Navarre, Pamplona, Navarra, Spain
- Epidemiology of Cancer and Other Chronic Diseases Research Group, Healthcare Research Institute of Navarre (IdiSNA), Pamplona, Spain
| | - Bethany van Guelpen
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Elom K Aglago
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Karl Smith-Byrne
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Krasimira Aleksandrova
- Biomarkers and Metabolism Research Group, Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology, Bremen, Germany
- Faculty of Human and Health Sciences, University of Bremen, Bremen, Germany
| |
Collapse
|
5
|
He F, Huang X, Wang Z, Qin M, Chen C, Huang Z, Wu Y, Huang Y, Tang B, Long C, Mo X, Tang W, Liu J. The Effect of Gender on the Intestinal Flora of Colorectal Cancer Under Different Stages. Mol Carcinog 2025; 64:526-542. [PMID: 39692233 DOI: 10.1002/mc.23863] [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: 05/26/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
This study aims to determine whether gender is a factor in the interplay between the human intestinal flora and colorectal cancer (CRC), ultimately providing new evidence for the clinical prediction and management of CRC in different genders. In this study, we included 186 untreated CRC patients, and classified them into two groups based on pathological staging: Groups Ⅰ-Ⅱ and Groups Ⅲ-Ⅳ, with male and female groups within each group. We collected preoperative fecal samples from these patients and performed 16S rRNA gene sequencing to analyze their intestinal flora. In the CRC Stages I-II cohort, the gut microbiota of the female group exhibited greater diversity and abundance compared to the male group, with a total of 13 gut microbiota demonstrating significant disparities. Notably, s__Parabacteroides gordonii, s__Bacteroides faecis, and s__Bacteroides nordii were found to be more prevalent in the female group relative to the male group. Within the CRC Stages III-IV cohort, 51 gut microbiota exhibited significant differences between the genders. In the immunocyte composition of fecal samples from patients with CRC, a higher proportion of naive B cells is observed in the male group as compared to the female group. In female CRC patients within the CRC Stages III-IV cohort, Actinomyces exhibited a significant negative correlation with activated dendritic cells, CD4+ memory T cells, and eosinophils. In male CRC patients within the CRC Stages III-IV cohort, Actinomyces demonstrated a significant positive correlation with naive B cells and a significant positive correlation with immune activation genes TNFRSF25 and TMIGD2. In female CRC patients within the CRC Stages III-IV cohort, Actinomyces showed a significant negative correlation with activated dendritic cells, CD4+ memory T cells, and eosinophils, and a significant positive correlation with immune activation genes TNFSF13B, LTA, KLRK1, and CXCL12. In the CRC Stages I-II group, the female group's intestinal flora is more diverse and richer than the male group. In the CRC Stages III-IV group, there are a total of 51 different intestinal flora in both the male and female groups. We also found that Actinomyces affects the occurrence and development of CRC in the male and female groups through different pathways. The results show that the intestinal flora differs between male and female CRC patients and is closely associated with cancer development.
Collapse
Affiliation(s)
- Fuhai He
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Xiaoliang Huang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Zhen Wang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Mingjian Qin
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Chuanbin Chen
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Zigui Huang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Yongzhi Wu
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Yongqi Huang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Binzhe Tang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Chenyan Long
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Xianwei Mo
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Jungang Liu
- Department of Gastrointestinal Surgery, Division of Colorectal and Anal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| |
Collapse
|
6
|
Deng D, Zhao L, Song H, Wang H, Cao H, Cui H, Zhou Y, Cui R. Microbiome analysis of gut microbiota in patients with colorectal polyps and healthy individuals. Sci Rep 2025; 15:7126. [PMID: 40021742 PMCID: PMC11871317 DOI: 10.1038/s41598-025-91626-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/21/2025] [Indexed: 03/03/2025] Open
Abstract
Colorectal polyps serve as the primary precursors for colorectal cancer. A close relationship has been observed between colorectal polyps and gut microbiota. However, the composition and role of the microbiome associated with tubular adenoma are not well understood. In this study, we prospectively evaluated alterations in gut microbiota among patients with colorectal polyps. A total of 60 subjects were enrolled in this study, including 30 patients with colorectal polyps (CP group) and 30 healthy controls (control group). The 16S rRNA sequencing was employed to characterize the gut microbiome in fecal samples. The results revealed that the beta diversity of the gut microbiota in the CP group significantly differs from that of the control group (p = 0.001). At the phylum level, the relative abundance of Bacteroides, Fusobacteria, and Proteobacteria was higher in the CP group compared to the control group (p < 0.05), whereas the relative abundance of Actinobacteria was higher in the control group in comparison to the CP group (p < 0.05). At the genus level, the abundance of Bacteroides increased in the CP group (p < 0.05), while Bifidobacterium declined in the CP group (p < 0.05). At the species level, the abundance of Clostridium perfringens, unidentified_Bacteroides, unidentified_Dorea, Escherichia coli, Clostridium ramosum, and Ruminococcus gnavus was higher (p < 0.05), whereas the abundance of Bifidobacterium adolescentis, unclassified_Bifidobacterium, Bifidobacterium longum, Faecalibacterium prausnitzii, and unidentified_Bifidobacterium is lower in CP group compared to the control group (p < 0.05). There was a structural imbalance in the composition of intestinal colonization flora for CP patients, characterized by a decrease in beneficial bacteria and an increase in harmful bacteria. Escherichia, Shigella, and Bacteroides may serve as promising biomarkers for early detection of colorectal polyps.
Collapse
Affiliation(s)
- Dayi Deng
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Lin Zhao
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Hui Song
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Houming Wang
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China
| | - Hengjie Cao
- Department of Surgery, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Huimin Cui
- Department of Surgery, Jinan Licheng District Hospital of Chinese Medicine, Jinan, 250000, China
| | - Yong Zhou
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China.
| | - Rong Cui
- Department of Surgery, Jiading Hospital of Traditional Chinese Medicine, 222 Bole Road, Jiading District, Shanghai, 201800, China.
| |
Collapse
|
7
|
Zhang H, Tian Y, Xu C, Chen M, Xiang Z, Gu L, Xue H, Xu Q. Crosstalk between gut microbiotas and fatty acid metabolism in colorectal cancer. Cell Death Discov 2025; 11:78. [PMID: 40011436 DOI: 10.1038/s41420-025-02364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy globally and the second leading cause of cancer-related mortality. Its development is a multifactorial and multistage process influenced by a dynamic interplay between gut microbiota, environmental factors, and fatty acid metabolism. Dysbiosis of intestinal microbiota and abnormalities in microbiota-associated metabolites have been implicated in colorectal carcinogenesis, highlighting the pivotal role of microbial and metabolic interactions. Fatty acid metabolism serves as a critical nexus linking dietary patterns with gut microbial activity, significantly impacting intestinal health. In CRC patients, reduced levels of short-chain fatty acids (SCFAs) and SCFA-producing bacteria have been consistently observed. Supplementation with SCFA-producing probiotics has demonstrated tumor-suppressive effects, while therapeutic strategies aimed at modulating SCFA levels have shown potential in enhancing the efficacy of radiation therapy and immunotherapy in both preclinical and clinical settings. This review explores the intricate relationship between gut microbiota, fatty acid metabolism, and CRC, offering insights into the underlying mechanisms and their potential translational applications. Understanding this interplay could pave the way for novel diagnostic, therapeutic, and preventive strategies in the management of CRC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yuan Tian
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Miaomiao Chen
- Department of Radiology, Huashan Hospital, National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200040, PR China
| | - Zeyu Xiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qing Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
8
|
Yang C, Qin LH, Li L, Wei QY, Long L, Liao JY. The causal relationship between the gut microbiota and endometrial cancer: a mendelian randomization study. BMC Cancer 2025; 25:248. [PMID: 39939905 PMCID: PMC11823214 DOI: 10.1186/s12885-025-13656-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Gut microbiota is associated with endometrial cancer (EC); however, the causal relationship remains unexplored. This study attempted to explore the relationship between gut microbiota and EC using Mendelian randomization (MR) methods. METHODS In this two-sample MR analysis, we used MiBioGen's gut microbiota data as the exposure and three datasets from European populations with EC as the outcome. The EC datasets included general EC, endometrioid histology, and non-endometrioid histology. Single nucleotide polymorphism (SNP) was used as the instrumental variable. Inverse variance weighted (IVW), multiplicative random effects IVW (MRE-IVW), Maximum likelihood (ML), MR Egger, MR-PRESSO, and the weighted median were used to perform MR analysis. Sensitivity analysis was conducted to assess the reliability of the results. RESULTS In this MR analysis of three EC datasets, specific gut microbiota were identified as potentially associated with different pathological types of EC. For general EC (ID: ebi-a-GCST006464), Family.Acidaminococcaceae (OR = 1.23, 95%CI: 1.02-1.48) and genus.Butyrivibrio (OR = 1.08, 95%CI: 1.01-1.16) were identified as risk factors, while genus.Ruminococcaceae UCG014 (OR = 0.82, 95%CI: 0.69-0.98) and genus.Turicibacter (OR = 0.84, 95%CI: 0.73-0.97) appeared to have protective effects. For endometrioid histology EC (ID: ebi-a-GCST006465), Family.Acidaminococcaceae (OR = 1.27, 95%CI: 1.01-1.59) and genus.Butyrivibrio (OR = 1.10, 95%CI: 1.01-1.19) were identified as risk factors, while several microbiota, including Family.Lactobacillaceae, genus.Coprococcus3, genus.Dorea, genus.Flavonifractor, genus.Lactobacillus, genus.Paraprevotella, and genus.Turicibacter, were identified as protective factors. For non-endometrioid histology EC (ID: ebi-a-GCST006466), Family.Rhodospirillaceae (OR = 1.41, 95%CI: 1.01-1.96) and genus.Peptococcus (OR = 1.43, 95%CI: 1.07-1.91) were identified as risk factors, while no significant protective factors were identified. CONCLUSIONS This two-sample MR study has identified gut microbiota with potential causal relationships with EC, varying by pathological type. These findings provide new insights into the pathogenesis of EC and suggest directions for future research on diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Chongze Yang
- Department of Radiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Lan-Hui Qin
- Department of Radiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Liwei Li
- Department of Gastroenterology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Qiu-Ying Wei
- Department of Radiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Liling Long
- Department of Radiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Jin-Yuan Liao
- Department of Radiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Zhuang Autonomous Region, Nanning, 530021, People's Republic of China.
| |
Collapse
|
9
|
Li G, Zhao D, Ouyang B, Chen Y, Zhao Y. Intestinal microbiota as biomarkers for different colorectal lesions based on colorectal cancer screening participants in community. Front Microbiol 2025; 16:1529858. [PMID: 39990152 PMCID: PMC11844352 DOI: 10.3389/fmicb.2025.1529858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction The dysregulation of intestinal microbiota has been implicated in the pathogenesis of colorectal cancer (CRC). However, the utilization of intestinal microbiota for identify the lesions in different procedures in CRC screening populations remains limited. Methods A total of 529 high-risk individuals who underwent CRC screening were included, comprising 13 advanced adenomas (Aade), 5 CRC, 59 non-advanced adenomas (Nade), 129 colon polyps (Pol), 99 cases of colorectal inflammatory disease (Inf), and 224 normal controls (Nor). 16S rRNA gene sequencing was used to profile the intestinal microbiota communities. The Gut Microbiota Health Index (GMHI) and average variation degree (AVD) were employed to assess the health status of the different groups. Results Our findings revealed that the Nor group exhibited significantly higher GMHIs and the lowest AVD compared to the four Lesion groups. The model incorporating 13 bacterial genera demonstrated optimal efficacy in distinguishing CRC and Aade from Nor, with an area under the curve (AUC) of 0.81 and a 95% confidence interval (CI) of 0.72 to 0.89. Specifically, the 55 bacterial genera combination model exhibited superior performance in differentiating CRC from Nor (AUC 0.98; 95% CI, 0.96-1), the 25 bacterial genera combination showed superior performance in distinguishing Aade from Nor (AUC 0.95). Additionally, the 27 bacterial genera combination demonstrated superior efficacy in differentiating Nade from Nor (AUC 0.82). The 13 bacterial genera combination exhibited optimal performance in distinguishing Inf from Nor (AUC 0.71). Discussion Our study has identified specific microbial biomarkers that can differentiate between colorectal lesions and healthy individuals. The intestinal microbiota markers identified may serve as valuable tools in community-based CRC screening programs.
Collapse
Affiliation(s)
- Gairui Li
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, China
- Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, China
| | - Dan Zhao
- Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, China
| | - Binfa Ouyang
- Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, China
| | - Yinggang Chen
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yashuang Zhao
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Amaro-da-Cruz A, Rubio-Tomás T, Álvarez-Mercado AI. Specific microbiome patterns and their association with breast cancer: the intestinal microbiota as a potential biomarker and therapeutic strategy. Clin Transl Oncol 2025; 27:15-41. [PMID: 38890244 PMCID: PMC11735593 DOI: 10.1007/s12094-024-03554-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer (BC) is one of the most diagnosed cancers in women. Based on histological characteristics, they are classified as non-invasive, or in situ (tumors located within the milk ducts or milk lobules) and invasive. BC may develop from in situ carcinomas over time. Determining prognosis and predicting response to treatment are essential tools to manage this disease and reduce its incidence and mortality, as well as to promote personalized therapy for patients. However, over half of the cases are not associated with known risk factors. In addition, some patients develop resistance to treatment and relapse. Therefore, it is necessary to identify new biomarkers and treatment strategies that improve existing therapies. In this regard, the role of the microbiome is being researched as it could play a role in carcinogenesis and the efficacy of BC therapies. This review aims to describe specific microbiome patterns associated with BC. For this, a literature search was carried out in PubMed database using the MeSH terms "Breast Neoplasms" and "Gastrointestinal Microbiome", including 29 publications. Most of the studies have focused on characterizing the gut or breast tissue microbiome of the patients. Likewise, studies in animal models and in vitro that investigated the impact of gut microbiota (GM) on BC treatments and the effects of the microbiome on tumor cells were included. Based on the results of the included articles, BC could be associated with an imbalance in the GM. This imbalance varied depending on molecular type, stage and grade of cancer, menopause, menarche, body mass index, and physical activity. However, a specific microbial profile could not be identified as a biomarker. On the other hand, some studies suggest that the GM may influence the efficacy of BC therapies. In addition, some microorganisms and bacterial metabolites could improve the effects of therapies or influence tumor development.
Collapse
Affiliation(s)
- Alba Amaro-da-Cruz
- Department of Chemical Engineering, Faculty of Science, University of Granada, 18071, Granada, Spain
| | - Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Ana I Álvarez-Mercado
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014, Granada, Spain.
- Institute of Nutrition and Food Technology, Biomedical Research Center, University of Granada, 18016, Armilla, Spain.
- Department of Pharmacology School of Pharmacy, University of Granada, 18071, Granada, Spain.
| |
Collapse
|
11
|
Zong Z, Zeng W, Li Y, Wang M, Cao Y, Cheng X, Jin Z, Mao S, Zhu X. Intratumor microbiota and colorectal cancer: Comprehensive and lucid review. Chin J Cancer Res 2024; 36:683-699. [PMID: 39802896 PMCID: PMC11724182 DOI: 10.21147/j.issn.1000-9604.2024.06.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
As a key component of tumor microenvironment, the microbiota has gradually played a key role in cancer research. Particularly in colorectal cancer, the specific population of microbiota within the tumor shows a strong association with the tumor type. Although the existence and potential role of microbiota in tumors have been recognized, the specific associations between the microbiota and tumor tissue and the mechanism of action still need to be further explored. This paper reviews the discovery, origin, and emerging role of the intratumor microbiota in the immune microenvironment and systematically outlines the oncogenic and metastasis-promoting strategies of the intratumor microbiota. Moreover, it comprehensively and holistically evaluates therapeutic strategies and prognostic performance on the basis of the intratumor microbiota, with the goal of providing strong support for future research and clinical practice.
Collapse
Affiliation(s)
- Zhen Zong
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Wenjuan Zeng
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yin Li
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Menghui Wang
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yuke Cao
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Xifu Cheng
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Zhenhua Jin
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Shengxun Mao
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Xingen Zhu
- Department of Neurosurgey, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
- Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang 330006, China
- Institute of Neuroscience, Nanchang University, Nanchang 330006, China
- Jiangxi Provincial Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006, China
| |
Collapse
|
12
|
Zhang J, Hou L, Ma L, Cai Z, Ye S, Liu Y, Ji P, Zuo Z, Zhao F. Real-time and programmable transcriptome sequencing with PROFIT-seq. Nat Cell Biol 2024; 26:2183-2194. [PMID: 39443694 PMCID: PMC11628399 DOI: 10.1038/s41556-024-01537-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
The high diversity and complexity of the eukaryotic transcriptome make it difficult to effectively detect specific transcripts of interest. Current targeted RNA sequencing methods often require complex pre-sequencing enrichment steps, which can compromise the comprehensive characterization of the entire transcriptome. Here we describe programmable full-length isoform transcriptome sequencing (PROFIT-seq), a method that enriches target transcripts while maintaining unbiased quantification of the whole transcriptome. PROFIT-seq employs combinatorial reverse transcription to capture polyadenylated, non-polyadenylated and circular RNAs, coupled with a programmable control system that selectively enriches target transcripts during sequencing. This approach achieves over 3-fold increase in effective data yield and reduces the time required for detecting specific pathogens or key mutations by 75%. We applied PROFIT-seq to study colorectal polyp development, revealing the intricate relationship between host immune responses and bacterial infection. PROFIT-seq offers a powerful tool for accurate and efficient sequencing of target transcripts while preserving overall transcriptome quantification, with broad applications in clinical diagnostics and targeted enrichment scenarios.
Collapse
Affiliation(s)
- Jinyang Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lingling Hou
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lianjun Ma
- Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhengyi Cai
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shujun Ye
- Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Liu
- Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peifeng Ji
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhenqiang Zuo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Fangqing Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
13
|
Liu Y, Lin XX, Hu SS, Zheng ED, Ye Y, Xu BB, Wu LC. The microbiota comparative analysis of the characteristics between colorectal adenomatous polyps and normal mucosal intestinal. Eur J Gastroenterol Hepatol 2024; 36:1305-1313. [PMID: 39166388 DOI: 10.1097/meg.0000000000002836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
OBJECTIVE The aim of this study is to systematically examine and compare the characteristics distinguishing colorectal adenomatous polyps from normal mucosal intestinal microbiota. METHODS A total of 30 specimens were obtained from patients diagnosed with colorectal adenomatous polyps (adenoma group) who underwent endoscopic removal at Wenzhou People's Hospital between September 2021 and November 2021. Concurrently, 30 normal mucosal specimens were collected from patients without adenomatous polyps (control group). Subsequently, microbiome total DNA extraction was carried out, followed by PCR amplification targeting the V3-V4 region of the 16S rDNA. High-throughput sequencing was conducted using the Illumina MiSeq platform. Subsequent to sequencing, bioinformatics analysis was used to assess the diversity, composition, and functional aspects of the intestinal microbiota in both study groups. RESULTS A notable dissimilarity in the microbiota structure was identified, specifically within the transverse colon, between these two groups ( P < 0.05). Species composition analysis revealed that Escherichia , Fusobacterium , and Bacteroides were predominant bacteria in both groups, with Escherichia and Enterobacter displaying significant differences at the genera level between the control group and the adenoma group ( P < 0.05). Correlation analysis and functional prediction demonstrated substantial disparities in interactions among dominant intestinal microbial genera within patients from both groups. Additionally, it was discovered that the intestinal microbiomes in patients in the adenoma group exhibited a significantly higher pathogenic potential. CONCLUSION Upon conducting a comprehensive analysis, it was discerned that the microbiota present in the transverse colon of the control group exhibited distinctive characteristics that may contribute to the maintenance of intestinal health.
Collapse
Affiliation(s)
- Ya Liu
- Department of Gastroenterology, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Bucher-Johannessen C, Senthakumaran T, Avershina E, Birkeland E, Hoff G, Bemanian V, Tunsjø H, Rounge TB. Species-level verification of Phascolarctobacterium association with colorectal cancer. mSystems 2024; 9:e0073424. [PMID: 39287376 PMCID: PMC11494908 DOI: 10.1128/msystems.00734-24] [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: 05/28/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
We have previously demonstrated an association between increased abundance of Phascolarctobacterium and colorectal cancer (CRC) and adenomas in two independent Norwegian cohorts. Here we seek to verify our previous findings using new cohorts and methods. In addition, we characterize lifestyle and sex specificity, the functional potential of the Phascolarctobacterium species, and their interaction with other microbial species. We analyze Phascolarctobacterium with 16S rRNA sequencing, shotgun metagenome sequencing, and species-specific qPCR, using 2350 samples from three Norwegian cohorts-CRCAhus, NORCCAP, and CRCbiome-and a large publicly available data set, curatedMetagenomicData. Using metagenome-assembled genomes from the CRCbiome study, we explore the genomic characteristics and functional potential of the Phascolarctobacterium pangenome. Three species of Phascolarctobacterium associated with adenoma/CRC were consistently detected by qPCR and sequencing. Positive associations with adenomas/CRC were verified for Phascolarctobacterium succinatutens and negative associations were shown for Phascolarctobacterium faecium and adenoma in curatedMetagenomicData. Men show a higher prevalence of P. succinatutens across cohorts. Co-occurrence among Phascolarctobacterium species was low (<6%). Each of the three species shows distinct microbial composition and forms distinct correlation networks with other bacterial taxa, although Dialister invisus was negatively correlated to all investigated Phascolarctobacterium species. Pangenome analyses showed P. succinatutens to be enriched for genes related to porphyrin metabolism and degradation of complex carbohydrates, whereas glycoside hydrolase enzyme 3 was specific to P. faecium.IMPORTANCEUntil now Phascolarctobacterium has been going under the radar as a CRC-associated genus despite having been noted, but overseen, as such for over a decade. We found not just one, but two species of Phascolarctobacterium to be associated with CRC-Phascolarctobacterium succinatutens was more abundant in adenoma/CRC, while Phascolarctobacterium faecium was less abundant in adenoma. Each of them represents distinct communities, constituted by specific microbial partners and metabolic capacities-and they rarely occur together in the same patients. We have verified that P. succinatutens is increased in adenoma and CRC and this species should be recognized among the most important CRC-associated bacteria.
Collapse
Affiliation(s)
- Cecilie Bucher-Johannessen
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
- Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
- Center for Bioinformatics, Department of Pharmacy, University of Oslo, Oslo, Norway
| | | | - Ekaterina Avershina
- Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
- Center for Bioinformatics, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Einar Birkeland
- Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Geir Hoff
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
- Telemark Hospital, Skien, Norway
| | - Vahid Bemanian
- Department of Pathology, Akershus University Hospital, Lørenskog, Norway
| | - Hege Tunsjø
- Department of Life Sciences and Health, Oslo Metropolitan University, Oslo, Norway
| | - Trine B. Rounge
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
- Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
- Center for Bioinformatics, Department of Pharmacy, University of Oslo, Oslo, Norway
| |
Collapse
|
15
|
García Menéndez G, Sichel L, López MDC, Hernández Y, Arteaga E, Rodríguez M, Fleites V, Fernández LT, Cano RDJ. From colon wall to tumor niche: Unraveling the microbiome's role in colorectal cancer progression. PLoS One 2024; 19:e0311233. [PMID: 39436937 PMCID: PMC11495602 DOI: 10.1371/journal.pone.0311233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Colorectal cancer (CRC) is influenced by perturbations in the colonic microbiota, characterized by an imbalance favoring pathogenic bacteria over beneficial ones. This dysbiosis contributes to CRC initiation and progression through mechanisms such as carcinogenic metabolite production, inflammation induction, DNA damage, and oncogenic signaling activation. Understanding the role of external factors in shaping the colonic microbiota is crucial for mitigating CRC progression. This study aims to elucidate the gut microbiome's role in CRC progression by analyzing paired tumor and mucosal tissue samples obtained from the colon walls of 17 patients. Through sequencing of the V3-V4 region of the 16S rRNA gene, we characterized the tumor microbiome and assessed its association with clinical variables. Our findings revealed a significant reduction in alpha diversity within tumor samples compared to paired colon biopsy samples, indicating a less diverse microbial environment within the tumor microenvironment. While both tissues exhibited dominance of similar bacterial phyla, their relative abundances varied, suggesting potential colon-specific effects. Fusobacteriota enrichment, notably in the right colon, may be linked to MLH1 deficiency. Taxonomy analysis identified diverse bacterial genera, with some primarily associated with the colon wall and others unique to this region. Conversely, several genera were exclusively expressed in tumor tissue. Functional biomarker analysis identified three key genes with differential abundance between tumor microenvironment and colon tissue, indicating distinct metabolic activities. Functional biomarker analysis revealed three key genes with differential abundance: K11076 (putrescine transport system) and K10535 (nitrification) were enriched in the tumor microenvironment, while K11329 (SasA-RpaAB circadian timing mediator) dominated colon tissue. Metabolic pathway analysis linked seven metabolic pathways to the microbiome. Collectively, these findings highlight significant gut microbiome alterations in CRC and strongly suggest that long-term dysbiosis profoundly impacts CRC progression.
Collapse
Affiliation(s)
- Gissel García Menéndez
- Pathology Department, Clinical Hospital Hermanos Ameijeiras, Centro Habana, La Habana, Cuba
| | - Liubov Sichel
- Stellar Biotics, LLC, Rockleigh, New Jersey, United States of America
| | | | - Yasel Hernández
- Pathology Department, Clinical Hospital Hermanos Ameijeiras, Centro Habana, La Habana, Cuba
| | - Ernesto Arteaga
- Pathology Department, Clinical Hospital Hermanos Ameijeiras, Centro Habana, La Habana, Cuba
| | - Marisol Rodríguez
- Pathology Department, Clinical Hospital Hermanos Ameijeiras, Centro Habana, La Habana, Cuba
| | - Vilma Fleites
- Oncology Department Clinical Hospital Hermanos Ameijeiras, Centro Habana, La Habana, Cuba
| | - Lipsy Teresa Fernández
- Surgery Department Clinical Hospital Hermanos Ameijeiras, Centro Habana, La Habana, Cuba
| | - Raúl De Jesus Cano
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA, United States of America
| |
Collapse
|
16
|
Chen G, Ren Q, Zhong Z, Li Q, Huang Z, Zhang C, Yuan H, Feng Z, Chen B, Wang N, Feng Y. Exploring the gut microbiome's role in colorectal cancer: diagnostic and prognostic implications. Front Immunol 2024; 15:1431747. [PMID: 39483461 PMCID: PMC11524876 DOI: 10.3389/fimmu.2024.1431747] [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: 05/12/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
The intricate interplay between the gut microbiome and colorectal cancer (CRC) presents novel avenues for early diagnosis and prognosis, crucial for improving patient outcomes. This comprehensive review synthesizes current findings on the gut microbiome's contribution to CRC pathogenesis, highlighting its potential as a biomarker for non-invasive CRC screening strategies. We explore the mechanisms through which the microbiome influences CRC, including its roles in inflammation, metabolism, and immune response modulation. Furthermore, we assess the viability of microbial signatures as predictive tools for CRC prognosis, offering insights into personalized treatment approaches. Our analysis underscores the necessity for advanced metagenomic studies to elucidate the complex microbiome-CRC nexus, aiming to refine diagnostic accuracy and prognostic assessment in clinical settings. This review propels forward the understanding of the microbiome's diagnostic and prognostic capabilities, paving the way for microbiome-based interventions in CRC management.
Collapse
Affiliation(s)
- Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qing Ren
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zilan Zhong
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianfan Li
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiqiang Huang
- The First Clinical College of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hongchao Yuan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zixin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
González A, Fullaondo A, Odriozola I, Odriozola A. Microbiota and beneficial metabolites in colorectal cancer. ADVANCES IN GENETICS 2024; 112:367-409. [PMID: 39396841 DOI: 10.1016/bs.adgen.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related death worldwide. In recent years, the impact of the gut microbiota on the development of CRC has become clear. The gut microbiota is the community of microorganisms living in the gut symbiotic relationship with the host. These microorganisms contribute to the development of CRC through various mechanisms that are not yet fully understood. Increasing scientific evidence suggests that metabolites produced by the gut microbiota may influence CRC development by exerting protective and deleterious effects. This article reviews the metabolites produced by the gut microbiota, which are derived from the intake of complex carbohydrates, proteins, dairy products, and phytochemicals from plant foods and are associated with a reduced risk of CRC. These metabolites include short-chain fatty acids (SCFAs), indole and its derivatives, conjugated linoleic acid (CLA) and polyphenols. Each metabolite, its association with CRC risk, the possible mechanisms by which they exert anti-tumour functions and their relationship with the gut microbiota are described. In addition, other gut microbiota-derived metabolites that are gaining importance for their role as CRC suppressors are included.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Iñaki Odriozola
- Health Department of Basque Government, Donostia-San Sebastián, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
18
|
Liu G, Su L, Kong C, Huang L, Zhu X, Zhang X, Ma Y, Wang J. Improved diagnostic efficiency of CRC subgroups revealed using machine learning based on intestinal microbes. BMC Gastroenterol 2024; 24:315. [PMID: 39289618 PMCID: PMC11409688 DOI: 10.1186/s12876-024-03408-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common cancer that causes millions of deaths worldwide each year. At present, numerous studies have confirmed that intestinal microbes play a crucial role in the process of CRC. Additionally, studies have shown that CRC can be divided into several consensus molecular subtypes (CMS) based on tumor gene expression, and CRC microbiomes have been reported related to CMS. However, most previous studies on intestinal microbiome of CRC have only compared patients with healthy controls, without classifying of CRC patients based on intestinal microbial composition. RESULTS In this study, a CRC cohort including 339 CRC samples and 333 healthy controls was selected as the discovery set, and the CRC samples were divided into two subgroups (234 Subgroup1 and 105 Subgroup2) using PAM clustering algorithm based on the intestinal microbial composition. We found that not only the microbial diversity was significantly different (Shannon index, p-value < 0.05), but also 129 shared genera altered (p-value < 0.05) between the two CRC subgroups, including several marker genera in CRC, such as Fusobacterium and Bacteroides. A random forest algorithm was used to construct diagnostic models, which showed significantly higher efficiency when the CRC samples were divided into subgroups. Then an independent cohort including 187 CRC samples (divided into 153 Subgroup1 and 34 Subgroup2) and 123 healthy controls was chosen to validate the models, and confirmed the results. CONCLUSIONS These results indicate that the divided CRC subgroups can improve the efficiency of disease diagnosis, with various microbial composition in the subgroups.
Collapse
Affiliation(s)
- Guang Liu
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Guangdong Hongyuan Pukang Medical Technology Co, Ltd, Guangzhou, 510000, China
| | - Lili Su
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Guangdong Hongyuan Pukang Medical Technology Co, Ltd, Guangzhou, 510000, China
| | - Cheng Kong
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Liang Huang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510000, China
| | - Xiaoyan Zhu
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xuanping Zhang
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Jiayin Wang
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
19
|
Zhou C, Ye X, Liu Z, Liu T, Li S, Yang J, Wei J, Yu P, Jia R, Zhao W. Dissecting the causal links between gut microbiome, immune traits and polyp using genetic evidence. Front Immunol 2024; 15:1431990. [PMID: 39346904 PMCID: PMC11427361 DOI: 10.3389/fimmu.2024.1431990] [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: 05/13/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024] Open
Abstract
Background Previous research has demonstrated an association between gut microbiota and immune status with the development of several diseases. However, whether these factors contribute to polyps remains unclear. This study aims to use Mendelian randomization (MR) to investigate the causal relationship between gut microbiota and 4 types of polyps (nasal, gallbladder, colon, and gastric polyps), as well as to analyze the mediating role of immune traits. Methods This study utilized large-scale GWAS meta-analyses of gut microbiota (MiBioGen Consortium), 731 immune traits, and 4 types of polyps (one from the FinnGen Consortium and three from the NBDC Human Database). Univariate MR with the inverse variance weighted (IVW) estimation method was employed as the primary analytical approach. A two-step MR analysis was performed to identify potential mediating immune traits. Additionally, multivariable MR approach based on Bayesian model averaging (MR-BMA) was employed to further prioritize gut microbiota and immune traits associated with polyp development. Results Based on IVW method in univariate MR analysis, we identified 39 gut microbial taxa and 135 immune traits significantly causally associated with at least one type of polyp. For nasal polyps, 13 microbial taxa and 61 immune traits were causally associated. After false discovery rate (FDR) correction, CD3 on Central Memory CD8+ T cells and CD3 on CD4 regulatory T cells remained significant. MR-BMA identified 4 gut microbial taxa and 4 immune traits as high priority. For gallbladder polyps, 9 microbial taxa and 30 immune traits were causally associated. MR-BMA identified 8 microbial taxa and 6 immune traits as higher importance. For colon polyps, 6 microbial taxa and 21 immune traits were causally associated. MR-BMA identified 4 microbial taxa and 3 immune traits as higher importance. For gastric polyps, 12 microbial taxa and 33 immune traits were causally associated. Actinobacteria remained significant after FDR correction, and MR-BMA identified 7 gut microbial taxa and 6 immune traits as high priority. We identified 16 causal pathways with mediator directions consistent with the direction of gut microbiome-polyp association. Of these, 6 pathways were associated with the mechanism of nasal polyps, 1 with gallbladder polyps, 2 with colon polyps, and 7 with gastric polyps. Conclusions Our findings shed light on the causal relationships between gut microbiota, immune traits, and polyp development, underscoring the crucial roles of gut microbiota and immune status in polypogenesis. Furthermore, these findings suggest potential applications in polyp prevention, early screening, and the development of effective strategies to reduce polyp risk.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Gastroenterology, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Xiaofeng Ye
- Department of Gastroenterology, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Zhinuo Liu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tong Liu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Shanzheng Li
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jinqiu Yang
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jingjing Wei
- Heart Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Yu
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ran Jia
- The First College of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Wenxia Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
20
|
Kamath HS, Shukla R, Shah U, Patel S, Das S, Chordia A, Satish P, Ghosh D. Role of Gut Microbiota in Predisposition to Colon Cancer: A Narrative Review. Indian J Microbiol 2024; 64:1-13. [PMID: 39282181 PMCID: PMC11399513 DOI: 10.1007/s12088-024-01242-5] [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: 09/20/2023] [Accepted: 02/28/2024] [Indexed: 09/18/2024] Open
Abstract
Globally, colorectal cancer (CRC) is a leading cause of cancer-related mortality. Dietary habits, inflammation, hereditary characteristics, and gut microbiota are some of its causes. The gut microbiota, a diverse population of bacteria living in the digestive system, has an impact on a variety of parameters, including inflammation, DNA damage, and immune response. The gut microbiome has a significant role in colon cancer susceptibility. Many studies have highlighted dysbiosis, an imbalance in the gut microbiota's makeup, as a major factor in colon cancer susceptibility. Dysbiosis has the potential to produce toxic metabolites and pro-inflammatory substances, which can hasten the growth of tumours. The ability of the gut microbiota to affect the host's immune system can also influence whether cancer develops or not. By better comprehending these complex interactions between colon cancer predisposition and gut flora, new preventive and therapeutic techniques might be developed. Targeting the gut microbiome with dietary modifications, probiotics, or faecal microbiota transplantation may offer cutting-edge approaches to reducing the risk of colon cancer and improving patient outcomes. The complex connection between the makeup of the gut microbiota and the emergence of colorectal cancer is explored in this narrative review.
Collapse
Affiliation(s)
- Hattiangadi Shruthi Kamath
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Rushikesh Shukla
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Urmil Shah
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Siddhi Patel
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Soumyajit Das
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Ayush Chordia
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Poorvikha Satish
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Dibyankita Ghosh
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| |
Collapse
|
21
|
Han S, Zhuang J, Song Y, Wu X, Yu X, Tao Y, Chu J, Qu Z, Wu Y, Han S, Yang X. Gut microbial subtypes and clinicopathological value for colorectal cancer. Cancer Med 2024; 13:e70180. [PMID: 39234654 PMCID: PMC11375334 DOI: 10.1002/cam4.70180] [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: 02/03/2024] [Revised: 08/03/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Gut bacteria are related to colorectal cancer (CRC) and its clinicopathologic characteristics. OBJECTIVE To develop gut bacterial subtypes and explore potential microbial targets for CRC. METHODS Stool samples from 914 volunteers (376 CRCs, 363 advanced adenomas, and 175 normal controls) were included for 16S rRNA sequencing. Unsupervised learning was used to generate gut microbial subtypes. Gut bacterial community composition and clustering effects were plotted. Differences of gut bacterial abundance were analyzed. Then, the association of CRC-associated bacteria with subtypes and the association of gut bacteria with clinical information were assessed. The CatBoost models based on gut differential bacteria were constructed to identify the diseases including CRC and advanced adenoma (AA). RESULTS Four gut microbial subtypes (A, B, C, D) were finally obtained via unsupervised learning. The characteristic bacteria of each subtype were Escherichia-Shigella in subtype A, Streptococcus in subtype B, Blautia in subtype C, and Bacteroides in subtype D. Clinical information (e.g., free fatty acids and total cholesterol) and CRC pathological information (e.g., tumor depth) varied among gut microbial subtypes. Bacilli, Lactobacillales, etc., were positively correlated with subtype B. Positive correlation of Blautia, Lachnospiraceae, etc., with subtype C and negative correlation of Coriobacteriia, Coriobacteriales, etc., with subtype D were found. Finally, the predictive ability of CatBoost models for CRC identification was improved based on gut microbial subtypes. CONCLUSION Gut microbial subtypes provide characteristic gut bacteria and are expected to contribute to the diagnosis of CRC.
Collapse
Affiliation(s)
- Shuwen Han
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
- Institut Catholique de Lille, Junia (ICL), Université Catholique de Lille, Laboratoire Interdisciplinaire des Transitions de Lille (LITL), Lille, France
| | - Jing Zhuang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Yifei Song
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Xinyue Wu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Xiaojian Yu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Ye Tao
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Shanghai Biozeron Biotechnology Co., Ltd., Shanghai, China
| | - Jian Chu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Zhanbo Qu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Yinhang Wu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Shugao Han
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Yang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| |
Collapse
|
22
|
Yin LL, Qi PQ, Hu YF, Fu XJ, He RS, Wang MM, Deng YJ, Xiong SY, Yu QW, Hu JP, Zhou L, Zhou ZB, Xiong Y, Deng H. Dysbiosis promotes recurrence of adenomatous polyps in the distal colorectum. World J Gastrointest Oncol 2024; 16:3600-3623. [PMID: 39171160 PMCID: PMC11334022 DOI: 10.4251/wjgo.v16.i8.3600] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/19/2024] [Accepted: 06/14/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Colorectal polyps, which are characterized by a high recurrence rate, represent preneoplastic conditions of the intestine. Due to unclear mechanisms of pathogenesis, first-line therapies for non-hereditary recurrent colorectal polyps are limited to endoscopic resection. Although recent studies suggest a mechanistic link between intestinal dysbiosis and polyps, the exact compositions and roles of bacteria in the mucosa around the lesions, rather than feces, remain unsettled. AIM To clarify the composition and diversity of bacteria in the mucosa surrounding or 10 cm distal to recurrent intestinal polyps. METHODS Mucosal samples were collected from four patients consistently with adenomatous polyps (Ade), seven consistently with non-Ade (Pol), ten with current Pol but previous Ade, and six healthy individuals, and bacterial patterns were evaluated by 16S rDNA sequencing. Linear discriminant analysis and Student's t-tests were used to identify the genus-level bacteria differences between groups with different colorectal polyp phenotypes. Pearson's correlation coefficients were used to evaluate the correlation between intestinal bacteria at the genus level and clinical indicators. RESULTS The results confirmed a decreased level of probiotics and an enrichment of pathogenic bacteria in patients with all types of polyps compared to healthy individuals. These changes were not restricted to the mucosa within 0.5 cm adjacent to the polyps, but also existed in histologically normal tissue 10 cm distal from the lesions. Significant differences in bacterial diversity were observed in the mucosa from individuals with normal conditions, Pol, and Ade. Increased abundance of Gram-negative bacteria, including Klebsiella, Plesiomonas, and Cronobacter, was observed in Pol group and Ade group, suggesting that resistance to antibiotics may be one risk factor for bacterium-related harmful environment. Meanwhile, age and gender were linked to bacteria changes, indicating the potential involvement of sex hormones. CONCLUSION These preliminary results support intestinal dysbiosis as an important risk factor for recurrent polyps, especially adenoma. Targeting specific pathogenic bacteria may attenuate the recurrence of polyps.
Collapse
Affiliation(s)
- Li-Li Yin
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Ping-Qian Qi
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yun-Fei Hu
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Xiao-Jun Fu
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Rui-Shan He
- The Second College of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Meng-Meng Wang
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yan-Juan Deng
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Su-Yi Xiong
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Qi-Wen Yu
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Jin-Ping Hu
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Lv Zhou
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zhi-Bin Zhou
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Ying Xiong
- Department of General Medicine, The Second College of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi Province, China
| | - Huan Deng
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang University, Nanchang 330006, Jiangxi Province, China
- The Ministry of Education Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi Province, China
| |
Collapse
|
23
|
Baas FS, Brusselaers N, Nagtegaal ID, Engstrand L, Boleij A. Navigating beyond associations: Opportunities to establish causal relationships between the gut microbiome and colorectal carcinogenesis. Cell Host Microbe 2024; 32:1235-1247. [PMID: 39146796 DOI: 10.1016/j.chom.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024]
Abstract
The gut microbiota has been recognized as an important determinant in the initiation and progression of colorectal cancer (CRC), with recent studies shining light on the molecular mechanisms that may contribute to the interactions between microbes and the CRC microenvironment. Despite the increasing wealth of associations being established in the field, proving causality remains challenging. Obstacles include the high variability of the microbiome and its context, both across individuals and across time. Additionally, there is a lack of large and representative cohort studies with long-term follow-up and/or appropriate sampling methods for studying the mucosal microbiome. Finally, most studies focus on CRC, whereas interactions between host and bacteria in early events in carcinogenesis remain elusive, reinforced by the heterogeneity of CRC development. Here, we discuss these current most prominent obstacles, the recent developments, and research needs.
Collapse
Affiliation(s)
- Floor S Baas
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nele Brusselaers
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Karolinska Hospital, Stockholm, Sweden; Global Health Institute, University of Antwerp, Antwerp, Belgium
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Karolinska Hospital, Stockholm, Sweden
| | - Annemarie Boleij
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
24
|
Sun J, Zhao J, Zhou S, Li X, Li T, Wang L, Yuan S, Chen D, Law PJ, Larsson SC, Farrington SM, Houlston RS, Dunlop MG, Theodoratou E, Li X. Systematic investigation of genetically determined plasma and urinary metabolites to discover potential interventional targets for colorectal cancer. J Natl Cancer Inst 2024; 116:1303-1312. [PMID: 38648753 PMCID: PMC11308169 DOI: 10.1093/jnci/djae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/27/2024] [Accepted: 04/13/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND We aimed to identify plasma and urinary metabolites related to colorectal cancer (CRC) risk and elucidate their mediator role in the associations between modifiable risk factors and CRC. METHODS Metabolite quantitative trait loci were derived from 2 published metabolomics genome-wide association studies, and summary-level data were extracted for 651 plasma metabolites and 208 urinary metabolites. Genetic associations with CRC were obtained from a large-scale genome-wide association study meta-analysis (100 204 cases, 154 587 controls) and the FinnGen cohort (4957 cases, 304 197 controls). Mendelian randomization and colocalization analyses were performed to evaluate the causal roles of metabolites in CRC. Druggability evaluation was employed to prioritize potential therapeutic targets. Multivariable Mendelian randomization and mediation estimation were conducted to elucidate the mediating effects of metabolites on the associations between modifiable risk factors and CRC. RESULTS The study identified 30 plasma metabolites and 4 urinary metabolites for CRC. Plasma sphingomyelin and urinary lactose, which were positively associated with CRC risk, could be modulated by drug interventions (ie, olipudase alfa, tilactase). Thirteen modifiable risk factors were associated with 9 metabolites, and 8 of these modifiable risk factors were associated with CRC risk. These 9 metabolites mediated the effect of modifiable risk factors (Actinobacteria, body mass index, waist to hip ratio, fasting insulin, smoking initiation) on CRC. CONCLUSION This study identified key metabolite biomarkers associated with CRC and elucidated their mediator roles in the associations between modifiable risk factors and CRC. These findings provide new insights into the etiology and potential therapeutic targets for CRC and the etiological pathways of modifiable environmental factors with CRC.
Collapse
Affiliation(s)
- Jing Sun
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianhui Zhao
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyun Zhou
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinxuan Li
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tengfei Li
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lijuan Wang
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susan M Farrington
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Malcolm G Dunlop
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, and Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Intarajak T, Udomchaiprasertkul W, Khoiri AN, Sutheeworapong S, Kusonmano K, Kittichotirat W, Thammarongtham C, Cheevadhanarak S. Distinct gut microbiomes in Thai patients with colorectal polyps. World J Gastroenterol 2024; 30:3336-3355. [PMID: 39086748 PMCID: PMC11287419 DOI: 10.3748/wjg.v30.i27.3336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Colorectal polyps that develop via the conventional adenoma-carcinoma sequence [e.g., tubular adenoma (TA)] often progress to malignancy and are closely associated with changes in the composition of the gut microbiome. There is limited research concerning the microbial functions and gut microbiomes associated with colorectal polyps that arise through the serrated polyp pathway, such as hyperplastic polyps (HP). Exploration of microbiome alterations associated with HP and TA would improve the understanding of mechanisms by which specific microbes and their metabolic pathways contribute to colorectal carcinogenesis. AIM To investigate gut microbiome signatures, microbial associations, and microbial functions in HP and TA patients. METHODS Full-length 16S rRNA sequencing was used to characterize the gut microbiome in stool samples from control participants without polyps [control group (CT), n = 40], patients with HP (n = 52), and patients with TA (n = 60). Significant differences in gut microbiome composition and functional mechanisms were identified between the CT group and patients with HP or TA. Analytical techniques in this study included differential abundance analysis, co-occurrence network analysis, and differential pathway analysis. RESULTS Colorectal cancer (CRC)-associated bacteria, including Streptococcus gallolyticus (S. gallolyticus), Bacteroides fragilis, and Clostridium symbiosum, were identified as characteristic microbial species in TA patients. Mediterraneibacter gnavus, associated with dysbiosis and gastrointestinal diseases, was significantly differentially abundant in the HP and TA groups. Functional pathway analysis revealed that HP patients exhibited enrichment in the sulfur oxidation pathway exclusively, whereas TA patients showed dominance in pathways related to secondary metabolite biosynthesis (e.g., mevalonate); S. gallolyticus was a major contributor. Co-occurrence network and dynamic network analyses revealed co-occurrence of dysbiosis-associated bacteria in HP patients, whereas TA patients exhibited co-occurrence of CRC-associated bacteria. Furthermore, the co-occurrence of SCFA-producing bacteria was lower in TA patients than HP patients. CONCLUSION This study revealed distinct gut microbiome signatures associated with pathways of colorectal polyp development, providing insights concerning the roles of microbial species, functional pathways, and microbial interactions in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Thoranin Intarajak
- Bioinformatics Unit, Chulabhorn Royal Academy, Lak Si 10210, Bangkok, Thailand
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | | | - Ahmad Nuruddin Khoiri
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Sawannee Sutheeworapong
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Kanthida Kusonmano
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology, and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Chinae Thammarongtham
- National Center for Genetic Engineering and Biotechnology, King Mongkut's University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| | - Supapon Cheevadhanarak
- Systems Biology and Bioinformatics Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
- School of Bioresources and Technology, King Mongkut’s University of Technology Thonburi, Bank Khun Thian 10150, Bangkok, Thailand
- Fungal Biotechnology Unit, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bang Khun Thian 10150, Bangkok, Thailand
| |
Collapse
|
26
|
Lang Y, Zhong C, Guo L, Liu Z, Zuo D, Chen X, Ding L, Huang B, Li B, Yuan Y, Niu Y, Qiu J, Qian C. Monoacylglycerol acyltransferase-2 inhibits colorectal carcinogenesis in APC min+/- mice. iScience 2024; 27:110205. [PMID: 39055928 PMCID: PMC11269928 DOI: 10.1016/j.isci.2024.110205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 04/18/2024] [Accepted: 06/04/2024] [Indexed: 07/28/2024] Open
Abstract
Monoacylglycerol acyltransferase-2 (MOGAT2), encodes MOGAT enzyme in the re-synthesis of triacylglycerol and protects from metabolism disorders. While, its precise involvement in colorectal cancer (CRC) progression remains inadequately understood. Our study demonstrated that knockout of Mogat2 in Apcmin/+ mice expedited intestinal tumor growth and progression, indicating that Mogat2 plays a tumor-suppressing role in CRC. Mechanically, Mogat2 deletion resulted in a significant alter the gut microbiota, while Fecal Microbiota Transplantation (FMT) experiments demonstrated that the gut microbiota in Mogat2 deleted mice promoted tumor growth. Furthermore, we identified Mogat2 as a functional regulator suppressing CRC cell proliferation and tumor growth by inhibiting the NF-κB signaling pathway in vivo. Collectively, these results provide novel insights into the protective double roles of Mogat2, inhibiting of NF-κB pathway and keeping gut microbiota homeostasis in colorectal cancer, which may help the development of novel cancer treatments for CRC.
Collapse
Affiliation(s)
- Yanhong Lang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Chengrui Zhong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Lingling Guo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Zhijie Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Dinglan Zuo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Xi Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Liuyan Ding
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Bijun Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Binkui Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Yi Niu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jiliang Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Liver Surgery, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou 510060, P.R. China
| | - Chaonan Qian
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Radiation Oncology, Guangzhou Concord Cancer Center, 9 Ciji Road, Huangpu District, Guangzhou 510555, P.R. China
| |
Collapse
|
27
|
Wang X, Zhang Q, Xu R, Li X, Hong Z. Research progress on the correlation between intestinal flora and colorectal cancer. Front Oncol 2024; 14:1416806. [PMID: 39087025 PMCID: PMC11288818 DOI: 10.3389/fonc.2024.1416806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal malignancies in the world. With the rapid pace of life and changes in diet structure, the incidence and mortality of CRC increase year by year posing a serious threat to human health. As the most complex and largest microecosystem in the human body, intestinal microecology is closely related to CRC. It is an important factor that affects and participates in the occurrence and development of CRC. Advances in next-generation sequencing technology and metagenomics have provided new insights into the ecology of gut microbes. It also helps to link intestinal flora with CRC, and the relationship between intestinal flora and CRC can be continuously understood from different levels. This paper summarizes the relationship between intestinal flora and CRC and its potential role in the diagnosis of CRC providing evidence for early screening and treatment of CRC.
Collapse
Affiliation(s)
- Xinyu Wang
- The Health Management Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qian Zhang
- Department of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Rongxuan Xu
- Department of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaofeng Li
- Department of Public Health, Dalian Medical University, Dalian, Liaoning, China
| | - Zhijun Hong
- The Health Management Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
28
|
Chen Q, Huang X, Zhang H, Jiang X, Zeng X, Li W, Su H, Chen Y, Lin F, Li M, Gu X, Jin H, Wang R, Diao D, Wang W, Li J, Wei S, Zhang W, Liu W, Huang Z, Deng Y, Luo W, Liu Z, Zhang B. Characterization of tongue coating microbiome from patients with colorectal cancer. J Oral Microbiol 2024; 16:2344278. [PMID: 38686186 PMCID: PMC11057396 DOI: 10.1080/20002297.2024.2344278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 04/13/2024] [Indexed: 05/02/2024] Open
Abstract
Background Tongue coating microbiota has aroused particular interest in profiling oral and digestive system cancers. However, little is known on the relationship between tongue coating microbiome and colorectal cancer (CRC). Methods Metagenomic shotgun sequencing was performed on tongue coating samples collected from 30 patients with CRC, 30 patients with colorectal polyps (CP), and 30 healthy controls (HC). We further validated the potential of the tongue coating microbiota to predict the CRC by a random forest model. Results We found a greater species diversity in CRC samples, and the nucleoside and nucleotide biosynthesis pathway was more apparent in the CRC group. Importantly, various species across participants jointly shaped three distinguishable fur types.The tongue coating microbiome profiling data gave an area under the receiver operating characteristic curve (AUC) of 0.915 in discriminating CRC patients from control participants; species such as Atopobium rimae, Streptococcus sanguinis, and Prevotella oris aided differentiation of CRC patients from healthy participants. Conclusion These results elucidate the use of tongue coating microbiome in CRC patients firstly, and the fur-types observed contribute to a better understanding of the microbial community in human. Furthermore, the tongue coating microbiota-based biomarkers provide a valuable reference for CRC prediction and diagnosis.
Collapse
Affiliation(s)
- Qubo Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xiaoting Huang
- Medical Research Center, Huazhong University of Science and Technology Union Shenzhen, Shenzhen, China
| | - Haiyan Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuanting Jiang
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Xuan Zeng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Wanhua Li
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hairong Su
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengye Lin
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Man Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xiangyu Gu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huihui Jin
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruohan Wang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dechang Diao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Colorectal surgery of Guangdong Provincial Hospital of TCM, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Gastrointestinal Surgery Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jin Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Gastrointestinal Surgery Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sufen Wei
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weizheng Zhang
- Medical Laboratory, Guangzhou Cadre Health Management Center, Guangzhou No.11 People’s Hospital, Guangzhou, China
| | - Wofeng Liu
- Medical Laboratory, Guangzhou Cadre Health Management Center, Guangzhou No.11 People’s Hospital, Guangzhou, China
| | - Zhiping Huang
- Information Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yusheng Deng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Wen Luo
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Zuofeng Liu
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Beiping Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
29
|
Hu S, Tang C, Wang L, Feng F, Li X, Sun M, Yao L. Causal relationship between gut microbiota and differentiated thyroid cancer: a two-sample Mendelian randomization study. Front Oncol 2024; 14:1375525. [PMID: 38737897 PMCID: PMC11082393 DOI: 10.3389/fonc.2024.1375525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
Background The gut microbiota has been significantly associated with differentiated thyroid cancer (DTC). However, the causal relationship between the gut microbiota and DTC remains unexplored. Methods Genome-wide association study (GWAS) summary databases were utilized to select exposures and outcomes. The Mendelian randomization (MR) method was employed to investigate the causal relationship between the gut microbiota and DTC. A sensitivity analysis was performed to assess the reliability of the findings. Results Four bacterial traits were associated with the risk of DTC: Class Mollicutes [odds ratio (OR) = 10.953, 95% confidence interval (95% CI): 2.333-51.428, p = 0.002], Phylum Tenericutes (OR = 10.953, 95% CI: 2.333-51.428, p = 0.002), Genus Eggerthella (OR = 3.219, 95% CI: 1.033-10.024, p = 0.044), and Order Rhodospirillales (OR = 2.829, 95% CI: 1.096-7.299, p = 0.032). The large 95% CI range for the Class Mollicutes and the Phylum Tenericutes may be attributed to the small sample size. Additionally, four other bacterial traits were negatively associated with DTC: Genus Eubacterium fissicatena group (OR = 0.381, 95% CI: 0.148-0.979, p = 0.045), Genus Lachnospiraceae UCG008 (OR = 0.317, 95% CI: 0.125-0.801, p = 0.015), Genus Christensenellaceae R-7 group (OR = 0.134, 95% CI: 0.020-0.886, p = 0.037), and Genus Escherichia Shigella (OR = 0.170, 95% CI: 0.037-0.769, p = 0.021). Conclusion These findings contribute to our understanding of the pathological mechanisms underlying DTC and provide novel insights for the clinical treatment of DTC.
Collapse
Affiliation(s)
- Shaojun Hu
- Department of Oncology, Suzhou Ninth People’s Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Chuangang Tang
- Department of Breast Surgery, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, China
| | - Ling Wang
- Department of Critical Care Medicine, The People’s Hospital of Huaiyin, Jinan, China
| | - Fang Feng
- Department of Oncology, Suzhou Ninth People’s Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Xiaoxin Li
- Department of Pathology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, China
| | - Mingyu Sun
- Department of Breast Surgery, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, China
| | - Lijun Yao
- Department of Oncology, Suzhou Ninth People’s Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| |
Collapse
|
30
|
Alahdal H, Almuneef G, Alkhulaifi MM, Aldibasi O, Aljouie A, Alharbi O, Almohawes ZN, Basingab F, Rejili M. Gut microbiota composition in patients with Crohn's disease in Saudi Arabia. PLoS One 2024; 19:e0299749. [PMID: 38656971 PMCID: PMC11042705 DOI: 10.1371/journal.pone.0299749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/14/2024] [Indexed: 04/26/2024] Open
Abstract
Crohn's disease (CD) entails intricate interactions with gut microbiome diversity, richness, and composition. The relationship between CD and gut microbiome is not clearly understood and has not been previously characterized in Saudi Arabia. We performed statistical analysis about various factors influencing CD activity and microbiota dysbiosis, including diagnosis, treatment, and its impact on their quality of life as well as high-throughput metagenomic V3-V4 16S rRNA encoding gene hypervariable region of a total of eighty patients with CD, both in its active and inactive state with healthy controls. The results were correlated with the demographic and lifestyle information, which the participants provided via a questionnaire. α-diversity measures indicated lower bacterial diversity and richness in the active and inactive CD groups compared to the control group. Greater dysbiosis was observed in the active CD patients compared to the inactive form of the disease, showed by a reduction in microbial diversity. Specific pathogenic bacteria such as Filifactor, Peptoniphilus, and Sellimonas were identified as characteristic of CD groups. In contrast, anti-inflammatory bacteria like Defluviitalea, Papillibacter, and Petroclostridium were associated with the control group. Among the various factors influencing disease activity and microbiota dysbiosis, smoking emerged as the most significant, with reduced α-diversity and richness for the smokers in all groups, and proinflammatory Fusobacteria was more present (p<0.05). Opposite to the control group, microbial diversity and richness were lower in CD participants of older age compared to younger ones, and male CD participants showed less diversity compared to women participants from the same groups. Our results describe the first report on the relationship between microbiota and Crohn's disease progress in Saudi Arabia, which may provide a theoretical basis for the application of therapeutic methods to regulate gut microbes in CD.
Collapse
Affiliation(s)
- Hadil Alahdal
- Department of Biology, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ghaida Almuneef
- Department of Biology, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Manal Muhammed Alkhulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Omar Aldibasi
- Biostatistics Section, King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Abdulrahman Aljouie
- Artificial Intelligence and Bioinformatics Department, King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- Department of Health Informatics, College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Othman Alharbi
- Department of Medicine, College of Medicine, King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia
| | - Zakiah Naser Almohawes
- Department of Biology, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mokhtar Rejili
- Department of Life Sciences, College of Sciences, Al Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
31
|
Wu H, Ma W, Wang Y, Wang Y, Sun X, Zheng Q. Gut microbiome-metabolites axis: A friend or foe to colorectal cancer progression. Biomed Pharmacother 2024; 173:116410. [PMID: 38460373 DOI: 10.1016/j.biopha.2024.116410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
An expanding corpus of research robustly substantiates the complex interrelation between gut microbiota and the onset, progression, and metastasis of colorectal cancer. Investigations in both animal models and human subjects have consistently underscored the role of gut bacteria in a variety of metabolic activities, driven by dietary intake. These activities include amino acid metabolism, carbohydrate fermentation, and the generation and regulation of bile acids. These metabolic derivatives, in turn, have been identified as significant contributors to the progression of colorectal cancer. This thorough review meticulously explores the dynamic interaction between gut bacteria and metabolites derived from the breakdown of amino acids, fatty acid metabolism, and bile acid synthesis. Notably, bile acids have been recognized for their potential carcinogenic properties, which may expedite tumor development. Extensive research has revealed a reciprocal influence of gut microbiota on the intricate spectrum of colorectal cancer pathologies. Furthermore, strategies to modulate gut microbiota, such as dietary modifications or probiotic supplementation, may offer promising avenues for both the prevention and adjunctive treatment of colorectal cancer. Nevertheless, additional research is imperative to corroborate these findings and enhance our comprehension of the underlying mechanisms in colorectal cancer development.
Collapse
Affiliation(s)
- Hao Wu
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Wenmeng Ma
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yiyao Wang
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yuanyuan Wang
- Department of anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province, PR China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
32
|
Shang F, Jiang X, Wang H, Guo S, Kang S, Xu B, Wang X, Chen S, Li N, Liu B, Zhao Z. Bifidobacterium longum suppresses colorectal cancer through the modulation of intestinal microbes and immune function. Front Microbiol 2024; 15:1327464. [PMID: 38585690 PMCID: PMC10995357 DOI: 10.3389/fmicb.2024.1327464] [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: 10/25/2023] [Accepted: 02/12/2024] [Indexed: 04/09/2024] Open
Abstract
Colorectal cancer (CRC), one of the most common malignancies in the world, urgently requires more treatment strategies. Although there has been much research on probiotics, limited research has been done in treating cancer. The purpose of this study was to investigate the role of Bifidobacterium longum (B. longum) in the prevention and treatment of CRC. Through Cell Counting Kit-8 and Colony Formation Assays, 8 h and a B. longum count of 1 × 108 CFU/ml were chosen as the best cocultivation conditions with CRC cells. The role of B. longum in inhibiting the progression of CRC cells was verified by a series of functional and immunofluorescence assays. For instance, in vivo assays have verified that B. longum could alleviate CRC progression. In addition, according to the results of in vivo assays and clinical statistical analysis, B. longum could reduce diarrhea symptoms. Mechanistically, by 16S and RNA sequencing, it was found that B. longum could affect the development of CRC by regulating the composition of gut microbes and enhancing immune function. The B. longum might inhibit the occurrence and development of CRC and relieve diarrhea symptoms by regulating intestinal microbes and immune function.
Collapse
Affiliation(s)
- Fangjian Shang
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xia Jiang
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haobo Wang
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shang Guo
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuo Kang
- Medical Insurance Office, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bin Xu
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin Wang
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shihao Chen
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ning Li
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bo Liu
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zengren Zhao
- Department of General Surgery, Hebei Key Laboratory of Colorectal Cancer Precision Diagnosis and Treatment, The First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
33
|
Imanbayev N, Iztleuov Y, Bekmukhambetov Y, Abdelazim IA, Donayeva A, Amanzholkyzy A, Aigul Z, Aigerim I, Aslan Y. Colorectal cancer and microbiota: systematic review. PRZEGLAD GASTROENTEROLOGICZNY 2024; 16:380-396. [PMID: 39810864 PMCID: PMC11726231 DOI: 10.5114/pg.2024.136228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2025]
Abstract
Introduction The gut microbiome maintains the mucus membrane barrier's integrity, and it is modulated by the host's immune system. Aim To detect the effect of microbiota modulation using probiotics, prebiotics, symbiotics, and natural changes on colorectal cancers (CRCs). Methods A PubMed search was conducted to retrieve the original and in vivo articles published in English language from 2010 until 2021 containing the following keywords: 1) CRCs, 2) CRCs treatment (i.e. surgical, chemotherapy, radiotherapy and/or immunotherapy), and 3) microbiota probiotic(s), prebiotic(s), symbiotic(s), dysbiosis and/or nutritional treatment. A total of 198 PubMed records/articles were initially identified. 108 articles were excluded at the initial screening, and another 29 articles were excluded after reviewing the abstracts, and finally 61 studies were analysed for this systematic review. Results The gut microbiota metabolites and (SCFAs) short-chain fatty acids (i.e. acetate and butyrate) have a protective effect against CRCs. SCFAs reduce the inflammatory cytokines, inhibit colonocyte proliferation, and promote malignant cell apoptosis. Butyrate maintains the integrity of the mucus membrane barrier and reduces intestinal mucosal inflammation. Reduced butyric acid level and increased inflammatory cytokines were observed after reduced Bacteroides fragilis and Bacteroides vulgatus species in the colon. Akkermansia muciniphila bacterium decreased in patients with CRCs. Conclusions Prebiotics (i.e. inulin and resistant starch, SCFAs producers) and consumption of unprocessed plant products are useful for developing and maintaining healthy gut microbiota. The pro-, pre- and/or symbiotics may be useful when carefully selected for CRC patients, to restore beneficial gut microbiota and support treatment efficacy.
Collapse
Affiliation(s)
- Nauryzbay Imanbayev
- Department of Oncology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Yerbolat Iztleuov
- Department of Radiology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Yerbol Bekmukhambetov
- Association of Individual Entrepreneurs and Legal Entities, National Chamber of Health, Astana, Kazakhstan
| | - Ibrahim A. Abdelazim
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ainur Donayeva
- Department of Normal Physiology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Ainur Amanzholkyzy
- Department of Normal Physiology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Zhumasheva Aigul
- Department of Pathomorphology, Medical Centre of West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Imanbayeva Aigerim
- Department of Oncology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Yergaliyev Aslan
- Department of General Surgery, Medical Centre of West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| |
Collapse
|
34
|
Vázquez-Cuesta S, Lozano García N, Rodríguez-Fernández S, Fernández-Avila AI, Bermejo J, Fernández-Avilés F, Muñoz P, Bouza E, Reigadas E. Impact of the Mediterranean Diet on the Gut Microbiome of a Well-Defined Cohort of Healthy Individuals. Nutrients 2024; 16:793. [PMID: 38542704 PMCID: PMC10974552 DOI: 10.3390/nu16060793] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 01/04/2025] Open
Abstract
A comprehensive understanding of gut microbiota in a clearly defined group of healthy individuals is essential when making meaningful comparisons with various diseases. The Mediterranean diet (MD), renowned for its potential health benefits, and the influence of adherence thereto on gut microbiota have become a focus of research. Our aim was to elucidate the impact of adherence to the MD on gut microbiota composition in a well-defined cohort. In this prospective study, healthy volunteers completed a questionnaire to provide demographic data, medical history, and dietary intake. Adherence was evaluated using the Med-DQI. The V4 region of the 16S rRNA gene was sequenced. Analysis of sequencing data and statistical analysis were performed using MOTHUR software and R. The study included 60 patients (51.7% females). Adherence correlated with alpha diversity, and higher values were recorded in good adherers. Good adherers had a higher abundance of Paraprevotella and Bacteroides (p < 0.001). Alpha diversity correlated inversely with fat intake and positively with non-starch polysaccharides (NSPs). Evenness correlated inversely with red meat intake and positively with NSPs. Predicted functional analysis highlighted metabolic pathway differences based on adherence to the MD. In conclusion, our study adds useful information on the relationship between the MD and the gut microbiome.
Collapse
Affiliation(s)
- Silvia Vázquez-Cuesta
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Biochemistry and Molecular Biology Department, School of Biology, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - Nuria Lozano García
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
| | - Sara Rodríguez-Fernández
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
| | - Ana I. Fernández-Avila
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Javier Bermejo
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Francisco Fernández-Avilés
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES CB06/06/0058), 28029 Madrid, Spain
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES CB06/06/0058), 28029 Madrid, Spain
| | - Elena Reigadas
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (S.V.-C.); (N.L.G.); (S.R.-F.); (E.B.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.-A.); (J.B.); (F.F.-A.)
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| |
Collapse
|
35
|
Mishra Y, Ranjan A, Mishra V, Chattaraj A, Aljabali AAA, El-Tanani M, Hromić-Jahjefendić A, Uversky VN, Tambuwala MM. The role of the gut microbiome in gastrointestinal cancers. Cell Signal 2024; 115:111013. [PMID: 38113978 DOI: 10.1016/j.cellsig.2023.111013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
The gut microbiota present in the human digestive system is incredibly varied and is home to trillions of microorganisms. The gut microbiome is shaped at birth, while numerous genetic, dietary, and environmental variables primarily influence the microbiome composition. The importance of gut microbiota on host health is becoming more widely acknowledged. Digestion, intestinal permeability, and immunological and metabolism responses can all be affected by changes in the composition and function of the gut microbiota. There is mounting evidence that the microbial population's complex traits are important biomarkers and indicators of patient outcomes in cancer and its therapies. Numerous studies have demonstrated that changed commensal gut microorganisms contribute to the development and spread of cancer through various routes. Despite the ongoing controversy surrounding the gut microbiome and gastrointestinal cancer, accumulating evidence points to a potentially far more intricate connection than a simple cause-and-effect relationship. SIMPLE SUMMARY: Due to their high frequency and fatality rate, gastrointestinal cancers are regarded as a severe public health issue with complex medical and economic burdens. The gut microbiota may directly or indirectly interact with existing therapies like immunotherapy and chemotherapy, affecting how well a treatment works. The gut microbiome influences the immune response's activity, function, and development. Generally, certain gut bacteria impact the antitumor actions during cancer by creating particular metabolites or triggering T-cell responses. Yet, certain bacterial species have been found to promote cellular proliferation and metastasis in cancer, and comprehending these interactions in the context of cancer may help identify possible treatment targets. Notwithstanding the improvements in the field, additional research is still required to comprehend the underlying processes, examine the effects on existing therapies, and pinpoint certain bacteria and immune cells that can cause this interaction.
Collapse
Affiliation(s)
- Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Abhigyan Ranjan
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A A Aljabali
- Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Alkhama Medical and Health Sciences University, United Arab Emirates
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln LN6 7TS, England, United Kingdom.
| |
Collapse
|
36
|
Prakash A, Rubin N, Staley C, Onyeaghala G, Wen YF, Shaukat A, Milne G, Straka RJ, Church TR, Prizment A. Effect of ginger supplementation on the fecal microbiome in subjects with prior colorectal adenoma. Sci Rep 2024; 14:2988. [PMID: 38316805 PMCID: PMC10844320 DOI: 10.1038/s41598-024-52658-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Ginger has been associated with a decreased incidence of colorectal cancer (CRC) through reduction in inflammatory pathways and inhibition of tumor growth. Recent pre-clinical models have implicated changes in the gut microbiome as a possible mediator of the ginger effect on CRC. We hypothesized that, in adults previously diagnosed with a colorectal adenoma, ginger supplementation would alter the fecal microbiome in the direction consistent with its CRC-inhibitory effect. Sixty-eight adults were randomized to take either ginger or placebo daily for 6 weeks, with a 6-week washout and longitudinal stool collection throughout. We performed 16S rRNA sequencing and evaluated changes in overall microbial diversity and the relative abundances of pre-specified CRC-associated taxa using mixed-effects logistic regression. Ginger supplementation showed no significant effect on microbial community structure through alpha or beta diversity. Of 10 pre-specified CRC-associated taxa, there were significant decreases in the relative abundances of the genera Akkermansia (p < 0.001), Bacteroides (p = 0.018), and Ruminococcus (p = 0.013) after 6-week treatment with ginger compared to placebo. Ginger supplementation led to decreased abundances of Akkermansia and Bacteroides, which suggests that ginger may have an inhibitory effect on CRC-associated taxa. Overall, ginger supplementation appears to have a limited effect on gut microbiome in patients with colorectal adenomas.
Collapse
Affiliation(s)
- Ajay Prakash
- Division of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Nathan Rubin
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Christopher Staley
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Guillaume Onyeaghala
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Ya-Feng Wen
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | | | - Ginger Milne
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert J Straka
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Timothy R Church
- Division of Environmental Health Sciences, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Anna Prizment
- Division of Hematology, Oncology, and Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Environmental Health Sciences, University of Minnesota School of Public Health, Minneapolis, MN, USA
| |
Collapse
|
37
|
Schult D, Maurer HC, Frolova M, Ringelhan M, Mayr U, Ulrich J, Heilmaier M, Rasch S, Lahmer T, Reitmeier S, Hennig C, Gassner C, Thur N, Will T, Janssen KP, Steiger K, Jesinghaus M, Neuhaus K, Quante M, Haller D, Abdelhafez M, Schmid RM, Middelhoff M. Systematic Evaluation of Clinical, Nutritional, and Fecal Microbial Factors for Their Association With Colorectal Polyps. Clin Transl Gastroenterol 2024; 15:e00660. [PMID: 38088370 PMCID: PMC10887443 DOI: 10.14309/ctg.0000000000000660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/29/2023] [Indexed: 02/25/2024] Open
Abstract
INTRODUCTION The identification of risk factors for precursor lesions of colorectal cancer (CRC) holds great promise in the context of prevention. With this study, we aimed to identify patient characteristics associated with colorectal polyps (CPs) and polyp features of potential malignant progression. Furthermore, a potential association with gut microbiota in this context was investigated. METHODS In this single-center study, a total of 162 patients with CPs and 91 control patients were included. Multiple variables including information on lifestyle, diet, serum parameters, and gut microbiota, analyzed by 16S-rRNA gene amplicon sequencing and functional imputations (Picrust2), were related to different aspects of CPs. RESULTS We observed that elevated serum alkaline phosphatase (AP) levels were significantly associated with the presence of high-grade dysplastic polyps. This association was further seen for patients with CRC. Thereby, AP correlated with other parameters of liver function. We did not observe significant changes in the gut microbiota between patients with CP and their respective controls. However, a trend toward a lower alpha-diversity was seen in patients with CRC. Interestingly, AP was identified as a possible clinical effect modifier of stool sample beta diversity. DISCUSSION We show for the first time an increased AP in premalignant CP. Furthermore, AP showed a significant influence on the microbial composition of the intestine. Relatively elevated liver enzymes, especially AP, may contribute to the detection of precancerous dysplastic or neoplastic changes in colorectal lesions. The association between elevated AP, premalignant CP, and the microbiome merits further study.
Collapse
Affiliation(s)
- David Schult
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - H. Carlo Maurer
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Marina Frolova
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Marc Ringelhan
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ulrich Mayr
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jörg Ulrich
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Markus Heilmaier
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Sebastian Rasch
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Tobias Lahmer
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Sandra Reitmeier
- ZIEL—Institute for Food & Health, Technische Universität München, Freising, Germany
- Chair of Nutrition and Immunology, Technische Universität München, Freising, Germany
| | - Chiara Hennig
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christina Gassner
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Niklas Thur
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Theresa Will
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, Technische Universität München, Munich, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, Technische Universität München, Munich, Germany
- Institute of Pathology, University Hospital Marburg, Marburg, Germany
| | - Klaus Neuhaus
- ZIEL—Institute for Food & Health, Technische Universität München, Freising, Germany
| | - Michael Quante
- Department of Internal Medicine II, Universitätsklinikum Freiburg, Universität Freiburg, Freiburg, Germany
| | - Dirk Haller
- ZIEL—Institute for Food & Health, Technische Universität München, Freising, Germany
- Chair of Nutrition and Immunology, Technische Universität München, Freising, Germany
| | - Mohamed Abdelhafez
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Roland M. Schmid
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Moritz Middelhoff
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
38
|
Kwao-Zigah G, Bediako-Bowan A, Boateng PA, Aryee GK, Abbang SM, Atampugbire G, Quaye O, Tagoe EA. Microbiome Dysbiosis, Dietary Intake and Lifestyle-Associated Factors Involve in Epigenetic Modulations in Colorectal Cancer: A Narrative Review. Cancer Control 2024; 31:10732748241263650. [PMID: 38889965 PMCID: PMC11186396 DOI: 10.1177/10732748241263650] [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: 11/02/2023] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Background: Colorectal cancer is the second cause of cancer mortality and the third most commonly diagnosed cancer worldwide. Current data available implicate epigenetic modulations in colorectal cancer development. The health of the large bowel is impacted by gut microbiome dysbiosis, which may lead to colon and rectum cancers. The release of microbial metabolites and toxins by these microbiotas has been shown to activate epigenetic processes leading to colorectal cancer development. Increased consumption of a 'Westernized diet' and certain lifestyle factors such as excessive consumption of alcohol have been associated with colorectal cancer.Purpose: In this review, we seek to examine current knowledge on the involvement of gut microbiota, dietary factors, and alcohol consumption in colorectal cancer development through epigenetic modulations.Methods: A review of several published articles focusing on the mechanism of how changes in the gut microbiome, diet, and excessive alcohol consumption contribute to colorectal cancer development and the potential of using these factors as biomarkers for colorectal cancer diagnosis.Conclusions: This review presents scientific findings that provide a hopeful future for manipulating gut microbiome, diet, and alcohol consumption in colorectal cancer patients' management and care.
Collapse
Affiliation(s)
- Genevieve Kwao-Zigah
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Antionette Bediako-Bowan
- Department of Surgery, University of Ghana Medical School, Accra, Ghana
- Department of Surgery, Korle Bu Teaching Hospital, Accra, Ghana
| | - Pius Agyenim Boateng
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Gloria Kezia Aryee
- Department of Medical Laboratory Sciences, University of Ghana, Accra, Ghana
| | - Stacy Magdalene Abbang
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Gabriel Atampugbire
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Osbourne Quaye
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Emmanuel A. Tagoe
- Department of Medical Laboratory Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
39
|
Ruiz-Saavedra S, Arboleya S, Nogacka AM, González del Rey C, Suárez A, Diaz Y, Gueimonde M, Salazar N, González S, de los Reyes-Gavilán CG. Commensal Fecal Microbiota Profiles Associated with Initial Stages of Intestinal Mucosa Damage: A Pilot Study. Cancers (Basel) 2023; 16:104. [PMID: 38201530 PMCID: PMC10778549 DOI: 10.3390/cancers16010104] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Progressive intestinal mucosal damage occurs over years prior to colorectal cancer (CRC) development. The endoscopic screening of polyps and histopathological examination are used clinically to determine the risk and progression of mucosal lesions. We analyzed fecal microbiota compositions using 16S rRNA gene-based metataxonomic analyses and the levels of short-chain fatty acids (SCFAs) using gas chromatography in volunteers undergoing colonoscopy and histopathological analyses to determine the microbiota shifts occurring at the early stages of intestinal mucosa alterations. The results were compared between diagnosis groups (nonpathological controls and polyps), between samples from individuals with hyperplastic polyps or conventional adenomas, and between grades of dysplasia in conventional adenomas. Some microbial taxa from the Bacillota and Euryarchaeota phyla were the most affected when comparing the diagnosis and histopathological groups. Deeper microbiota alterations were found in the conventional adenomas than in the hyperplastic polyps. The Ruminococcus torques group was enriched in both the hyperplastic polyps and conventional adenomas, whereas the family Eggerthellaceae was enriched only in the hyperplastic polyps. The abundance of Prevotellaceae, Oscillospiraceae, Methanobacteriaceae, Streptococcaceae, Christensenellaceae, Erysipelotrichaceae, and Clostridiaceae shifted in conventional adenomas depending on the grade of dysplasia, without affecting the major SCFAs. Our results suggest a reorganization of microbial consortia involved in gut fermentative processes.
Collapse
Affiliation(s)
- Sergio Ruiz-Saavedra
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.R.-S.); (S.A.); (A.M.N.); (M.G.); (N.S.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.R.-S.); (S.A.); (A.M.N.); (M.G.); (N.S.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.R.-S.); (S.A.); (A.M.N.); (M.G.); (N.S.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Carmen González del Rey
- Department of Anatomical Pathology, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain;
| | - Adolfo Suárez
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
- Digestive Service, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain
| | - Ylenia Diaz
- Digestive Service, Carmen and Severo Ochoa Hospital, 33819 Cangas del Narcea, Spain;
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.R.-S.); (S.A.); (A.M.N.); (M.G.); (N.S.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.R.-S.); (S.A.); (A.M.N.); (M.G.); (N.S.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Sonia González
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.R.-S.); (S.A.); (A.M.N.); (M.G.); (N.S.)
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| |
Collapse
|
40
|
Crossland NA, Beck S, Tan WY, Lo M, Mason JB, Zhang C, Guo W, Crott JW. Fecal microbiota transplanted from old mice promotes more colonic inflammation, proliferation, and tumor formation in azoxymethane-treated A/J mice than microbiota originating from young mice. Gut Microbes 2023; 15:2288187. [PMID: 38031252 PMCID: PMC10730208 DOI: 10.1080/19490976.2023.2288187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023] Open
Abstract
Aging is a strong risk factor for colorectal cancer (CRC). It is well established that gut microbial dysbiosis can play a role in the etiology of CRC. Although the composition of the gut microbial community changes with age and is reported to become more pro-inflammatory, it is unclear whether such changes are also pro-tumorigenic for the colon. To address this gap, we conducted fecal microbiota transplants (FMT) from young (DY, ~6 wk) and old (DO, ~72 wk) donor mice into young (8 wk) recipient mice that were pre-treated with antibiotics. After initiating tumorigenesis with azoxymethane, recipients were maintained for 19 wk during which time they received monthly FMT boosters. Compared to recipients of young donors (RY), recipients of old donors (RO) had an approximately 3-fold higher prevalence of histologically confirmed colon tumors (15.8 vs 50%, Chi2 P = .03), approximately 2-fold higher proliferating colonocytes as well as significantly elevated colonic IL-6, IL-1β and Tnf-α. Transcriptomics analysis of the colonic mucosa revealed a striking upregulation of mitochondria-related genes in the RO mice, a finding corroborated by increased mitochondrial abundance. Amongst the differences in fecal microbiome observed between DY and DO mice, the genera Ruminoclostridium, Lachnoclostridium and Marvinbryantia were more abundant in DY mice while the genera Bacteroides and Akkermansia were more abundant in DO mice. Amongst recipients, Ruminoclostridium and Lachnoclostridium were higher in RY mice while Bacteroides was higher in RO mice. Differences in fecal microbiota were observed between young and old mice, some of which persisted upon transplant into recipient mice. Recipients of old donors displayed significantly higher colonic proliferation, inflammation and tumor abundance compared to recipients of young donors. These findings support an etiological role for altered gut microbial communities in the increased risk for CRC with increasing age and establishes that such risk can be transmitted between individuals.
Collapse
Affiliation(s)
- Nicholas A. Crossland
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, USA
- Comparative Pathology Laboratory, Boston University National Emerging Infectious Disease Laboratories, Boston, MA, USA
| | - Samuel Beck
- Department of Dermatology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Wei Yu Tan
- Comparative Pathology Laboratory, Boston University National Emerging Infectious Disease Laboratories, Boston, MA, USA
| | - Ming Lo
- Comparative Pathology Laboratory, Boston University National Emerging Infectious Disease Laboratories, Boston, MA, USA
| | - Joel B. Mason
- Vitamins and Carcinogenesis Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Chao Zhang
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Weimin Guo
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, USA
| | - Jimmy W. Crott
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, USA
- Vitamins and Carcinogenesis Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
41
|
Ruiz-Saavedra S, Zapico A, González S, Salazar N, de los Reyes-Gavilán CG. Role of the intestinal microbiota and diet in the onset and progression of colorectal and breast cancers and the interconnection between both types of tumours. MICROBIOME RESEARCH REPORTS 2023; 3:6. [PMID: 38455079 PMCID: PMC10917624 DOI: 10.20517/mrr.2023.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/12/2023] [Accepted: 11/21/2023] [Indexed: 03/09/2024]
Abstract
Colorectal cancer (CRC) is among the leading causes of mortality in adults of both sexes worldwide, while breast cancer (BC) is among the leading causes of death in women. In addition to age, gender, and genetic predisposition, environmental and lifestyle factors exert a strong influence. Global diet, including alcohol consumption, is one of the most important modifiable factors affecting the risk of CRC and BC. Western dietary patterns promoting high intakes of xenobiotics from food processing and ethanol have been associated with increased cancer risk, whereas the Mediterranean diet, generally leading to a higher intake of polyphenols and fibre, has been associated with a protective effect. Gut dysbiosis is a common feature in CRC, where the usual microbiota is progressively replaced by opportunistic pathogens and the gut metabolome is altered. The relationship between microbiota and BC has been less studied. The estrobolome is the collection of genes from intestinal bacteria that can metabolize oestrogens. In a dysbiosis condition, microbial deconjugating enzymes can reactivate conjugated-deactivated oestrogens, increasing the risk of BC. In contrast, intestinal microorganisms can increase the biological activity and bioavailability of dietary phytochemicals through diverse microbial metabolic transformations, potentiating their anticancer activity. Members of the intestinal microbiota can increase the toxicity of xenobiotics through metabolic transformations. However, most of the microorganisms involved in diet-microbiota interactions remain poorly characterized. Here, we provide an overview of the associations between microbiota and diet in BC and CRC, considering the diverse types and heterogeneity of these cancers and their relationship between them and with gut microbiota.
Collapse
Affiliation(s)
- Sergio Ruiz-Saavedra
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa 33300, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Aida Zapico
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
- Department of Functional Biology, University of Oviedo, Oviedo 33006, Spain
| | - Sonia González
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
- Department of Functional Biology, University of Oviedo, Oviedo 33006, Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa 33300, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa 33300, Spain
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo 33011, Spain
| |
Collapse
|
42
|
Ruiz-Saavedra S, González Del Rey C, Suárez A, Díaz Y, Zapico A, Arboleya S, Salazar N, Gueimonde M, de Los Reyes-Gavilán CG, González S. Associations of dietary factors and xenobiotic intake with faecal microbiota composition according to the presence of intestinal mucosa damage. Food Funct 2023; 14:9591-9605. [PMID: 37740374 DOI: 10.1039/d3fo01356a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
Diet is a major modulator of gut microbiota, which plays a key role in the health status, including colorectal cancer (CRC) development. Several studies and meta-analyses have evidenced an association of certain dietary factors and xenobiotic intake with the incidence of CRC. Nevertheless, how these dietary factors impact the first stages of intestinal mucosa damage is still uncertain. This study aimed at exploring the associations of relevant dietary factors with the gut microbiota of control individuals and subjects diagnosed with intestinal polyps. A total of 60 volunteers were recruited, clinically classified according to colonoscopy criteria and interviewed using food frequency questionnaires (FFQs). The nutritional status of each volunteer was determined and the intake of dietary xenobiotics was quantified. The relative abundance of faecal microbiota taxonomic groups was obtained through 16S rRNA gene sequencing. The association of dietary factors and xenobiotics with faecal microbiota composition showed differences according to the clinical diagnosis group. Our results showed that the intake of red meat (≥50 g day-1) and total polycyclic aromatic hydrocarbons (PAHs) (≥0.75 μg day-1) was associated with a decreased abundance of the family Bacteroidaceae and an increased abundance of Coriobacteriaceae in control subjects. The intake of the heterocyclic amines 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine (PhIP) (≥40 ng day-1) and 2-amino-3,8 dimethylimidazo(4,5,f) quinoxaline (MeIQx) (≥50 ng day-1) was associated with a decreased abundance of Akkermansiaceae in the control diagnosis group. Moreover, N-nitroso compounds (NOCs), nitrites (≥1.69 mg day-1) and N-nitrosodimethylamine (NDMA) (≥0.126 μg day-1) were associated with a decreased abundance of Bifidobacteriaceae. The intake of ethanol (≥12 g day-1) in the polyps group was associated with an increased abundance of Peptostreptococcaceae and a decreased abundance of Veillonellaceae. Moreover, linear regression analyses allowed us to identify ethanol, calcium, bioactive compounds such as flavonoids, stilbenes, cellulose, phenolic acids or total polyphenols, and dietary xenobiotics such as PhIP and MeIQx, the NOC N-nitrosopyrrolidine (NPYR) or the total PAHs as potential predictors of faecal microbiota group abundances. These results indicated that the consumption of milk, red meat, processed meat and ethanol and the intake of polyphenols, dietary PAHs, HAs and NOCs are associated with specific groups of the intestinal microbiota, depending on the clinical diagnosis group.
Collapse
Affiliation(s)
- Sergio Ruiz-Saavedra
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain.
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Carmen González Del Rey
- Anatomical Pathology Service, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain
| | - Adolfo Suárez
- Digestive Service, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Spain
| | - Ylenia Díaz
- Digestive Service, Carmen and Severo Ochoa Hospital, 33819 Cangas del Narcea, Spain
| | - Aida Zapico
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain.
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain.
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain.
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Clara G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain.
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Sonia González
- Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
43
|
Guan X, Liu N, Zhu Z, Xu Y, Xiong D, Li X. Association of tea and its extracts with colorectal adenomas: meta-analysis and systematic review. Front Nutr 2023; 10:1241848. [PMID: 37867491 PMCID: PMC10585173 DOI: 10.3389/fnut.2023.1241848] [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: 06/28/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Background There are many studies on the association of tea and its extracts with colorectal adenomas, but the results have varied. The study aims to investigate the effect of tea and its extracts on colorectal adenomas using meta analysis and systematic review. Methods Literature was obtained through PubMed, Cochrane Library, Embase and Chinese BioMedical Literature Service System since the establishment of the database until April 31, 2023. Search terms include adenomas, polyps, colorectal, rectal, rectum, tea, epigallocatechin, drinking and beverages. Meta-regression analysis was used to infer the source of heterogeneity. Heterogeneity was assessed using I2 statistics and Q test. The effect measures were odds ratio (OR) and 95% confidence interval (95% CI). Stata17.0 software was used for data processing. Results The findings indicated that study design (t = 0.78, P = 0.454), types of tea intake (t = 1.35, P = 0.205), occurrences (t = -0.19, P = 0.852), regions (t = 1.13, P = 0.281) and grades of adenomas (t = 0.06, P = 0.952) were statistical homogeneity. Tea and its extracts were negatively correlated with the risk of colorectal adenomas (OR = 0.81, 95% CI: 0.66-0.98). No publication bias was found in this study (t = -0.22, P = 0.828) and the results are robust. Conclusion This study suggests that tea and its extracts have a certain protective effect on colorectal adenomas, which provides scientific evidence for preventive strategies for colorectal adenomas. As for the causal relationship between tea and its extracts on colorectal adenomas, further prospective studies are needed.
Collapse
Affiliation(s)
- Xifei Guan
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nawen Liu
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhixin Zhu
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanxue Xu
- Department of Nursing, College of Medicine, Zhejiang University, Hangzhou, China
| | - Dehai Xiong
- Department of General Surgery, Three Gorges Affiliated Hospital, Chongqing University, Chongqing, China
| | - Xiuyang Li
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Widjaja F, Rietjens IMCM. From-Toilet-to-Freezer: A Review on Requirements for an Automatic Protocol to Collect and Store Human Fecal Samples for Research Purposes. Biomedicines 2023; 11:2658. [PMID: 37893032 PMCID: PMC10603957 DOI: 10.3390/biomedicines11102658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/24/2023] [Indexed: 10/29/2023] Open
Abstract
The composition, viability and metabolic functionality of intestinal microbiota play an important role in human health and disease. Studies on intestinal microbiota are often based on fecal samples, because these can be sampled in a non-invasive way, although procedures for sampling, processing and storage vary. This review presents factors to consider when developing an automated protocol for sampling, processing and storing fecal samples: donor inclusion criteria, urine-feces separation in smart toilets, homogenization, aliquoting, usage or type of buffer to dissolve and store fecal material, temperature and time for processing and storage and quality control. The lack of standardization and low-throughput of state-of-the-art fecal collection procedures promote a more automated protocol. Based on this review, an automated protocol is proposed. Fecal samples should be collected and immediately processed under anaerobic conditions at either room temperature (RT) for a maximum of 4 h or at 4 °C for no more than 24 h. Upon homogenization, preferably in the absence of added solvent to allow addition of a buffer of choice at a later stage, aliquots obtained should be stored at either -20 °C for up to a few months or -80 °C for a longer period-up to 2 years. Protocols for quality control should characterize microbial composition and viability as well as metabolic functionality.
Collapse
Affiliation(s)
- Frances Widjaja
- Division of Toxicology, Wageningen University & Research, 6708 WE Wageningen, The Netherlands;
| | | |
Collapse
|
45
|
Li X, Feng J, Wang Z, Liu G, Wang F. Features of combined gut bacteria and fungi from a Chinese cohort of colorectal cancer, colorectal adenoma, and post-operative patients. Front Microbiol 2023; 14:1236583. [PMID: 37614602 PMCID: PMC10443710 DOI: 10.3389/fmicb.2023.1236583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
Colorectal cancer (CRC) accounts for the third highest morbidity burden among malignant tumors worldwide. Previous studies investigated gut microbiome changes that occur during colorectal adenomas (CRA) progression to overt CRC, thus highlighting the importance of the gut microbiome in carcinogenesis. However, few studies have examined gut microbiome characteristics across the entire spectrum, from CRC development to treatment. The study used 16S ribosomal ribonucleic acid and internal transcribed spacer amplicon sequencing to compare the composition of gut bacteria and fungi in a Chinese cohort of healthy controls (HC), CRC patients, CRA patients, and CRC postoperative patients (PP). Our analysis showed that beta diversity was significantly different among the four groups based on the gut bacterial and fungal data. A total of 51 species of bacteria and 8 species of fungi were identified in the HC, CRA, CRC, and PP groups. Correlation networks for both the gut bacteria and fungi in HC vs. CRA, HC vs. CRC, and HC vs. PP indicated some hub bacterial and fungal genera in each model, and the correlation between bacterial and fungal data indicated that a highly significant negative correlation exists among groups. Quantitative polymerase chain reaction (qPCR) analysis in a large cohort of HC, CRC, CRA, and PP patients demonstrated a significantly increasing trend of Fusobacterium nucleatum, Bifidobacterium bifidum, Candida albicans, and Saccharomyces cerevisiae in the feces of CRC patients than that of HC patients (p < 0.01). However, the abundance levels of CRA and PP were significantly lower in HC patients than those in CRC patients. Further studies are required to identify the functional consequences of the altered bacterial/fungal composition on metabolism and CRC tumorigenesis in the host.
Collapse
Affiliation(s)
- Xiaopeng Li
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Radiation Oncology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Jiahui Feng
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Zhanggui Wang
- Department of Radiation Oncology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Gang Liu
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Fan Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
46
|
Qu R, Zhang Y, Ma Y, Zhou X, Sun L, Jiang C, Zhang Z, Fu W. Role of the Gut Microbiota and Its Metabolites in Tumorigenesis or Development of Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205563. [PMID: 37263983 PMCID: PMC10427379 DOI: 10.1002/advs.202205563] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/20/2023] [Indexed: 06/03/2023]
Abstract
Colorectal cancer (CRC) is the most common cancer of the digestive system with high mortality and morbidity rates. Gut microbiota is found in the intestines, especially the colorectum, and has structured crosstalk interactions with the host that affect several physiological processes. The gut microbiota include CRC-promoting bacterial species, such as Fusobacterium nucleatum, Escherichia coli, and Bacteroides fragilis, and CRC-protecting bacterial species, such as Clostridium butyricum, Streptococcus thermophilus, and Lacticaseibacillus paracasei, which along with other microorganisms, such as viruses and fungi, play critical roles in the development of CRC. Different bacterial features are identified in patients with early-onset CRC, combined with different patterns between fecal and intratumoral microbiota. The gut microbiota may be beneficial in the diagnosis and treatment of CRC; some bacteria may serve as biomarkers while others as regulators of chemotherapy and immunotherapy. Furthermore, metabolites produced by the gut microbiota play essential roles in the crosstalk with CRC cells. Harmful metabolites include some primary bile acids and short-chain fatty acids, whereas others, including ursodeoxycholic acid and butyrate, are beneficial and impede tumor development and progression. This review focuses on the gut microbiota and its metabolites, and their potential roles in the development, diagnosis, and treatment of CRC.
Collapse
Affiliation(s)
- Ruize Qu
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Yi Zhang
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Yanpeng Ma
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Xin Zhou
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Lulu Sun
- State Key Laboratory of Women's Reproductive Health and Fertility PromotionPeking UniversityBeijing100191P. R. China
- Department of Endocrinology and MetabolismPeking University Third HospitalBeijing100191P. R. China
| | - Changtao Jiang
- Center of Basic Medical ResearchInstitute of Medical Innovation and ResearchThird HospitalPeking UniversityBeijing100191P. R. China
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesPeking University and the Key Laboratory of Molecular Cardiovascular Science (Peking University)Ministry of EducationBeijing100191P. R. China
- Center for Obesity and Metabolic Disease ResearchSchool of Basic Medical SciencesPeking UniversityBeijing100191P. R. China
| | - Zhipeng Zhang
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| | - Wei Fu
- Department of General SurgeryPeking University Third HospitalBeijing100191P. R. China
- Cancer CenterPeking University Third HospitalBeijing100191P. R. China
| |
Collapse
|
47
|
Zhu Z, Guan X, Liu N, Zhu X, Dai S, Xiong D, Li X. Association between dietary factors and colorectal serrated polyps: a systematic review and meta-analysis. Front Nutr 2023; 10:1187539. [PMID: 37575321 PMCID: PMC10413578 DOI: 10.3389/fnut.2023.1187539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/27/2023] [Indexed: 08/15/2023] Open
Abstract
Background Dietary factors may affect the incidence of colorectal serrated polyps (SP). However, its effects on SP are unclear as epidemiological studies on this topic have showed inconsistent results. The present systematic review and meta-analysis sought to evaluate the effects of dietary factors on SPs. Methods Studies regarding the association between dietary factors and SPs were identified by searching PubMed, Cochrane library, Embase and Chinese Biomedical Literature database from inception until 27 February 2023. Search terms include serrated, hyperplastic, adenoma, polyps, colorectal, rectal, rectum and risk. Heterogeneity was assessed using I2 statistics. The meta-analysis was conducted by using a random-effects model, and the pooled effects were expressed with odds ratios (OR) and 95% confidence intervals (95% CI). Probable sources of heterogeneity were identified through meta-regression. Subgroup analysis were based on lesion types, study designs, countries, and so on. Results 28 studies were ultimately eligible after scanning, and five dietary factors including vitamin D, calcium, folate, fiber and red or processed meat were excerpted. Higher intakes of vitamin D (OR = 0.95, 95%CI:0.90-1.02), calcium (OR = 0.97, 95%CI: 0.91-1.03) and folate (OR = 0.82, 95% CI: 0.6-1.13) were not significantly associated with SP. Fiber intake (OR = 0.90, 95% CI: 0.82-0.99) was a protective factor against SPs. Red meat intake increased the risk of SPs by 30% for the highest versus lowest intakes (OR = 1.30, 95% CI: 1.13-1.51). For different lesion types, higher folate intake was associated with a decreased risk of HPs (OR = 0.59, 95%CI: 0.44-0.79), and higher vitamin D intake decreased the risk of SPs including SSA/P (OR = 0.93, 95%CI: 0.88-0.98). Conclusions Higher dietary fiber intake plays an effective role in preventing SP, while red meat intake is associated with an increased risk of SP. This evidence provides guidance for us to prevent SP from a dietary perspective. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?, RecordID=340750.
Collapse
Affiliation(s)
- Zhixin Zhu
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xifei Guan
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nawen Liu
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoxia Zhu
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sheng Dai
- Department of General Surgery, School of Medicine, Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dehai Xiong
- Department of General Surgery, Three Gorges Hospital, Chongqing University, Chongqing, China
| | - Xiuyang Li
- Department of Big Data in Health Science, and Center for Clinical Big Data and Statistics, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
48
|
Katsaounou K, Yiannakou D, Nikolaou E, Brown C, Vogazianos P, Aristodimou A, Chi J, Costeas P, Agapiou A, Frangou E, Tsiaoussis G, Potamitis G, Antoniades A, Shammas C, Apidianakis Y. Fecal Microbiota and Associated Volatile Organic Compounds Distinguishing No-Adenoma from High-Risk Colon Adenoma Adults. Metabolites 2023; 13:819. [PMID: 37512526 PMCID: PMC10383435 DOI: 10.3390/metabo13070819] [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: 05/22/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Microbiota and the metabolites they produce within the large intestine interact with the host epithelia under the influence of a range of host-derived metabolic, immune, and homeostatic factors. This complex host-microbe interaction affects intestinal tumorigenesis, but established microbial or metabolite profiles predicting colorectal cancer (CRC) risk are missing. Here, we aimed to identify fecal bacteria, volatile organic compounds (VOC), and their associations that distinguish healthy (non-adenoma, NA) from CRC prone (high-risk adenoma, HRA) individuals. Analyzing fecal samples obtained from 117 participants ≥15 days past routine colonoscopy, we highlight the higher abundance of Proteobacteria and Parabacteroides distasonis, and the lower abundance of Lachnospiraceae species, Roseburia faecis, Blautia luti, Fusicatenibacter saccharivorans, Eubacterium rectale, and Phascolarctobacterium faecium in the samples of HRA individuals. Volatolomic analysis of samples from 28 participants revealed a higher concentration of five compounds in the feces of HRA individuals, isobutyric acid, methyl butyrate, methyl propionate, 2-hexanone, and 2-pentanone. We used binomial logistic regression modeling, revealing 68 and 96 fecal bacteria-VOC associations at the family and genus level, respectively, that distinguish NA from HRA endpoints. For example, isobutyric acid associations with Lachnospiraceae incertae sedis and Bacteroides genera exhibit positive and negative regression lines for NA and HRA endpoints, respectively. However, the same chemical associates with Coprococcus and Colinsella genera exhibit the reverse regression line trends. Thus, fecal microbiota and VOC profiles and their associations in NA versus HRA individuals indicate the significance of multiple levels of analysis towards the identification of testable CRC risk biomarkers.
Collapse
Affiliation(s)
- Kyriaki Katsaounou
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus
| | | | | | | | | | | | | | | | - Agapios Agapiou
- Department of Chemistry, University of Cyprus, Nicosia 2109, Cyprus
| | | | | | | | | | | | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, Nicosia 2109, Cyprus
| |
Collapse
|
49
|
Li W, Zhou X, Yuan S, Wang L, Yu L, Sun J, Chen J, Xiao Q, Wan Z, Zheng JS, Zhang CX, Larsson SC, Farrington SM, Law P, Houlston RS, Tomlinson I, Ding KF, Dunlop MG, Theodoratou E, Li X. Exploring the Complex Relationship between Gut Microbiota and Risk of Colorectal Neoplasia Using Bidirectional Mendelian Randomization Analysis. Cancer Epidemiol Biomarkers Prev 2023; 32:809-817. [PMID: 37012201 PMCID: PMC10233354 DOI: 10.1158/1055-9965.epi-22-0724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/07/2022] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Human gut microbiome has complex relationships with the host, contributing to metabolism, immunity, and carcinogenesis. METHODS Summary-level data for gut microbiota and metabolites were obtained from MiBioGen, FINRISK and human metabolome consortia. Summary-level data for colorectal cancer were derived from a genome-wide association study meta-analysis. In forward Mendelian randomization (MR), we employed genetic instrumental variables (IV) for 24 gut microbiota taxa and six bacterial metabolites to examine their causal relationship with colorectal cancer. We also used a lenient threshold for nine apriori gut microbiota taxa as secondary analyses. In reverse MR, we explored association between genetic liability to colorectal neoplasia and abundance of microbiota studied above using 95, 19, and 7 IVs for colorectal cancer, adenoma, and polyps, respectively. RESULTS Forward MR did not find evidence indicating causal relationship between any of the gut microbiota taxa or six bacterial metabolites tested and colorectal cancer risk. However, reverse MR supported genetic liability to colorectal adenomas was causally related with increased abundance of two taxa: Gammaproteobacteria (β = 0.027, which represents a 0.027 increase in log-transformed relative abundance values of Gammaproteobacteria for per one-unit increase in log OR of adenoma risk; P = 7.06×10-8), Enterobacteriaceae (β = 0.023, P = 1.29×10-5). CONCLUSIONS We find genetic liability to colorectal neoplasia may be associated with abundance of certain microbiota taxa. It is more likely that subset of colorectal cancer genetic liability variants changes gut biology by influencing both gut microbiota and colorectal cancer risk. IMPACT This study highlights the need of future complementary studies to explore causal mechanisms linking both host genetic variation with gut microbiome and colorectal cancer susceptibility.
Collapse
Affiliation(s)
- Wanxin Li
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan Zhou
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuai Yuan
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lijuan Wang
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Yu
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Sun
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Chen
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qian Xiao
- Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Ju-Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Cai-Xia Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Susanna C. Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Susan M. Farrington
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Cancer Research UK Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Ian Tomlinson
- Cancer Research UK Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Ke-Feng Ding
- Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Malcolm G. Dunlop
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Cancer Research UK Edinburgh Cancer Research Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Evropi Theodoratou
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, Centre of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Colon Cancer Genetics Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| |
Collapse
|
50
|
Heng D, Zhang M, Yuan Y, Qiu X. Alteration of Colonic Bacterial and Fungal Composition and Their Inter- and Intra-Kingdom Interaction in Patients with Adenomas with Low-Grade Dysplasia. Microorganisms 2023; 11:1327. [PMID: 37317301 PMCID: PMC10223777 DOI: 10.3390/microorganisms11051327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
Colorectal cancer (CRC) develops from pre-cancerous cellular lesions in the gut epithelium and mainly originates from specific types of colonic adenomas with dysplasia. However, gut microbiota signatures among sampling sites in patients with colorectal adenomas with low-grade dysplasia (ALGD) and normal control (NC) remain uncharacterized. To characterize gut microbial and fungal profiles in ALGD and normal colorectal mucosa tissues. We used 16S and ITS1-2 rRNA gene sequencing and bioinformatics analysis on the microbiota of ALGD and normal colorectal mucosa from 40 subjects. Bacterial sequences in the ALGD group showed an increase in Rhodobacterales, Thermales, Thermaceae, Rhodobacteraceae, and several genera, including Thermus, Paracoccus, Sphingobium, and Pseudomonas, compared to the NC group. Fungal sequences in the ALGD group showed an increase in Helotiales, Leotiomycetes, and Basidiomycota, while several orders, families, and genera, including Verrucariales, Russulales, and Trichosporonales, were decreased. The study found various interactions between intestinal bacteria and fungi. The bacterial functional analysis showed increased glycogen and vanillin degradation pathways in the ALGD group. Meanwhile, the fungal functional analysis showed a decrease in pathways related to the biosynthesis of gondoate and stearate, as well as degradation of glucose, starch, glycogen, sucrose, L-tryptophan, and pantothenate, and an increase in the octane oxidation pathway in the ALGD group. The mucosal microbiota in ALGD exhibits altered fungal and microbial composition compared to the NC mucosa, potentially contributing to the development of intestinal cancer by regulating specific metabolic pathways. Therefore, these changes in microbiota and metabolic pathways may be potential markers for diagnosing and treating colorectal adenoma and carcinoma.
Collapse
Affiliation(s)
- Ding Heng
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China; (D.H.); (M.Z.)
| | - Min Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China; (D.H.); (M.Z.)
| | - Yuhan Yuan
- Department of Endoscopic Center, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China;
| | - Xinyun Qiu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing 210029, China; (D.H.); (M.Z.)
- F. Widjaja Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|