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
|
Jing Z, Yinhang W, Jian C, Zhanbo Q, Xinyue W, Shuwen H. Interaction between gut microbiota and T cell immunity in colorectal cancer. Autoimmun Rev 2025; 24:103807. [PMID: 40139455 DOI: 10.1016/j.autrev.2025.103807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 02/26/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
This review delves into the complex and multi-layered mechanisms that govern the interaction between gut microbiota and T cells in the context of colorectal cancer (CRC), revealing a novel "microbiota-immune regulatory landscape" within the tumor microenvironment. As CRC progresses, the gut microbiota experiences a significant transformation in both its composition and metabolic patterns. On one hand, specific microbial entities within the gut microbiota can directly engage with T cells, functioning as "immunological triggers" that shape T-cell behavior. Simultaneously, microbial metabolites, such as short-chain fatty acids and bile acids, serve as "molecular regulators" that intricately govern T-cell function and differentiation, fine-tuning the immune response. On the other hand, the quorum-sensing mechanism, a recently recognized communication network among bacteria, also plays a pivotal role in orchestrating T-cell immunity. Additionally, the gut microbiota forms an intriguing connection with the neuro-immune regulatory axis, a largely unexplored "territory" in CRC research. Regarding treatment strategies, a diverse array of intervention approaches-including dietary modifications, the utilization of probiotics, bacteriophages, and targeted antibiotic therapies-offer promising prospects for restoring the equilibrium of the gut microbiota, thereby acting as "ecosystem renovators" that impede tumor initiation and progression. Nevertheless, the current research landscape in this field is fraught with challenges. These include significant variations in microbial composition, dietary preferences, and tumor microenvironments among individuals, a lack of large-scale cohort studies, and insufficient research that integrates tumor mutation analysis, gut microbiota investigations, and immune microenvironment evaluations. This review emphasizes the necessity for future research efforts to seamlessly incorporate multiple factors and utilize bioinformatics analysis to construct a more comprehensive "interactive map" of the gut microbiota-T cell relationship in CRC. The aim is to establish a solid theoretical basis for the development of highly effective and personalized treatment regimens, ultimately transforming the therapeutic approach to CRC.
Collapse
Affiliation(s)
- Zhuang Jing
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Xinyue
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; ASIR (Institute - Association of intelligent systems and robotics), 14B rue Henri Sainte Claire Deville, 92500 Rueil-Malmaison, France.
| |
Collapse
|
3
|
San-Martin MI, Chamizo-Ampudia A, Sanchiz Á, Ferrero MÁ, Martínez-Blanco H, Rodríguez-Aparicio LB, Navasa N. Microbiome Markers in Gastrointestinal Disorders: Inflammatory Bowel Disease, Colorectal Cancer, and Celiac Disease. Int J Mol Sci 2025; 26:4818. [PMID: 40429958 DOI: 10.3390/ijms26104818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/06/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Intestinal microbiota and the host's immune system form a symbiotic alliance that sustains normal development and function in the human gut. Changes such as dietary habits among societies in developed countries have led to the development of unbalanced microbial populations in the gut, likely contributing to the dramatic increase in inflammatory diseases in the last few decades. Recent advances in DNA sequencing technologies have tremendously helped to characterize the microbiome associated with disease, both in identifying global alterations and discovering specific biomarkers that potentially contribute to disease pathogenesis, as evidenced by animal studies. Beyond bacterial alterations, non-bacterial components such as fungi, viruses, and microbial metabolites have been implicated in these diseases, influencing immune responses and gut homeostasis. Multi-omics approaches integrating metagenomics, metabolomics, and transcriptomics offer a more comprehensive understanding of the microbiome's role in disease pathogenesis, paving the way for innovative diagnostic and therapeutic strategies. Unraveling the metagenomic profiles associated with disease may facilitate earlier diagnosis and intervention, as well as the development of more personalized and effective therapeutic strategies. This review synthesizes recent and relevant microbiome research studies aimed at characterizing the microbial signatures associated with inflammatory bowel disease, colorectal cancer, and celiac disease.
Collapse
Affiliation(s)
- M Isabel San-Martin
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Alejandro Chamizo-Ampudia
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - África Sanchiz
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Miguel Ángel Ferrero
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Honorina Martínez-Blanco
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Leandro Benito Rodríguez-Aparicio
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| | - Nicolás Navasa
- Area Biochemistry, Department of Molecular Biology, University of León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, Campus de Vegazana, 24071 León, Spain
| |
Collapse
|
4
|
Hong Y, Wang D, Liu Z, Chen Y, Wang Y, Li J. Decoding per- and polyfluoroalkyl substances (PFAS) in hepatocellular carcinoma: a multi-omics and computational toxicology approach. J Transl Med 2025; 23:504. [PMID: 40317014 PMCID: PMC12049027 DOI: 10.1186/s12967-025-06517-z] [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: 02/25/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS), particularly perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS), are synthetic chemicals known for their widespread use and environmental persistence. These compounds have been increasingly linked to hepatotoxicity and the development of hepatocellular carcinoma (HCC). However, the molecular mechanisms by which PFAS contribute to HCC remain underexplored. METHODS This study employs a multi-omics approach that combines network toxicology, integrated machine learning, single-cell RNA sequencing, spatial transcriptomics, experimental validation, and molecular docking simulations to uncover the mechanisms through which PFAS exposure drives HCC. We analyzed publicly available transcriptomic data from several HCC cohorts and used differential gene expression analysis to identify targets associated with both PFAS exposure and HCC. We constructed a protein-protein interaction (PPI) network and a survival risk model, the PFAS-related HCC signature (PFASRHSig), based on integrated machine learning to identify prognostic biomarkers, with the goal of identifying core targets of PFAS in HCC progression and prognosis. RT-qPCR and immunohistochemical (IHC) staining were used to validate the expression levels of the targets in both tumor and normal tissues. Molecular docking simulations were conducted to assess the binding affinities between PFAS compounds and selected target proteins. RESULTS Functional enrichment studies revealed that PFAS targets were associated with metabolic signaling pathways, which are actively involved in lipid, glucose, drug metabolism, etc. Through integrated machine learning and PPI network analysis, we identified six genes, APOA1, ESR1, IGF1, PPARGC1A, SERPINE1, and PON1, that serve as core targets of PFAS in both HCC progression and prognosis. These targets were further validated via bulk RNA-seq, single-cell RNA-seq, and spatial transcriptomics, which revealed differential expression patterns across various cell types in the HCC tumor microenvironment. The results of RT-qPCR and IHC staining were consistent with the in silico findings. Molecular docking simulations revealed strong binding affinities between PFAS compounds and these core targets, supporting their potential roles in PFAS-induced hepatocarcinogenesis. CONCLUSIONS Our study highlights key molecular targets and pathways involved in PFAS-induced liver carcinogenesis and proposes a robust survival risk model (PFASRHSig) for HCC. These findings provide new insights into PFAS toxicity mechanisms and offer potential therapeutic targets for mitigating the health risks associated with PFAS exposure. Collectively, our findings help in advancing clinical applications by providing insights into disease mechanisms and potential therapeutic interventions.
Collapse
Affiliation(s)
- Yanggang Hong
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Deqi Wang
- The First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zeyu Liu
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yuxin Chen
- The First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi Wang
- The First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jiajun Li
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
5
|
Costa MADC, da Silva Duarte V, Fraiz GM, Cardoso RR, da Silva A, Martino HSD, Dos Santos D'Almeida CT, Ferreira MSL, Corich V, Hamaker BR, Giacomini A, Bressan J, Barros FARD. Regular Consumption of Black Tea Kombucha Modulates the Gut Microbiota in Individuals with and without Obesity. J Nutr 2025; 155:1331-1349. [PMID: 39732435 DOI: 10.1016/j.tjnut.2024.12.013] [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: 08/26/2024] [Revised: 11/22/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Kombucha, a fermented beverage obtained from a Symbiotic Culture of Bacteria and Yeast, has shown potential in modulating gut microbiota, although no clinical trials have been done. OBJECTIVES We aimed to evaluate the effects of regular black tea kombucha consumption on intestinal health in individuals with and without obesity. METHODS A pre-post clinical intervention study was conducted lasting 8 wk. Forty-six participants were allocated into 2 groups: normal weight + black tea kombucha (n = 23); and obese + black tea kombucha (n = 23). Blood, urine, and stool samples were collected at baseline (T0) and after 8 wk of intervention (T8). RESULTS A total of 145 phenolic compounds were identified in the kombucha, primarily flavonoids (81%) and phenolic acids (19%). Kombucha favored commensal bacteria such as Bacteroidota and Akkermanciaceae, especially in the obese group. Subdoligranulum, a butyrate producer, also increased in the obese group after kombucha consumption (P = 0.031). Obesity-associated genera Ruminococcus and Dorea were elevated in the obese group at baseline (P < 0.05) and reduced after kombucha consumption, becoming similar to the normal weight group (Ruminococcus: obese T8 × normal weight T8: P = 0.27; Dorea: obese T8 × normal weight T0: P = 0.57; obese T8 × normal weight T8: P = 0.32). Fungal diversity increased, with a greater abundance of Saccharomyces in both groups and reductions in Exophiala and Rhodotorula, particularly in the obese group. Pichia and Dekkera, key microorganisms in kombucha, were identified as biomarkers after the intervention. CONCLUSIONS Regular kombucha consumption positively influenced gut microbiota in both normal and obese groups, with more pronounced effects in the obese group, suggesting that it may be especially beneficial for those individuals. This trial was registered at Brazilian Clinical Trial Registry - ReBEC as UTN code U1111-1263-9550 (https://ensaiosclinicos.gov.br/rg/RBR-9832wsx).
Collapse
Affiliation(s)
- Mirian Aparecida de Campos Costa
- Bioactive Compounds and Carbohydrates (BIOCARB) Research Group, Department of Food Science and Technology, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Vinícius da Silva Duarte
- Faculty of Chemistry, Biotechnology, and Food Science, The Norwegian University of Life Sciences, Ås, Norway
| | - Gabriela Macedo Fraiz
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil; Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, Universidad de Navarra, Pamplona, Spain
| | - Rodrigo Rezende Cardoso
- Bioactive Compounds and Carbohydrates (BIOCARB) Research Group, Department of Food Science and Technology, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Alessandra da Silva
- Public Health Epidemiology Graduate Program, Environmental and Health Education Laboratory, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | | | - Carolina Thomaz Dos Santos D'Almeida
- Laboratory of Bioactives, Food and Nutrition Graduate Program, Federal University of State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
| | - Mariana Simões Larraz Ferreira
- Laboratory of Bioactives, Food and Nutrition Graduate Program, Federal University of State of Rio de Janeiro - UNIRIO, Rio de Janeiro, RJ, Brazil
| | - Viviana Corich
- Department of Agronomy, Food Natural Resources, Animals, and Environment, Università degli Studi di Padova, Legnaro, Padova, PD, Italy
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, United States
| | - Alessio Giacomini
- Department of Agronomy, Food Natural Resources, Animals, and Environment, Università degli Studi di Padova, Legnaro, Padova, PD, Italy
| | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Frederico Augusto Ribeiro de Barros
- Bioactive Compounds and Carbohydrates (BIOCARB) Research Group, Department of Food Science and Technology, Universidade Federal de Viçosa, Viçosa, MG, Brazil.
| |
Collapse
|
6
|
Torshizi Esfahani A, Zafarjafarzadeh N, Vakili F, Bizhanpour A, Mashaollahi A, Karimi Kordestani B, Baratinamin M, Mohammadpour S. Gut microbiome in colorectal cancer: metagenomics from bench to bedside. JNCI Cancer Spectr 2025; 9:pkaf026. [PMID: 40045177 DOI: 10.1093/jncics/pkaf026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/27/2024] [Accepted: 02/27/2025] [Indexed: 05/27/2025] Open
Abstract
Colorectal cancer (CRC) is a major global health challenge. Emerging research highlights the pivotal role of the gut microbiota in influencing CRC risk, progression, and treatment response. Metagenomic approaches, especially high-throughput shotgun sequencing, have provided unprecedented insights into the intricate connections between the gut microbiome and CRC. By enabling comprehensive taxonomic and functional profiling, metagenomics has revealed microbial signatures, activities, and biomarkers associated with colorectal tumorigenesis. Furthermore, metagenomics has shown a potential to guide patient stratification, predict treatment outcomes, and inform microbiome-targeted interventions. Despite remaining challenges in multi-omics data integration, taxonomic gaps, and validation across diverse cohorts, metagenomics has propelled our comprehension of the intricate gut microbiome-CRC interplay. This review underscores the clinical relevance of microbial signatures as potential diagnostic and prognostic tools in CRC. Furthermore, it discusses personalized treatment strategies guided by this omics' approach.
Collapse
Affiliation(s)
- Amir Torshizi Esfahani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikta Zafarjafarzadeh
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Fatemeh Vakili
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Anahita Bizhanpour
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Amirhesam Mashaollahi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Bita Karimi Kordestani
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Mahdieh Baratinamin
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University Tehran, Tehran, Iran
| | - Somayeh Mohammadpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Guo H, Wang J, Huang S, Sooranna SR, Shu F, Li G. Long-Term Exposure to Microplastics Promotes Early-Stage Hepatocarcinogenesis Induced by Diethylnitrosamine in Rats by Modulation of Their Gut Microbiota. TOXICS 2025; 13:353. [PMID: 40423432 DOI: 10.3390/toxics13050353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/23/2025] [Accepted: 04/26/2025] [Indexed: 05/28/2025]
Abstract
Hepatocarcinogenesis is linked to environmental factors, with microplastics (MPs) emerging as a global environmental concern that may contribute to liver injury. However, the impact of MPs on the early stages of hepatocarcinogenesis has been largely ignored. Here we investigated the impact of long-term MP exposure on the formation of preneoplastic lesions during hepatocarcinogenesis induced by diethylnitrosamine (DEN) in rats. Rats were injected with DEN to induce preneoplastic lesions, and then they were orally administered with 1 µm MPs 0.5 mg/kg body weight per day for 20 weeks. The results revealed that long-term exposure to MPs did not induce the formation of glutathione S-transferase placental form (GST-P)-positive foci as preneoplastic lesions during hepatocarcinogenesis in these animals, thereby indicating non-carcinogenicity. However, MP exposure resulted in a 1-fold increase in both the number and size of GST-P-positive foci in rats initiated with DEN compared to those treated with DEN alone. Accordingly, MP exposure led to a 0.61-fold increase in the index of proliferating cell nuclear antigen (PCNA)-positive cells in DEN-initiated rats when compared to DEN treatment alone. In addition, the composition of the gut microbiota was significantly altered, accompanied by various levels of short-chain fatty acids. Our results suggest that long-term MP exposure can promote pre-neoplastic lesion formation in DEN-induced rats by increased cell proliferation as well as alterations in the gut microbiota and short-chain fatty acid levels. This highlights the potential health risks associated with hepatocarcinogenesis linked to long-term exposure to MPs.
Collapse
Affiliation(s)
- Huina Guo
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Jianan Wang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Shaowen Huang
- Department of Laboratory Medicine, Baise People's Hospital, Baise 533000, China
| | - Suren Rao Sooranna
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London SW10 9NH, UK
| | - Fangyi Shu
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Genliang Li
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise 533000, China
| |
Collapse
|
8
|
Zhou K, Yu Y, Li W, Zhu M. Clostridium butyricum Regulates the Inflammatory and Immunoregulatory Pathway Through NFKB1 in Colorectal Cancer Treatment. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10547-w. [PMID: 40279041 DOI: 10.1007/s12602-025-10547-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Colorectal cancer (CRC) ranks among the top three most prevalent malignancies globally and is a leading cause of cancer-related mortality. Traditional therapeutic approaches usually cause significant adverse effects, highlighting the urgent demand for alternative, more effective treatments. Probiotics have gained attentions as potential cancer therapy due to their beneficial impacts on host health. Clostridium butyricum (Cl. butyricum) has shown anticancer properties in recent studies, though the underlying mechanisms remain inadequately understood. This study presents an integrative analysis of network pharmacology and proteomics to elucidate the key targets of Cl. butyricum in CRC treatment. The network pharmacology analysis identified 72 overlapping genes, and functional analysis of these genes indicated that most pathways were related to pathways in cancer and inflammation, and butyrate emerging as the pivotal product of Cl. butyricum due to its strong associations with the identified hub genes. In parallel, proteomics analysis revealed 168 differential expressed proteins (DEPs) in Cl. butyricum-treated HCT-116 cells, comprising 78 upregulated and 90 downregulated proteins. These DEPs were primarily enriched in apoptosis and inflammatory pathways. PPI analysis further highlighted NFKB1 as key contributors to the anticancer effects of Cl. butyricum. The integrative analysis revealed a significant convergence of pathways enrichment patterns, particularly in inflammatory and immune-related pathways. Computational and experimental validation identified NFKB1 as a pivotal molecular target in CRC intervention. These collective findings elucidate the mechanistic basis of the antitumor properties of Cl. butyricum, highlighting its regulatory effects on NFKB1 through both inflammatory and, to a lesser extent, immunoregulatory pathways.
Collapse
Affiliation(s)
- Kun Zhou
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China
| | - Yue Yu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China
| | - Wei Li
- College of Medicine, Translational Medicine Research Institute, Yangzhou University, Yangzhou, 225001, China
| | - Mingchang Zhu
- The Key Laboratory of the Inorganic Molecule-Based Chemistry of Liaoning Province, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China.
- College of Environmental and Safety Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, Liaoning, China.
| |
Collapse
|
9
|
Abdel Hamid M, Pammer LM, Oberparleiter S, Günther M, Amann A, Gruber RA, Mair A, Nocera FI, Ormanns S, Zimmer K, Gerner RR, Kocher F, Vorbach SM, Wolf D, Riedl JM, Huemer F, Seeber A. Multidimensional differences of right- and left-sided colorectal cancer and their impact on targeted therapies. NPJ Precis Oncol 2025; 9:116. [PMID: 40263545 PMCID: PMC12015310 DOI: 10.1038/s41698-025-00892-y] [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: 12/05/2024] [Accepted: 03/31/2025] [Indexed: 04/24/2025] Open
Abstract
Despite advances in metastatic colorectal cancer (mCRC) treatment, long-term survival remains poor, particularly in right-sided colorectal cancer (RCRC), which has a worse prognosis compared to left-sided CRC (LCRC). This disparity is driven by the complex biological diversity of these malignancies. RCRC and LCRC differ not only in clinical presentation and outcomes but also in their underlying molecular and genetic profiles. This article offers a detailed literature review focusing on the distinctions between RCRC and LCRC. We explore key differences across embryology, anatomy, pathology, omics, and the tumor microenvironment (TME), providing insights into how these factors contribute to prognosis and therapeutic responses. Furthermore, we examine the therapeutic implications of these differences, considering whether the conventional classification of CRC into right- and left-sided forms should be refined. Recent molecular findings suggest that this binary classification may overlook critical biological complexities. Therefore, we propose that future approaches should integrate molecular insights to better guide personalized treatments, especially anti-EGFR therapies, and improve patient outcomes.
Collapse
Affiliation(s)
- Marwa Abdel Hamid
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Lorenz M Pammer
- Department of Gastroenterology and Hepatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Silvia Oberparleiter
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Günther
- INNPATH, Institute of Pathology, Tirol Kliniken GmBH, Innsbruck, Austria
| | - Arno Amann
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Rebecca A Gruber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Mair
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Fabienne I Nocera
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Steffen Ormanns
- INNPATH, Institute of Pathology, Tirol Kliniken GmBH, Innsbruck, Austria
| | - Kai Zimmer
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Romana R Gerner
- Department of Medicine III, Hematology and Oncology, University Hospital Rechts der Isar, Technical University of Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, ZIEL Institute for Food & Health, 85354, Freising, Germany
| | - Florian Kocher
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Samuel M Vorbach
- Department of Radiation Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob M Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Huemer
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Center for Clinical Cancer and Immunology Trials (CCCIT), Paracelsus Medical University, Salzburg, Austria
| | - Andreas Seeber
- Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria.
- Department of Oncology, Hematology and Palliative Care, General Hospital Oberwart, Oberwart, Austria.
| |
Collapse
|
10
|
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
|
11
|
Zhang H, Zhou Y, Jiang YH, Hu WP, Huang LL, Lin HX, Zuo ZG, Du JM, Lou YL. Altered microbiota of rectal mucosa in rectal cancer patients. World J Gastroenterol 2025; 31:105248. [PMID: 40248061 PMCID: PMC12001164 DOI: 10.3748/wjg.v31.i13.105248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND With advances in sequencing techniques, microbiota dysbiosis and pathogenic microbes that accelerate colorectal cancer progression have been identified and widely reported. However, few studies have focused on the microbiota taxa of rectal mucus in rectal cancer (RC) patients. Here, we analyzed the composition and characteristics of the rectal mucosa microbiota of RC patients from Wenzhou city, China, and compared the results with those of healthy controls. AIM To explore the changes in the characteristics of the rectal mucosal flora associated with RC, and identify biomarkers of microbe taxa for RC. METHODS Rectal mucosa samples from a Chinese cohort of 72 recently diagnosed RC patients and 71 healthy controls were obtained. A validation cohort, which included 22 RC patients and 60 healthy controls, was also established. Changes in the rectal mucosal flora were observed by cultivation, 16S ribosomal DNA gene sequencing analysis and quantitative polymerase chain reaction analysis. RESULTS The 16S ribosomal DNA results demonstrated that RC patients presented increased bacterial community richness and alpha diversity as well as an altered rectal mucosal microbiota, with depletion of Proteobacteria and Thermi and enrichment of Bacteroidetes and Fusobacteria in cancerous mucosal tissues (CM) and enrichment of Firmicutes and Cyanobacteria in adjacent noncancerous mucosal tissues (AM). The culture results showed that the mean loads of Escherichia coli, Bifidobacterium, Enterococcus, and Lactobacillus were significantly reduced in RC patients. The ratios of Prevotella to Ruminococcus [areas under the receiver operating curve: 0.795 in AM vs normal control mucosa (NM), 0.77 in CM vs NM] and of Prevotella stercorea to Propionibacterium acnes (areas under the receiver operating curve: 0.808 in AM vs NM, 0.843 in CM vs NM) exhibited excellent abilities to differentiate between healthy controls and RC patients. CONCLUSION RC patients have an altered rectal mucosal microbiota, and the ratio of Prevotella to Ruminococcus or the ratio of Prevotella stercorea to Propionibacterium acnes may serve as a marker for RC diagnosis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Laboratory Medicine, Hangzhou Geriatric Hospital, Hangzhou 310022, Zhejiang Province, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yan Zhou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - You-Heng Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, Guangdong Province, China
| | - Wan-Ping Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Lu-Lu Huang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Hai-Xia Lin
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Zhi-Gui Zuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Ji-Mei Du
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yong-Liang Lou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| |
Collapse
|
12
|
McDonald HG, Reagan AM, Faisal ASM, Goettl R, Wang C, Hoyd R, Spakowicz D, Evers BM, Kim J, Bhakta AS. Survival Disparity and the Unique Genomic and Microbiome Profiles of Colon Cancer in Appalachian Kentucky. J Am Coll Surg 2025; 240:612-624. [PMID: 39868697 DOI: 10.1097/xcs.0000000000001299] [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: 01/28/2025]
Abstract
BACKGROUND Colon cancer is a leading cause of mortality in Appalachian Kentucky. Studies suggest that the microbiome may influence cancer outcomes. We investigate differential gene expression, the tumor microbiome, and their association as potential drivers of disparities in colon cancer outcomes. STUDY DESIGN This study analyzed patients diagnosed with colon adenocarcinoma between 2010 and 2023. Demographic data were extracted from Kentucky Cancer Registry. Somatic mutations and significantly mutated genes were identified using Fisher's exact t -test. RNASeq data were processed for gene expression analysis and Holm-Bonferroni method was used to adjust p values for multiple comparisons. The STAR aligner (exotic), v2.1 pipeline, and KrakenUniq database were used to classify microbes in human samples. The R package (exotic) was then used to decontaminate the results. RESULTS The final cohort included 2,276 patients, 321 of which had available somatic mutation sequencing data. Demographic differences between Appalachian and non-Appalachian patients included marital status (p = 0.0005), race (p < 0.0001), insurance status (p = 0.0005), BMI (p = 0.001), type 2 diabetes (p < 0.0001), and Charlson Comorbidity Index (p = 0.03). There was no difference in gene mutation frequency. There was differential expression of 228 genes. Differential abundance analysis revealed differences in 381 bacterial species. Importantly, 3 microbiota significantly correlated with survival disparities between Appalachian and non-Appalachian patients: Clostridium cadaveris (adjusted p = 0.009), Ligilactobacillus salivarius (adjusted p = 0.048), and Sutterella wadsworthensis (adjusted p = 0.009). CONCLUSIONS This is the first report of the distinct tumor microbiome in Appalachian Kentucky and its impact on survival. Further studies are needed to better characterize the unique tumor and gut microbiome of Appalachian patients with colon cancer.
Collapse
Affiliation(s)
- Hannah G McDonald
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Anna M Reagan
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Abu Saleh Mosa Faisal
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Ryan Goettl
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Chi Wang
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Rebecca Hoyd
- The Ohio State University Comprehensive Cancer Center, Division of Medical Oncology, Columbus, OH (Hoyd, Spakowicz)
| | - Daniel Spakowicz
- The Ohio State University Comprehensive Cancer Center, Division of Medical Oncology, Columbus, OH (Hoyd, Spakowicz)
| | - B Mark Evers
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Joseph Kim
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| | - Avinash S Bhakta
- From the Department of Surgery (McDonald, Reagan, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
- Markey Cancer Center (McDonald, Reagan, Faisal, Goettl, Wang, Evers, Kim, Bhakta), University of Kentucky Medical Center, Lexington, KY
| |
Collapse
|
13
|
Chan YL, Liao JC, Li TL, Wu CJ, Chiu YH. Bifidobacterium lactis ameliorates AOM/DSS-induced inflammation, dysbiosis, and colonic precancerous lesions. Appl Microbiol Biotechnol 2025; 109:69. [PMID: 40116950 PMCID: PMC11928396 DOI: 10.1007/s00253-025-13445-x] [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: 03/13/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/23/2025]
Abstract
Bowel cancer is the third most common malignancy of tumors and one of the major causes of cancer-related death. Bowel precancerous conditions can develop without any symptoms, which either makes it difficult for early diagnosis or poses a poor prognosis/gloomy relapse. This study aimed to investigate the effects of Bifidobacterium animalis subsp. lactis TCI604 (B. lactis) on inflammatory responses, gut microbiome, and protectiveness against azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colonic precancerous lesions. The AOM/DSS-induced colonic precancerous lesion murine model was studied with 24 female C57BL/6 J mice assigned to the control group, AOM/DSS-induced colonic precancerous lesion group (AOM/DSS), AOM/DSS treated with B. lactis probiotic group (B. lactis P), and AOM/DSS treated with B. lactis cell-free supernatant group (B. lactis S). The results showed that both B. lactis P and B. lactis S could attenuate AOM/DSS-induced body weight loss and intestine damage, reduce aberrant crypt foci (ACF) and the formation of colonic polyps, and significantly inhibit pro-inflammatory cytokines and the NF-κB signaling pathway, in which the B. lactis S group outperformed others. Further analysis using 16S rDNA sequencing suggested that both B. lactis P and B. lactis S optimize gut microbiota. Several bacteria, including Muribaculaceae, Prevotellaceae_UCG-001, Anaerostipes, Ruminococcaceae, Mucispirillum, Clostridia_UCG-014, and Clostridia_vadinBB60 that were known in close relation to colonic precancerous lesions, were sequenced at taxonomic level. Our results indicated that both B. lactis P and B. lactis S improved AOM/DSS-induced colonic precancerous lesions by regulating inflammation as well as optimizing gut microbiota, thereby establishing reciprocally cooperative net benefits between probiotics/postbiotics and mice with colonic precancerous lesions. KEY POINTS: • Prophylactic administration of probiotic and postbiotic of B. lactis is capable of alleviating the AOM/DSS-induced body weight loss and colon shortening, as well as diminishing the development of colonic precancerous lesions, such as the formation of ACF and colonic polyps, in an AOM/DSS mouse model • Either probiotic or postbiotic of B. lactis has a positive role in mediating immune imbalance and colonic inflammation via suppression of inflammatory immune cells, pro-inflammatory cytokines, and the NF-κB signaling pathway • AOM/DSS-induced dysbiosis can be reversed with the probiotic and postbiotic of B. lactis supplementation.
Collapse
Affiliation(s)
- Yi-Lin Chan
- Department of Chemistry, Chinese Culture University, Taipei, Taiwan, ROC
| | - Jun-Cheng Liao
- Department of Microbiology, Soochow University, 70, Linhsi Road, Taipei, Taiwan, ROC
| | - Tsung-Lin Li
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Chang-Jer Wu
- Department of Food Science and Center of Excellence for the Oceans, National Taiwan Ocean University, 2, Pei Ning Road, Keelung, Taiwan, ROC.
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC.
| | - Yi-Han Chiu
- Department of Microbiology, Soochow University, 70, Linhsi Road, Taipei, Taiwan, ROC.
| |
Collapse
|
14
|
Qin Y, Wang Q, Lin Q, Liu F, Pan X, Wei C, Chen J, Huang T, Fang M, Yang W, Pan L. Multi-omics analysis reveals associations between gut microbiota and host transcriptome in colon cancer patients. mSystems 2025; 10:e0080524. [PMID: 40013792 PMCID: PMC11915798 DOI: 10.1128/msystems.00805-24] [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: 06/13/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Colon cancer (CC) is one of the most common cancers globally, which is associated with the gut microbiota intimately. In current research, exploring the complex interaction between microbiomes and CC is a hotspot. However, the information on microbiomes in most previous studies is based on fecal, which does not fully display the microbial environment of CC. Herein, we collected mucosal and tissue samples from both the tumor and normal regions of 19 CC patients and clarified the composition of mucosal microbiota by 16S rRNA and metagenomic sequencing. Additionally, RNA-Seq was also conducted to identify the different expression genes between tumor and normal tissue samples. We revealed significantly different microbial community structures and expression profiles to CC. Depending on correlation analysis, we demonstrated that 1,472 genes were significantly correlated with CC tumor microbiota. Our study reveals a significant enrichment of Campylobacter jejuni in the mucosa of CC, which correlates with bile secretion. Additionally, we observe a negative correlation between C. jejuni and immune cells CD4+ Tem and mast cells. Finally, we discovered that metabolic bacterial endosymbiont of Bathymodiolus sp., Bacillus wiedmannii, and Mycobacterium tuberculosis had a significant survival value for CC, which was ignored by previous research. Overall, our study expands the understanding of the complex interplay between microbiota and CC and provides new targets for the treatment of CC. IMPORTANCE This study contributes to our understanding of the interaction between microbiota and colon cancer (CC). By examining mucosal and tissue samples rather than solely relying on fecal samples, we have uncovered previously unknown aspects of CC-associated microbiota. Our findings reveal distinct microbial community structures and gene expression profiles correlated with CC progression. Notably, the enrichment of Campylobacter jejuni in CC mucosa, linked to bile secretion, underscores potential mechanisms in CC pathogenesis. Additionally, observed correlations between microbial taxa and immune cell populations offer new avenues for immunotherapy research in CC. Importantly, this study introduces CC-associated microbiota with survival implications for CC, expanding therapeutic targets beyond conventional strategies. By elucidating these correlations, our study not only contributes to uncovering the potential role of gut microbiota in colon cancer but also establishes a foundation for mechanistic studies of gut microbiota in colon cancer, emphasizing the broader impact of microbiota research on cancer biology.
Collapse
Affiliation(s)
- Yuling Qin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiang Wang
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fengfei Liu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaolan Pan
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junxian Chen
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Taijun Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Fang
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Weilong Yang
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Linghui Pan
- Guangxi Clinical Research Center for Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
15
|
Liu J, Yuan Q, Zhang Y, Wang X, Zhai L, Wang R, Zheng C, Hong Z. Sleep health: an unappreciated key player in colorectal cancer. J Cancer 2025; 16:1934-1943. [PMID: 40092705 PMCID: PMC11905398 DOI: 10.7150/jca.107117] [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: 11/17/2024] [Accepted: 02/16/2025] [Indexed: 03/19/2025] Open
Abstract
Colorectal cancer (CRC) poses a significant threat to human life and health. Global cancer prevalence data in 2022 indicated that the number of new cases of CRC was about 1.92 million and the deaths were around 900,000. A variety of risk factors, including genes and environment, can induce the occurrence of CRC. Previous studies have focused on the impact of dietary patterns on the development of CRC and have ignored sleep factors. Sleep deprivation is a common problem as people's work pressure increases. Sleep disorders can lead to metabolic and immune system dysregulation in people, contributing to the development and progression of many tumors. At present, there are few reports on the relationship between sleep disorders and tumors. Therefore, the purpose of this paper is to summarize and interpret the relationship between various sleep disorders and the onset and progression of CRC. This review is the first to investigate the possible mechanisms of sleep leading to CRC from the perspectives of metabolic reprogramming, intestinal microbiota disorders, and the release of inflammatory factors. In conclusion, this study highlights the rational sleep pattern and duration, which can help inhibit the occurrence of CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhijun Hong
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
Zhao F, An R, Ma Y, Yu S, Gao Y, Wang Y, Yu H, Xie X, Zhang J. Integrated spatial multi-omics profiling of Fusobacterium nucleatum in breast cancer unveils its role in tumour microenvironment modulation and cancer progression. Clin Transl Med 2025; 15:e70273. [PMID: 40070022 PMCID: PMC11897063 DOI: 10.1002/ctm2.70273] [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: 12/01/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
Tumour-associated microbiota are integral components of the tumour microenvironment (TME). However, previous studies on intratumoral microbiota primarily rely on bulk tissue analysis, which may obscure their spatial distribution and localized effects. In this study, we applied in situ spatial-profiling technology to investigate the spatial distribution of intratumoral microbiota in breast cancer and their interactions with the local TME. Using 5R 16S rRNA gene sequencing and RNAscope FISH/CISH on patients' tissue, we identified significant spatial heterogeneity in intratumoral microbiota, with Fusobacterium nucleatum (F. nucleatum) predominantly localized in tumour cell-rich areas. GeoMx digital spatial profiling (DSP) revealed that regions colonized by F. nucleatum exhibit significant influence on the expression of RNAs and proteins involved in proliferation, migration and invasion. In vitro studies indicated that co-culture with F. nucleatum significantly stimulates the proliferation and migration of breast cancer cells. Integrative spatial multi-omics and co-culture transcriptomic analyses highlighted the MAPK signalling pathways as key altered pathways. By intersecting these datasets, VEGFD and PAK1 emerged as critical upregulated proteins in F. nucleatum-positive regions, showing strong positive correlations with MAPK pathway proteins. Moreover, the upregulation of VEGFD and PAK1 by F. nucleatum was confirmed in co-culture experiments, and their knockdown significantly reduced F. nucleatum-induced proliferation and migration. In conclusion, intratumoral microbiota in breast cancer exhibit significant spatial heterogeneity, with F. nucleatum colonization markedly altering tumour cell protein expression to promote progression and migration. These findings provide novel perspectives on the role of microbiota in breast cancer, identify potential therapeutic targets, and lay the foundation for future cancer treatments. KEY POINTS: Intratumoral Fusobacterium nucleatum exhibits significant spatial heterogeneity within breast cancer tissues. F. nucleatum colonization alters the expression of key proteins involved in tumour progression and migration. The MAPK signalling pathway is a critical mediator of F. nucleatum-induced breast cancer cell proliferation and migration. VEGFD and PAK1 are potential therapeutic targets to mitigate F. nucleatum-induced tumour progression.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Rui An
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yilei Ma
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Shaobo Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yuzhen Gao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yanzhong Wang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Haitao Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Xinyou Xie
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Jun Zhang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| |
Collapse
|
17
|
Yuan K, Xu H, Li S, Coker OO, Liu W, Wang L, Zhang X, Yu J. Intraneoplastic fungal dysbiosis is associated with colorectal cancer progression and host gene mutation. EBioMedicine 2025; 113:105608. [PMID: 39970705 PMCID: PMC11876754 DOI: 10.1016/j.ebiom.2025.105608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The relationship between intraneoplastic fungi and colorectal cancer (CRC) progression remains largely unclear. Here, we investigated fungal community changes in adenoma and CRC and their correlation with host genetic mutations. METHODS We obtained 261 tissue biopsies from two geographically distinct cohorts of CRC and adenoma patients, with each individual contributing 2-5 biopsies from lesions and 2 from adjacent normal tissues. 18S ribosomal RNA gene sequencing was used for microbial profiling. Host genetic alterations including KRAS mutations and microsatellite instability (MSI) were detected concurrently. FINDINGS Intra-neoplastic fungal composition significantly differed between CRC and adenoma in two independent cohorts, with enrichment of highly variable fungi (HVF) in CRC. Six HVFs exhibited higher abundances in adenoma and CRC compared to adjacent normal tissues with Malassezia showing a progressive increase from adenoma to CRC. Fungi intratumoral heterogeneity index also increased from adenoma through stages I to IV of CRC. Intra-tumoral fungi-fungi co-abundance analysis indicated stronger positive interactions in CRC than in adenoma, with increasingly robust links among intra-tumoral fungi along adenoma-CRC progression, primarily driven by Malassezia and Aspergillus. Furthermore, fungal heterogeneity was significantly correlated with host genetic mutations, with higher risk indices in CRC tissues harboring KRAS and MSI mutations. Thirteen fungi stratified CRC samples with KRAS mutations, achieving an area under the curve (AUC) of 0.86, while those associated with MSI status showed an AUC of 0.89. INTERPRETATION This study demonstrates that intraneoplastic fungal community alterations occur between adenoma and CRC, with increasing heterogeneity associated with host genetic mutations, emphasizing the role of fungal dysbiosis in CRC. FUNDING This work was supported by RGC Research Impact Fund Hong Kong (R4032-21F); RGC-CRF (C4008-23W); Strategic Seed Funding Collaboration Research Scheme CUHK (3133344); Strategic Impact Enhancement Fund CUHK (3135509); Impact case for RAE CUHK (3134277).
Collapse
Affiliation(s)
- Kai Yuan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Hongzhi Xu
- Institute for Microbial Ecology, School of Medicine, Xiamen University, Department of Gastroenterology, Zhongshan Hospital, Xiamen, China
| | - Shengmian Li
- Department of Gastroenterology, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Olabisi Oluwabukola Coker
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Weixin Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Luyao Wang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
18
|
Geng W, Ma K, Jiang Y, Peng S, Wang X. Association between gallbladder disease and colorectal neoplasia: a meta-analysis. Sci Rep 2025; 15:6276. [PMID: 39979467 PMCID: PMC11842739 DOI: 10.1038/s41598-025-91002-2] [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: 10/20/2024] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Although studies are available on the impact of gallbladder disease on the risk of colorectal neoplasia (CRN), the results are still debatable. We conducted a meta-analysis to summarize the correlation between gallbladder diseases and CRN. Eligible studies up to June 2024 were screened and retrieved using PubMed and Web of Science as well as by performing a manual review of references. Subgroup analyses stratified by region, location, and pathology of CRN were performed. Subgroup analyses stratified by classification and size of gallbladder disease were also performed. The pooled odd ratios (ORs) with 95% confidence intervals (CIs) were calculated. Sensitivity analyses were also performed. Begg's test was conducted to determine the publication bias. A total of twenty studies were included. The results showed that gallbladder disease significantly increased the risk of CRN (OR = 1.20, 95%CI, 1.11-1.29, P < 0.001). Subgroup analyses showed that subjects with gallstones (OR = 1.14, 95%CI, 1.05-1.25, P = 0.003) or gallbladder polyps (OR = 1.23, 95%CI, 1.15-1.31, P < 0.001) had a significantly higher risk of developing CRN. Asians (OR = 1.21, 95%CI, 1.11-1.31, P < 0.001) with gallstones were more likely to develop CRN. Patients with larger gallbladder polyps (≥ 0.5 cm) were at a greater risk of developing CRN (OR = 1.96, 95%CI, 1.41-2.73, P < 0.001). Gallbladder polyps and gallstones increase the risk of CRN. Therefore, colonoscopy should be performed in patients with gallbladder disease, especially in those of Asian descent, as well as in people with large gallbladder polyps.
Collapse
Affiliation(s)
- Wenbin Geng
- Department of Gastroenterology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, 68 Gehu middle road, Wujing District, Changzhou, 213000, Jiangsu Province, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Kai Ma
- Department of Gastroenterology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, 68 Gehu middle road, Wujing District, Changzhou, 213000, Jiangsu Province, China
- The Third Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yizhou Jiang
- Department of Gastroenterology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, 68 Gehu middle road, Wujing District, Changzhou, 213000, Jiangsu Province, China
- The Third Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiyu Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Xiaoyong Wang
- Department of Gastroenterology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, 68 Gehu middle road, Wujing District, Changzhou, 213000, Jiangsu Province, China.
- The Third Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
19
|
Liu L, Zhou Q, Xu T, Deng Q, Sun Y, Fu J, Chen M, Chen X, Ma Z, Dong Q, Ma B, Jiao Y, Zhou Y, Wu T, Zou H, Shi J, Wang Y, Sheng Y, Tang L, Zheng C, Wu W, Ma W, Sun W, Hu S, Zhou H, He Y, Kong X, Chen L. Non-differential gut microbes contribute to hypertension and its severity through co-abundances: A multi-regional prospective cohort study. IMETA 2025; 4:e268. [PMID: 40027484 PMCID: PMC11865328 DOI: 10.1002/imt2.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 03/05/2025]
Abstract
Microbial dysbiosis, characterized by an imbalanced microbial community structure and function, has been linked to hypertension. While prior research has primarily focused on differential abundances, our study highlights the role of non-differential microbes in hypertension. We propose that non-differential microbes contribute to hypertension through their ecological interactions, as defined by co-abundances (pairs of microbes exhibiting correlated abundance patterns). Using gut microbiome data from the Guangdong Gut Microbiome Project, which includes 2355 hypertensive and 4644 non-hypertensive participants across 14 regions, we identified replicable hypertension-related microbial interactions. Notably, most co-abundances involved non-differential microbes, which were found to correlate with both hypertension severity and hypertension-related microbial metabolic pathways. These findings emphasize the importance of microbial interactions in hypertension pathogenesis and propose a novel perspective for microbiome-based therapeutic strategies.
Collapse
Affiliation(s)
- Lu Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Qianyi Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Tianbao Xu
- Department of Cardiology, The Affiliated Kezhou People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityXinjiangChina
| | - Qiufeng Deng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Yuhao Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Jingxiang Fu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Muxuan Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaojiao Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhenchao Ma
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Quanbin Dong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Beining Ma
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yuwen Jiao
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Yan Zhou
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Tingting Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Huayiyang Zou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Jing Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yifeng Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Yanhui Sheng
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouChina
| | - Liming Tang
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| | - Chao Zheng
- Huzhou Central Hospital, Affiliated Huzhou HospitalZhejiang University School of MedicineHuzhouChina
- Department of Endocrinology, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Wei Wu
- Guangdong Provincial Institute of Public HealthGuangdong Provincial Center for Disease Control and PreventionGuangzhouChina
| | - Wenjun Ma
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouChina
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Clinical Research Center for Laboratory MedicineGuangzhouChina
- State Key Laboratory of Organ Failure ResearchSouthern Medical UniversityGuangzhouChina
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Clinical Research Center for Laboratory MedicineGuangzhouChina
- State Key Laboratory of Organ Failure ResearchSouthern Medical UniversityGuangzhouChina
- Key Laboratory of Mental Health of the Ministry of EducationGuangzhouChina
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouChina
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingChina
- Changzhou Medical Center, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical UniversityNanjing Medical UniversityChangzhouChina
| |
Collapse
|
20
|
Senthakumaran T, Tannæs TM, Moen AEF, Brackmann SA, Jahanlu D, Rounge TB, Bemanian V, Tunsjø HS. Detection of colorectal-cancer-associated bacterial taxa in fecal samples using next-generation sequencing and 19 newly established qPCR assays. Mol Oncol 2025; 19:412-429. [PMID: 38970464 PMCID: PMC11793011 DOI: 10.1002/1878-0261.13700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/15/2024] [Accepted: 06/28/2024] [Indexed: 07/08/2024] Open
Abstract
We have previously identified increased levels of distinct bacterial taxa within mucosal biopsies from colorectal cancer (CRC) patients. Following prior research, the aim of this study was to investigate the detection of the same CRC-associated bacteria in fecal samples and to evaluate the suitability of fecal samples as a non-invasive material for the detection of CRC-associated bacteria. Next-generation sequencing (NGS) of the 16S ribosomal RNA (rRNA) V4 region was performed to evaluate the detection of the CRC-associated bacteria in the fecal microbiota of cancer patients, patients with adenomatous polyp and healthy controls. Furthermore, 19 novel species-specific quantitative PCR (qPCR) assays were established to detect the CRC-associated bacteria. Approximately, 75% of the bacterial taxa identified in biopsies were reflected in fecal samples. NGS failed to detect low-abundance CRC-associated taxa in fecal samples, whereas qPCR exhibited high sensitivity and specificity in identifying all targeted taxa. Comparison of fecal microbial composition between the different patient groups showed enrichment of Fusobacterium nucleatum, Parvimonas micra, and Gemella morbillorum in cancer patients. Our findings suggest that low-abundance mucosa-associated bacteria can be detected in fecal samples using sensitive qPCR assays.
Collapse
Affiliation(s)
| | - Tone M. Tannæs
- Section for Clinical Molecular Biology (EpiGen)Akershus University HospitalLørenskogNorway
- Department of Clinical Molecular Biology, Institute of Clinical MedicineUniversity of OsloNorway
| | - Aina E. F. Moen
- Section for Clinical Molecular Biology (EpiGen)Akershus University HospitalLørenskogNorway
- Department of Clinical Molecular Biology, Institute of Clinical MedicineUniversity of OsloNorway
- Department of Methods Development and AnalyticsNorwegian Institute of Public HealthOsloNorway
| | - Stephan A. Brackmann
- Department of Gastroenterology, Division of MedicineAkershus University HospitalLørenskogNorway
- Institute for Clinical MedicineUniversity of OsloNorway
| | - David Jahanlu
- Department of Life Sciences and HealthOslo Metropolitan UniversityNorway
| | - Trine B. Rounge
- Department of Pharmacy, Centre for BioinformaticsUniversity of OsloNorway
- Department of ResearchCancer Registry of NorwayOsloNorway
| | - Vahid Bemanian
- Department of PathologyAkershus University HospitalLørenskogNorway
| | - Hege S. Tunsjø
- Department of Life Sciences and HealthOslo Metropolitan UniversityNorway
| |
Collapse
|
21
|
Feizi H, Kafil HS, Plotnikov A, Kataev V, Balkin A, Filonchikova E, Rezaee MA, Ghotaslou R, Sadrkabir M, Kadkhoda H, Kamounah FS, Nikitin S. Polyp and tumor microenvironment reprogramming in colorectal cancer: insights from mucosal bacteriome and metabolite crosstalk. Ann Clin Microbiol Antimicrob 2025; 24:9. [PMID: 39881353 PMCID: PMC11780822 DOI: 10.1186/s12941-025-00777-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Highly frequent colorectal cancer (CRC) is predicted to have 3.2 million novel cases by 2040. Tumor microenvironment (TME) bacteriome and metabolites are proposed to be involved in CRC development. In this regard, we aimed to investigate the bacteriome and metabolites of healthy, adenomatous polyp, and CRC tissues. METHODS Sixty samples including healthy (H), adenomatous polyps (AP), adenomatous polyps-adjacent (APA), cancer tumor (CT), and cancer tumor-adjacent (CA) tissues were collected and analyzed by 16 S rRNA sequencing and 1H NMR spectroscopy. RESULTS Our results revealed that the bacteriome and metabolites of the H, AP, and CT groups were significantly different. We observed that the Lachnospiraceae family depleted concomitant with acetoacetate and beta-hydroxybutyric acid (BHB) accumulations in the AP tissues. In addition, some bacterial species including Gemella morbillorum, and Morganella morganii were enriched in the AP compared to the H group. Furthermore, fumarate was accumulated concomitant to Aeromonas enteropelogenes, Aeromonas veronii, and Fusobacterium nucleatum subsp. animalis increased abundance in the CT compared to the H group. CONCLUSION These results proposed that beneficial bacteria including the Lachnospiraceae family depletion cross-talk with acetoacetate and BHB accumulations followed by an increased abundance of driver bacteria including G. morbillorum, and M. morganii may reprogram polyp microenvironment leading to tumor initiation. Consequently, passenger bacteria accumulation like A. enteropelogenes, A.veronii, and F. nucleatum subsp. animalis cross-talking fumarate in the TME may aggravate cancer development. So, knowledge of TME bacteriome and metabolites might help in cancer prevention, early diagnosis, and a good prognosis.
Collapse
Affiliation(s)
- Hadi Feizi
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Microbiology, Aalinasab Hospital, Social Security Organization, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Andrey Plotnikov
- Institute for Cellular and Intracellular Symbiosis of the Ural Branch of the Russian Academy of Sciences, Orenburg, Russia
| | - Vladimir Kataev
- Institute for Cellular and Intracellular Symbiosis of the Ural Branch of the Russian Academy of Sciences, Orenburg, Russia
| | - Alexander Balkin
- Institute for Cellular and Intracellular Symbiosis of the Ural Branch of the Russian Academy of Sciences, Orenburg, Russia
| | - Ekaterina Filonchikova
- Institute for Cellular and Intracellular Symbiosis of the Ural Branch of the Russian Academy of Sciences, Orenburg, Russia
| | - Mohammad Ahangarzadeh Rezaee
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Ghotaslou
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sadrkabir
- Department of Internal Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Hiva Kadkhoda
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Mahabad Faculty of Medical Sciences, , Urmia University of Medical Sciences, Urmia, Iran
| | - Fadhil S Kamounah
- Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | - Sergei Nikitin
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| |
Collapse
|
22
|
Vega AA, Shah PP, Rouchka EC, Clem BF, Dean CR, Woodrum N, Tanwani P, Siskind LJ, Beverly LJ. E. coli Biomolecules Increase Glycolysis and Invasive Potential in Lung Adenocarcinoma. Cancers (Basel) 2025; 17:380. [PMID: 39941749 PMCID: PMC11815989 DOI: 10.3390/cancers17030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction: Recent studies have discovered that lung cancer subtypes possess distinct microbiome profiles within their tumor microenvironment. Additionally, the tumor-associated microbiome exhibits altered bacterial pathways, suggesting that certain bacterial families are more capable of facilitating tumor progression than others. We hypothesize that there exists a crosstalk between lung adenocarcinoma (LUAD) cells and bacterial cells. Methods and Materials: RNA sequencing (RNA-seq) was performed on LUAD cell lines to explore the paracrine signaling effects of bacterial biomolecules. Based on our RNA-seq data, we investigated glycolysis by measuring glucose uptake and lactate production, invasive potential through invasion assays, and epithelial-to-mesenchymal transition (EMT) markers. Since lipopolysaccharides (LPS), abundant on the cell walls of Gram-negative bacteria, can activate toll-like receptor 4 (TLR4), we inhibited TLR4 with C34 to assess its relationship with the observed phenotypic changes. To identify the bacterial biomolecules responsible for these changes, we treated the media with RNAse enzyme, charcoal or dialyzed away molecules larger than 3 kDa. Results and Discussion: RNA-seq revealed 948 genes upregulated in the presence of E. coli biomolecules. Among these, we observed increased expression of Hexokinase II (HKII), JUN proto-oncogene, and Snail Family Transcriptional Repressor 1. We verified the elevation of glycolytic enzymes through Western blot and saw elevation of 2-deoxyglucose uptake and lactate production in LUAD cell lines incubated in E. coli biomolecules. In addition to E. coli elevating glycolysis in LUAD cell lines, E. coli exposure enhanced invasive potential as demonstrated by Boyden chamber assays. Notably, inhibition of TLR4 did not reduce the impact of E. coli biomolecules on glycolysis or the invasive potential of LUAD. Modulating the E. coli-supplemented media with RNAse enzyme or dextran-coated charcoal or using a spin column to remove biomolecules smaller than 3 kDa resulted in changes in HKII and Claudin protein expression. These findings suggest a direct relationship between E. coli and LUAD, wherein several cancer hallmarks are upregulated. Future studies should further investigate these bacterial biomolecules and their role in the tumor microenvironment to fully understand the impact of microbial shifts on cancer progression.
Collapse
Affiliation(s)
- Alexis A. Vega
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA (E.C.R.); (B.F.C.)
| | - Parag P. Shah
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Eric C. Rouchka
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA (E.C.R.); (B.F.C.)
- KY INBRE Bioinformatics Core, University of Louisville, Louisville, KY 40202, USA
| | - Brian F. Clem
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA (E.C.R.); (B.F.C.)
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Calista R. Dean
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Natassja Woodrum
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Preeti Tanwani
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Leah J. Siskind
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| | - Levi J. Beverly
- Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, USA; (P.P.S.); (C.R.D.); (N.W.); (P.T.); (L.J.S.)
| |
Collapse
|
23
|
Wang X, Zheng Z, Yu D, Qiu X, Yang T, Li R, Liu J, Wang X, Jin P, Sheng J, Qin N, Li N, Xu J. Colorectal cancer in Lynch syndrome families: consequences of gene germline mutations and the gut microbiota. Orphanet J Rare Dis 2025; 20:30. [PMID: 39827259 PMCID: PMC11742751 DOI: 10.1186/s13023-025-03543-4] [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: 01/23/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Lynch syndrome (LS)-associated colorectal cancer (CRC) always ascribes to pathogenic germline mutations in mismatch repair (MMR) genes. However, the penetrance of CRC varies among those with the same MMR gene mutation. Thus, we hypothesized that the gut microbiota is also involved in CRC development in LS families. METHODS This prospective, observational study was performed from December 2020 to March 2023. We enrolled 72 individuals from 9 LS families across six provinces in China and employed 16S rRNA gene amplicon sequencing to analyze the fecal microbiota components among LS-related CRC patients (AS group), their spouses (BS group), mutation carriers without CRC (CS group), and non-mutation carriers (DS group) using alpha and beta diversity indices. RESULTS There were no apparent differences in age or gender among the four groups. Alpha and beta diversity indices exhibited no significant differences between the AS and BS groups, verifying the role of germline mutations in the occurrence of CRC in LS families. Beta diversity analysis exhibited significant differences between the AS and CS groups, revealing the importance of the gut microbiota for the occurrence of CRC in LS families. A greater difference (both alpha and beta diversity indices) was shown between the AS and DS groups, demonstrating the combined impact of the gut microbiota and genetic germline mutations on the occurrence of CRC in LS families. Compared with those in the CS and DS groups, we identified ten microbial genera enriched in the AS group, and one genus (Bacteroides) decreased in the AS group. Among the elevated genera in the AS group, Agathobacter, Coprococcus and Prevotellaceae_NK3B31_group were butyrate-producing genera. CONCLUSION This study found the development of CRC in the LS families can be attributed to the combined effects of gene germline mutations as well as the gut microbiota and provided novel insights into the prevention and treatment of CRC in the LS families.
Collapse
Affiliation(s)
- Xuexin Wang
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhijun Zheng
- Realbio Genomics Institute, Shanghai, 201114, China.
| | - Dongliang Yu
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China
| | - Xiaojue Qiu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Ting Yang
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Ruoran Li
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jing Liu
- Realbio Genomics Institute, Shanghai, 201114, China
- Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| | - Xin Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China
| | - Peng Jin
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianqiu Sheng
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China
| | - Nan Qin
- Tenth People's Hospital of Tongji University, Shanghai, 200072, China
- Qingdao Realbio Precision Medical Test Co. Ltd, Qingdao, 266071, China
| | - Na Li
- Medical School of Chinese PLA, Beijing, 100853, China.
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Junfeng Xu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
24
|
Kim MJ, Song MH, Ji YS, Park JW, Shin YK, Kim SC, Kim G, Cho B, Park H, Ku JL, Jeong SY. Cell free supernatants of Bifidobacterium adolescentis and Bifidobacterium longum suppress the tumor growth in colorectal cancer organoid model. Sci Rep 2025; 15:935. [PMID: 39762302 PMCID: PMC11704243 DOI: 10.1038/s41598-024-83048-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The probiotic gut microbiome and its metabolites are pivotal in regulating host metabolism, inflammation, and immunity. Host genetics, colonization at birth, the host lifestyle, and exposure to diseases and drugs determine microbial composition. Dysbiosis and disruption of homeostasis in the beneficial microbiome have been reported to be involved in the tumorigenesis and progression of colorectal cancer (CRC). However, the influence of bacteria-secreted metabolites on CRC growth is yet to be fully elucidated. In this study, we compared the microbial composition of CRC patients to healthy controls to identify distinct patterns of microbiota-derived metabolites in CRC patients. Metagenomic analysis demonstrated that beneficial bacteria strains; Blautia producta, Bifidobacterium adolescentis, and Bifidobacterium longum decreased, while Parabacteroides distasonis and Bacteroides ovatus were more prevalent in the CRC patient group. Treatment of cancer organoid lines with microbial culture supernatants from Blautia producta, Bifidobacterium adolescentis, and Bifidobacterium longum showed remarkable inhibition of cancer growth. This study demonstrates that the bacterial metabolites depleted in CRC patients may inhibit cancer growth and highlights the effects of microbiome-derived metabolites on CRC growth.
Collapse
Affiliation(s)
- Min Jung Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Myoung-Hyun Song
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yo-Sep Ji
- Holzapfel Effective Microbes (HEM) Pharma, Handong Global University, Pohang, Gyungbuk, Republic of Korea
| | - Ji Won Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Young-Kyoung Shin
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Soon-Chan Kim
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Gihyeon Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Beomki Cho
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Hansoo Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Ja-Lok Ku
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Seung-Yong Jeong
- Department of Surgery, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
25
|
Valciukiene J, Lastauskiene E, Laurinaviciene A, Jakubauskas M, Kryzauskas M, Valkiuniene RB, Augulis R, Garnelyte A, Kavoliunas J, Silinskaite U, Poskus T. Interaction of human gut microbiota and local immune system in progression of colorectal adenoma (MIMICA-1): a protocol for a prospective, observational cohort study. Front Oncol 2025; 14:1495635. [PMID: 39834942 PMCID: PMC11743970 DOI: 10.3389/fonc.2024.1495635] [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: 09/13/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION The current understanding of colorectal carcinogenesis is based on the adenoma-carcinoma sequence, where genetics, intestinal microbiota changes and local immunity shifts seem to play the key roles. Despite the emerging evidence of dysbiotic intestinal state and immune-cell infiltration changes in patients with colorectal adenocarcinoma, early and advanced adenoma as precursors of colorectal cancer, and carcinoma in situ as the following progression, are rather less studied. The newly colon-site adapted AI-based analysis of immune infiltrates is able to predict long-term outcomes of colon carcinoma. Though it could also facilitate the pathologic evaluation of precancerous lesion's potential to progress. Therefore, the purpose of this prospective cohort study (MIMICA-1) is, firstly, to identify the intestinal microbiota and immune infiltration patterns around the normal bowel tissue, early and advanced adenoma, carcinoma in situ, and adenocarcinoma, and secondly, to analyze the immune - microbiome interplay along the steps of conventional colorectal tumorigenesis. METHODS AND ANALYSES This study aims to prospectively recruit 40 patients (10 per group) with confirmed colorectal dysplasia undergoing endoscopic polypectomy, endoscopic mucosal resection for colorectal small (≤1cm), and large (>1cm) adenoma or carcinoma in situ, or biopsy and subsequent colon resection for invasive colorectal cancer, and 10 healthy patients undergoing screening colonoscopy. Stool samples will be collected prior to bowel preparation for the analysis of fecal (luminal) microbiota composition. Biopsy specimens will be taken from the terminal ileum, right colon, left colon, and a pathological lesion in the colon (if present) to assess mucosa-associated microbiota composition and intestinal immunity response. DNA will be extracted from all samples and sequenced using the Illumina MiSeq platform. Unifrac and Bray-Curtis methods will be used to assess microbial diversity. The intestinal immune system response will be examined using digital image analysis where primarily immunohistochemistry procedures for CD3, CD8, CD20 and CD68 immune cell markers will be performed. Thereafter, the count, density and distribution of immunocompetent cells in epithelial and stromal tissue compartments will be evaluated using AI-based platform. The interaction between the microbial shifts and intestinal immune system response in adenoma-carcinoma sequence and the healthy patients will be examined. In addition, fecal samples will be explored for gut microbiota's composition, comparing fecal- and tissue-derived bacterial patterns in healthy gut and along the adenoma-carcinoma sequence. DISCUSSION We hypothesize that changes within the human gut microbiota led to detectable alterations of the local immune response and correlate with the progression from normal mucosa to colorectal adenoma and invasive carcinoma. It is expectable to find more severe gut immune infiltration at dysplasia site, though analyzing invasive colorectal cancer we expect to detect broader mucosa-associated and luminal microbiota changes with subsequent local immune response at near-lesion site and possibly throughout the entire colon. We believe that specific compositional differences detected around premalignant colorectal lesions are critically important for its primary role in initiation and acceleration of colorectal carcinogenesis. Thus, these microbial patterns could potentially supplement fecal immunohistochemical tests for the early non-invasive detection of colorectal adenoma. Moreover, AI-based analysis of immune infiltrates could become additional diagnostic and prognostic tool in precancerous lesions prior to the development of colorectal cancer. REGISTRATION The study is registered at the Australian New Zealand Clinical Trials Registry (ACTRN12624000976583) https://www.anzctr.org.au/.
Collapse
Affiliation(s)
- Jurate Valciukiene
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Egle Lastauskiene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aida Laurinaviciene
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Matas Jakubauskas
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Marius Kryzauskas
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ruta Barbora Valkiuniene
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Renaldas Augulis
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ausra Garnelyte
- National Center of Pathology, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Justinas Kavoliunas
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Tomas Poskus
- Clinic of Gastroenterology, Nephro-Urology, and Surgery, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
26
|
Xia D, Jin L, Wang B, Jin Y, Zheng Q, Xu J, Chen S. Alpha-glucosidase inhibitor decreases the risk of colorectal adenoma in the aged with Type 2 diabetes. Sci Rep 2025; 15:583. [PMID: 39748054 PMCID: PMC11696837 DOI: 10.1038/s41598-024-84294-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
The rapidly aging population is fueling a surge in diabetes, especially Type 2, which heightens colorectal cancer (CRC) risk. Colorectal adenoma, a precursor, compounds this trend. Although alpha-glucosidase inhibitors are effective hypoglycemic drugs working in the GI tract, the link between them and colorectal adenoma formation remains unexplored. A retrospective cross-sectional study was conducted on type 2 diabetes patients aged 60 and above using data from Wenzhou Central Hospital from January 2021 to May 2024. We used multivariable logistic regression and propensity score matching analysis (PSM) to calculate adjusted ORs for colorectal adenoma, controlling for potential confounders. A total of 311 subjects were enrolled in the study, with a mean age of 67.55 years. 138 (44.4%) were diagnosed with colorectal adenoma. Multivariate logistic regression analysis revealed that the AGI (Alpha-glucosidase inhibitor) Group had an adjusted OR of 0.399 (95% CI = 0.22-0.723, p = 0.002) compared to those with AGI free people. A similar trend was also observed in the PSM analysis (OR = 0.362, 95% CI = 0.176-0.744, p = 0.004). Subgroup analysis reveals hypertension as a potential modulator of the inverse relationship between AGI and colorectal adenoma occurrence post-PSM (p = 0.049). And AGI reduces serum iron levels, both before (p = 0.01) and after PSM (p = 0.028). In summary, our findings indicate that AGI significantly mitigates the risk of colorectal adenoma among individuals aged 60 and above, particularly among those afflicted with hypertension. Additionally, it substantially decreases serum iron levels.
Collapse
Affiliation(s)
- Dingchao Xia
- Department of Infectious Diseases, Wenzhou Central Hospital, Wenzhou, 325000, Zhejiang, China
- Department of Infectious Diseases, Wenzhou Sixth People's Hospital, Wenzhou, 325000, Zhejiang, China
| | - Lanling Jin
- Department of Neurology, Pujiang County People's Hospital, Wenzhou, Jinhua, 322200, Zhejiang, China
| | - Binfeng Wang
- Department of Gastroenterology, Affiliated Yueqing Hospital,Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yi Jin
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qun Zheng
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jie Xu
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Senzhong Chen
- Department of Gerontology, Wenzhou Central Hospital, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
27
|
Jain P, Mohapatra S, Farooq U, Hassan N, Mirza MA, Iqbal Z. An Overview of the Dichotomous Role of Microbiota in Cancer Progression and Management. Curr Cancer Drug Targets 2025; 25:38-48. [PMID: 38409691 DOI: 10.2174/0115680096282503240124104029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/28/2024]
Abstract
It is a well-known fact that cancer is considered the second leading cause of mortality across the globe. Although the human oral cavity and intestine are the natural habitat of thousands of microbes, dysbiosis results in malignancies, such as oral squamous cell carcinoma and colorectal cancer. Amongst the intestinal microbes, H. pylori is a deadly carcinogen. Also, causative pathogens for the development of pancreatic and colorectal cancer are found in the oral cavity, such as Fusobacterium nucleatum and Porphyromonas gingivalis. Many periodontopathic micro- organisms, like Streptococcus sp., Peptostreptococcus sp., Prevotella sp., Fusobacterium sp., Porphyromonas gingivalis, and Capnocytophaga gingivalis, strongly have an impact on the development of oral cancers. Three basic mechanisms are involved in pathogen-mediated cancer development, like chronic inflammation-mediated angiogenesis, inhibition of cellular apoptosis, and release of carcinogenic by-products. Microbiota has a dichotomous role to play in cancer, i.e., microbiota can be used for cancer management too. Shreds of evidence are there to support the fact that microbiota enhances the chemotherapeutic drug efficacy. This review presents the possible mechanism of the oncogenic effect of microbiota with emphasis on the oral microbiome and also attempts to explain the intricate role of microbiota in cancer management.
Collapse
Affiliation(s)
- Pooja Jain
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Sradhanjali Mohapatra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Uzma Farooq
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Nazia Hassan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Mohd Aamir Mirza
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| | - Zeenat Iqbal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110030, India
| |
Collapse
|
28
|
Lattanzi G, Perillo F, Díaz-Basabe A, Caridi B, Amoroso C, Baeri A, Cirrincione E, Ghidini M, Galassi B, Cassinotti E, Baldari L, Boni L, Vecchi M, Caprioli F, Facciotti F, Strati F. Estrogen-related differences in antitumor immunity and gut microbiome contribute to sexual dimorphism of colorectal cancer. Oncoimmunology 2024; 13:2425125. [PMID: 39548749 PMCID: PMC11572150 DOI: 10.1080/2162402x.2024.2425125] [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: 05/21/2024] [Revised: 10/18/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024] Open
Abstract
Colorectal cancer (CRC) is a multifaceted disease whose development and progression varies depending on tumor location, age of patients, infiltration of immune cells within cancer lesions, and the tumor microenvironment. These pathophysiological characteristics are additionally influenced by sex-related differences. The gut microbiome plays a role in initiation and progression of CRC, and shapes anti-tumor immune responses but how responsiveness of the immune system to the intestinal microbiota may contribute to sexual dimorphism of CRC is largely unknown. We studied survival, tumor-infiltrating immune cell populations and tumor-associated microbiome of a cohort of n = 184 male and female CRC patients through high-dimensional single-cell flow cytometry and 16S rRNA gene sequencing. We functionally tested the immune system-microbiome interactions in in-vivo and in-vitro models of the disease. High-dimensional single-cell flow cytometry showed that female patients are enriched by tumor-infiltrating invariant Natural Killer T (iNKT) cells but depleted by cytotoxic T lymphocytes. The enrichment of oral pathobionts and a reduction of β-glucuronidase activity are distinctive traits characterizing the gut microbiome of female patients affected by CRC. Functional assays using a collection of human primary iNKT cell lines demonstrated that the gut microbiota of female patients functionally impairs iNKT cell anti-tumor functions interfering with the granzyme-perforin cytotoxic pathway. Our results highlight a sex-dependent functional relationship between the gut microbiome, estrogen metabolism, and the decline of cytotoxic T cell responses, contributing to the sexual dimorphism observed in CRC patients with relevant implications for precision medicine and the design of targeted therapeutic approaches addressing sex bias in cancer.
Collapse
Affiliation(s)
- Georgia Lattanzi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Perillo
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Angélica Díaz-Basabe
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Bruna Caridi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alberto Baeri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Elisa Cirrincione
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Michele Ghidini
- Medical Oncology, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Galassi
- Medical Oncology, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Cassinotti
- Department of General & Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Ludovica Baldari
- Department of General & Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Luigi Boni
- Department of General & Minimally Invasive Surgery, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Maurizio Vecchi
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Federica Facciotti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Francesco Strati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
29
|
Rong J, Chen X, Li Z, Li B, Sun Y, Miao Y. Dysregulation of saliva and fecal microbiota as novel biomarkers of colorectal cancer. Front Oncol 2024; 14:1498328. [PMID: 39743994 PMCID: PMC11688226 DOI: 10.3389/fonc.2024.1498328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
The aim of this study was to investigate the biomarkers of salivary and fecal microbiota in Colorectal cancer (CRC). Initially, the study scrutinized the microbial community composition disparities among groups. Utilizing Lasso analysis, it sifted through operational taxonomic units (OTUs) to pinpoint distinctive features. Subsequently, by intersecting feature OTUs across groups, it curated a set of core-shared OTUs and devised a corresponding network. Concluding with functional enrichment analysis, the research delved into the divergent biological functions of these microbial communities within the studied groups. Analysis revealed higher bacterial diversity in saliva compared to feces, with distinct differences at both phylum and genus levels. Feces primarily contained Firmicutes, while saliva was dominated by Bacteroidetes and Proteobacteria. Notably, Escherichia-Shigella and Fusobacterium in feces and Streptococcus in saliva showed increasing abundance from average to adenoma to colorectal cancer. Specific dominant flora was identified within and between groups, including CRC and adenomas across different stages. Seventeen core shared OTUs were identified, and networks of shared OTUs were constructed for each group. Functional enrichment analysis highlighted distinct microbial community functions among the groups. This study's findings on characteristic OTUs in saliva and fecal samples offer valuable insights for distinguishing between healthy individuals, adenoma patients, and those with colorectal cancer. This study identified distinctive OTUs in saliva and feces to distinguish between healthy individuals, adenoma patients, and those with CRC, offering a valuable diagnostic reference.
Collapse
Affiliation(s)
- Jiamei Rong
- Yan’an Hospital Affiliated To Kunming Medical University, Kunming, Yunnan, China
| | - Xiaocui Chen
- Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Zhangqin Li
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bona Li
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yang Sun
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yinglei Miao
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
30
|
Ma Y, Chen T, Sun T, Dilimulati D, Xiao Y. The oncomicrobiome: New insights into microorganisms in cancer. Microb Pathog 2024; 197:107091. [PMID: 39481695 DOI: 10.1016/j.micpath.2024.107091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
The discoveries of the oncomicrobiome (intratumoral microbiome) and oncomicrobiota (intratumoral microbiota) represent significant advances in tumor research and have rapidly become of key interest to the field. Within tumors, microorganisms such as bacteria, fungi, viruses, and archaea form the oncomicrobiota and are primarily found within tumor cells, immunocytes, and the intercellular matrix. The oncomicrobiome exhibits marked heterogeneity and is associated with tumor initiation, progression, metastasis, and treatment response. Interactions between the oncomicrobiome and the immune system can modulate host antitumor immunity, influencing the efficacy of immunotherapies. Oncomicrobiome research also faces numerous challenges, including overcoming methodological issues such as low target abundance, susceptibility to contamination, and biases in sample handling and analysis methods across different studies. Furthermore, studies of the oncomicrobiome may be confounded by baseline differences in microbiomes among populations driven by both environmental and genetic factors. Most studies to date have revealed associations between the oncomicrobiome and tumors, but very few have established mechanistic links between the two. This review introduces the relevant concepts, detection methods, sources, and characteristics of the oncomicrobiome. We then describe the composition of the oncomicrobiome in common tumors and its role in shaping the tumor microenvironment. We also discuss the current problems and challenges to be overcome in this rapidly progressing field.
Collapse
Affiliation(s)
- Yingying Ma
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingting Sun
- Department of Structure and Morphology, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China; Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Dilinuer Dilimulati
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Structure and Morphology, Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China; Peking Union Medical College & Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
31
|
Williams CJM, Peddle AM, Kasi PM, Seligmann JF, Roxburgh CS, Middleton GW, Tejpar S. Neoadjuvant immunotherapy for dMMR and pMMR colorectal cancers: therapeutic strategies and putative biomarkers of response. Nat Rev Clin Oncol 2024; 21:839-851. [PMID: 39317818 DOI: 10.1038/s41571-024-00943-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 09/26/2024]
Abstract
Approximately 15% of locally advanced colorectal cancers (CRC) have DNA mismatch repair deficiency (dMMR), resulting in high microsatellite instability and a high tumour mutational burden. These cancers are frequently sensitive to therapy with immune-checkpoint inhibitors (ICIs) in the metastatic setting. This sensitivity seems to be even more pronounced in locally advanced disease, and organ preservation has become a realistic aim in ongoing clinical trials involving patients with dMMR rectal cancer. By contrast, metastatic CRCs with proficient DNA mismatch repair (pMMR) are generally resistant to ICIs, although a proportion of locally advanced pMMR tumours seem to have a high degree of sensitivity to ICIs. In this Review, we describe the current and emerging clinical evidence supporting the use of neoadjuvant ICIs in patients with dMMR and pMMR CRC, and the potential advantages (based on a biological rationale) of such an approach. We discuss how neoadjuvant 'window-of-opportunity' trials are being leveraged to progress biomarker discovery and we provide an overview of potential predictive biomarkers of response to ICIs, exploring the challenges faced when evaluating such biomarkers in biopsy-derived samples. Lastly, we describe how these discoveries might be used to drive a rational approach to trialling novel immunotherapeutic strategies in patients with pMMR CRC, with the ultimate aim of disease eradication and the generation of long-term immunosurveillance.
Collapse
Affiliation(s)
| | | | - Pashtoon M Kasi
- Department of Gastrointestinal Oncology, City of Hope Orange County Lennar Foundation Cancer Center, Irvine, CA, USA
| | - Jenny F Seligmann
- Division of Oncology, Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | | | - Gary W Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | |
Collapse
|
32
|
Permain J, Hock B, Eglinton T, Purcell R. Functional links between the microbiome and the molecular pathways of colorectal carcinogenesis. Cancer Metastasis Rev 2024; 43:1463-1474. [PMID: 39340753 PMCID: PMC11554747 DOI: 10.1007/s10555-024-10215-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Colorectal cancer (CRC) is a common cancer, with a concerning rise in early-onset CRC cases, signalling a shift in disease epidemiology. Whilst our understanding of the molecular underpinnings of CRC has expanded, the complexities underlying its initiation remain elusive, with emerging evidence implicating the microbiome in CRC pathogenesis. This review synthesizes current knowledge on the intricate interplay between the microbiome, tumour microenvironment (TME), and molecular pathways driving CRC carcinogenesis. Recent studies have reported how the microbiome may modulate the TME and tumour immune responses, consequently influencing cancer progression, and whilst specific bacteria have been linked with CRC, the underlying mechanisms remains poorly understood. By elucidating the functional links between microbial landscapes and carcinogenesis pathways, this review offers insights into how bacteria orchestrate diverse pathways of CRC development, shedding light on potential therapeutic targets and personalized intervention strategies.
Collapse
Affiliation(s)
- Jessica Permain
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Barry Hock
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Timothy Eglinton
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Rachel Purcell
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
33
|
Zinkeng A, Taylor FL, Cheong SH, Song H, Merchant JL. Early Onset Colorectal Cancer: Molecular Underpinnings Accelerating Occurrence. Cell Mol Gastroenterol Hepatol 2024; 19:101425. [PMID: 39510499 PMCID: PMC11731505 DOI: 10.1016/j.jcmgh.2024.101425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
The onset of colorectal cancer (CRC) in patients younger than 50 continues to rapidly increase. This study highlights the epidemiologic changes, risk factors, clinical characteristics, and molecular profiles prevalent in early onset CRC patients, and identifies key areas for future research. It has been noted that only a small fraction of early onset CRC cases is attributed to known hereditary mutations and fit the canonical pathway of late-onset colorectal cancer development. To highlight this, we review the genetic and epigenetic modifications specific to early onset CRC. We also discuss the synergetic effect of single-nucleotide polymorphisms and environmental factors on the early onset of CRC. Additionally, we discuss the potential of noninvasive biomarker assays to enhance early detection, screening, diagnosis, and prognostic outcome predictions.
Collapse
Affiliation(s)
- Atehkeng Zinkeng
- Medical Scientist Training Program, University of Arizona College of Medicine, Tucson, Arizona
| | | | | | | | - Juanita L Merchant
- Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona.
| |
Collapse
|
34
|
Antonacci A, Bizzoca C, Barile G, Andriola V, Vincenti L, Bartolomeo N, Abbinante A, Orrù G, Corsalini M. Evaluation of Periodontitis and Fusobacterium nucleatum Among Colorectal Cancer Patients: An Observational Cross-Sectional Study. Healthcare (Basel) 2024; 12:2189. [PMID: 39517401 PMCID: PMC11545387 DOI: 10.3390/healthcare12212189] [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: 09/29/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Periodontitis has been associated with an increased risk of CRC, as well as a worse prognosis due to increased inflammation mediators and carcinogenic factors. Moreover, direct and indirect virulence factors from periodontal pathogens, such as Fusobacterium nucleatum, could play a pivotal role in malignant transformation and progression. This cross-sectional study aims to evaluate the presence and the stage of periodontitis in a cohort of patients with CRC. The secondary aim is to assess the presence of F. nucleatum and its relationship with patients' general characteristics, concomitant pathologies, tumor characteristics, and drug therapy. MATERIALS AND METHODS Patients affected by CRC underwent dental examination and periodontal charting with the "North Carolina" probe to assess the presence and stage of periodontitis, according to the new classification of periodontal diseases of the World Workshop of the European Federation of Periodontology (EFP) and the American Academy of Periodontology (AAP) 2017. F. nucleatum presence was assessed by a dorsal tongue swab and related to the patient's general characteristics, concomitant pathologies, tumor characteristics, and drug therapy. RESULTS Periodontal disease was found in 94.3% of I/II CRC stage patients and 100% of III/IV CRC stage patients. Severe periodontitis was found in 76% of the advanced CRC stage and 87.9% of patients with initial CRC, while initial periodontitis was found in 12.1% of initial CRC and 24% of late CRC stages, respectively, without significant differences. F. nucleatum presence showed no correlation between the patient's and tumor's characteristics, comorbidities, and drug assumed. CONCLUSIONS Periodontal disease showed a high prevalence among CRC patients. Moreover, severe periodontitis has a higher prevalence in CRC patients compared to initial periodontitis. F. nucleatum presence was unrelated to CRC stage, site, other comorbidities, and drug therapies. With these data, it is not possible to admit a direct relationship between CRC and periodontal disease, but further case-control studies must be carried out to further prove this aspect. Preventive and operative targeted strategies to maintain a healthy oral status are suggested in CRC patients.
Collapse
Affiliation(s)
- Anna Antonacci
- Department of Interdisciplinary Medicine, ‘Aldo Moro’, University of Bari, 70100 Bari, Italy; (A.A.); (N.B.); (A.A.)
| | - Cinzia Bizzoca
- Department of General Surgery “Ospedaliera”, Polyclinic Hospital of Bari, 70100 Bari, Italy; (C.B.); (V.A.); (M.C.)
| | - Giuseppe Barile
- Department of Interdisciplinary Medicine, ‘Aldo Moro’, University of Bari, 70100 Bari, Italy; (A.A.); (N.B.); (A.A.)
| | - Valeria Andriola
- Department of General Surgery “Ospedaliera”, Polyclinic Hospital of Bari, 70100 Bari, Italy; (C.B.); (V.A.); (M.C.)
| | - Leonardo Vincenti
- General Surgery Unit, National Institute of Gastroenterology IRCCS Saverio de Bellis, Research Hospital, Via Turi 27, 0013 Bari, Italy;
| | - Nicola Bartolomeo
- Department of Interdisciplinary Medicine, ‘Aldo Moro’, University of Bari, 70100 Bari, Italy; (A.A.); (N.B.); (A.A.)
| | - Antonia Abbinante
- Department of Interdisciplinary Medicine, ‘Aldo Moro’, University of Bari, 70100 Bari, Italy; (A.A.); (N.B.); (A.A.)
| | - Germano Orrù
- Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy;
| | - Massimo Corsalini
- Department of General Surgery “Ospedaliera”, Polyclinic Hospital of Bari, 70100 Bari, Italy; (C.B.); (V.A.); (M.C.)
| |
Collapse
|
35
|
Li Q, Geng S, Luo H, Wang W, Mo YQ, Luo Q, Wang L, Song GB, Sheng JP, Xu B. Signaling pathways involved in colorectal cancer: pathogenesis and targeted therapy. Signal Transduct Target Ther 2024; 9:266. [PMID: 39370455 PMCID: PMC11456611 DOI: 10.1038/s41392-024-01953-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Its complexity is influenced by various signal transduction networks that govern cellular proliferation, survival, differentiation, and apoptosis. The pathogenesis of CRC is a testament to the dysregulation of these signaling cascades, which culminates in the malignant transformation of colonic epithelium. This review aims to dissect the foundational signaling mechanisms implicated in CRC, to elucidate the generalized principles underpinning neoplastic evolution and progression. We discuss the molecular hallmarks of CRC, including the genomic, epigenomic and microbial features of CRC to highlight the role of signal transduction in the orchestration of the tumorigenic process. Concurrently, we review the advent of targeted and immune therapies in CRC, assessing their impact on the current clinical landscape. The development of these therapies has been informed by a deepening understanding of oncogenic signaling, leading to the identification of key nodes within these networks that can be exploited pharmacologically. Furthermore, we explore the potential of integrating AI to enhance the precision of therapeutic targeting and patient stratification, emphasizing their role in personalized medicine. In summary, our review captures the dynamic interplay between aberrant signaling in CRC pathogenesis and the concerted efforts to counteract these changes through targeted therapeutic strategies, ultimately aiming to pave the way for improved prognosis and personalized treatment modalities in colorectal cancer.
Collapse
Affiliation(s)
- Qing Li
- The Shapingba Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shan Geng
- Central Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Wang
- Chongqing Municipal Health and Health Committee, Chongqing, China
| | - Ya-Qi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lu Wang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jian-Peng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
36
|
Kane KJ, Jensen CD, Yang J, Dong H, Merchant SA, Koripella P, Li X, Hendel JM, Corley DA, Lee JK. Oral Antibiotic Use in Adulthood and Risk of Early-Onset Colorectal Cancer: A Case-Control Study. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00859-0. [PMID: 39368633 DOI: 10.1016/j.cgh.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND AND AIMS Prior antibiotic use may be a factor in the rising incidence of colorectal cancer seen in those under 50 years of age (early-onset colorectal cancer [EOCRC]); however, the few studies to examine this link have reported conflicting results. Therefore, we evaluated the association between oral antibiotic use in adulthood and EOCRC in a large integrated healthcare system in the United States. METHODS A population-based nested case-control study was conducted among Kaiser Permanente Northern California patients 18-49 years of age diagnosed with EOCRC (adenocarcinoma of the colon or rectum) in 1998-2020 who had ≥2 years of continuous pharmacy benefit prior to diagnosis. Cases were matched 4:1 to healthy controls on birth year, sex, race and ethnicity, medical facility, and duration of pharmacy benefit. Antibiotic exposure >1 year before the diagnosis/index date was assessed using prescribing records. Conditional logistic regression was used to estimate odds ratios and 95% confidence intervals. A sensitivity analysis was performed among those with ≥10 years of continuous prescribing records. RESULTS A total of 1359 EOCRC cases were matched to 4711 healthy controls. Antibiotic use in adulthood was not significantly associated with EOCRC in unadjusted or adjusted analyses (adjusted odds ratio, 1.04; 95% confidence interval, 0.94-1.26). No associations were seen for cumulative number of oral antibiotic dispensations or for any prior period of antibiotic exposure. CONCLUSIONS In a large U.S. healthcare setting, there was no conclusive evidence of an association between oral antibiotic use in adulthood and risk of EOCRC.
Collapse
Affiliation(s)
- Kevin J Kane
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California
| | - Christopher D Jensen
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Jingrong Yang
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Huyun Dong
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Sophie A Merchant
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Pradeep Koripella
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California
| | - Xiaoran Li
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California
| | - Jeffrey M Hendel
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California
| | - Douglas A Corley
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California; Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Jeffrey K Lee
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California; Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| |
Collapse
|
37
|
Guo C, An Q, Zhang L, Wei X, Xu J, Yu J, Wu G, Ma J. Intratumoral microbiota as cancer therapeutic target. Aging Med (Milton) 2024; 7:636-644. [PMID: 39507228 PMCID: PMC11535161 DOI: 10.1002/agm2.12359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
Intratumoral microbiota, which affects the physiological and pathological processes of the host, has attracted increasing attention from researchers. Microbials have been found in normal as well as tumor tissues that were originally thought to be sterile. Intratumoral microbiota is considered to play a significant role in the development of tumors and the reduction of clinical benefits. In addition, intratumoral microbiota are heterogeneous, which have different distribution in various types of tumors, and can influence tumor development through different mechanisms, including genome mutations, inflammatory responses, activated cancer pathways, and immunosuppressive microenvironments. Therefore, eliminating the intratumoral microbiota is considered one of the most promising ways to slow down the tumor progression and improve therapeutic outcomes. In this review, we systematically categorized the intratumoral microbiota and elucidated its role in the pathogenesis and therapeutic response of cancer. We have also described the novel strategies to mitigate the impact of tumor progression. We hope this review will provide new insights for the anti-tumor treatment, particularly for the elderly population, where such insights could significantly enhance treatment outcomes.
Collapse
Affiliation(s)
- Chang Guo
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
- Medical SchoolUniversity of Chinese Academy of SciencesBeijingPeople's Republic of China
| | - Qi An
- General Surgery Department, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Lu‐yao Zhang
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Xun‐dong Wei
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jing Xu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jiang‐yong Yu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Guo‐ju Wu
- General Surgery Department, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| |
Collapse
|
38
|
Flory M, Bravo P, Alam A. Impact of gut microbiota and its metabolites on immunometabolism in colorectal cancer. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00050. [PMID: 39624362 PMCID: PMC11608621 DOI: 10.1097/in9.0000000000000050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/17/2024] [Indexed: 01/25/2025]
Abstract
Colorectal cancer (CRC) is highly prevalent, accounting for approximately one-tenth of cancer cases and deaths globally. It stands as the second most deadly and third most common cancer type. Although the gut microbiota has been implicated in CRC carcinogenesis for the last several decades, it remains one of the least understood risk factors for CRC development, as the gut microbiota is highly diverse and variable. Many studies have uncovered unique microbial signatures in CRC patients compared with healthy matched controls, with variations dependent on patient age, disease stage, and location. In addition, mechanistic studies revealed that tumor-associated bacteria produce diverse metabolites, proteins, and macromolecules during tumor development and progression in the colon, which impact both cancer cells and immune cells. Here, we summarize microbiota's role in tumor development and progression, then we discuss how the metabolic alterations in CRC tumor cells, immune cells, and the tumor microenvironment result in the reprogramming of activation, differentiation, functions, and phenotypes of immune cells within the tumor. Tumor-associated microbiota also undergoes metabolic adaptation to survive within the tumor environment, leading to immune evasion, accumulation of mutations, and impairment of immune cells. Finally, we conclude with a discussion on the interplay between gut microbiota, immunometabolism, and CRC, highlighting a complex interaction that influences cancer development, progression, and cancer therapy efficacy.
Collapse
Affiliation(s)
- Madison Flory
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Paloma Bravo
- Department of Biology, Carleton College, Northfield, MN, USA
| | - Ashfaqul Alam
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
39
|
Wu YJ, Xiong JF, Zhan CN, Xu H. Gut microbiota alterations in colorectal adenoma-carcinoma sequence based on 16S rRNA gene sequencing: A systematic review and meta-analysis. Microb Pathog 2024; 195:106889. [PMID: 39197689 DOI: 10.1016/j.micpath.2024.106889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/12/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Most sporadic colorectal cancers (CRC) develop through the adenoma-carcinoma sequence. While dysbiosis of the intestinal flora contributes to CRC's pathogenesis, precise microbial taxa closely associated with the colorectal adenoma-carcinoma sequence remain elusive. This meta-analysis aimed to summarize the features of intestinal flora in patients with AD and CRC. METHODS PubMed, Embase, Cochrane Library, and Web of Science were searched for case-control studies comparing the relative abundance of gut microbiota in the feces of patients with AD, CRC, and healthy controls (HC) from inception to January 2024. The weighted mean difference (WMD) with a 95 % confidence interval (CI) was used to display the results. The Newcastle-Ottawa Scale (NOS) was used to assess the quality of the entailed literature. Publication bias was evaluated with the Egger's and Begg's tests. RESULTS Eleven studies were included, involving 477 CRC patients, 628 AD patients, and 864 healthy controls. Compared with HC, the patients with AD had a significantly lower Chao 1 index (WMD = -30.17, 95 % CI [-41.10, -19.23], P < 0.001) and Shannon index (WMD = -0.11 95 % CI [-0.18, -0.04], P = 0.002). Compared with AD, the CRC patients had a significantly higher Chao1 index (WMD = 22.09, 95 % CI [7.59, 36.00], P = 0.003) and Shannon index (WMD = 0.08, 95 % CI [0.00, 0.15], P = 0.037). Enterobacteriaceae (WMD = 0.03 95 % CI [0.00,0.05], P = 0.047; WMD = 0.02 95 % CI [0.00,0.04], P = 0.027) significantly increased in the order of Control-AD-CRC, while that of Blautia (WMD = -0.00 95 % CI [-0.01, -0.00], P = 0.001; WMD = -0.00 95 % CI [-0.00, -0.00], P = 0.002) was reduced. Compared with HC, the relative abundance of Proteobacteria (WMD = 0.05 95 % CI [0.03,0.07], P < 0.001), Fusobacteria (WMD = 0.02 95 % CI [0.00,0.03], P = 0.042), Streptococcaceae (WMD = 0.03 95 % CI [0.01,0.05], P = 0.017), Prevotellaceae (WMD = 0.02 95 % CI [0.00,0.04], P = 0.040), and Escherichia-Shigella (WMD = 0.06 95 % CI [0.01, 0.11], P = 0.021) was enriched in the CRC group. The relative abundance of Alistipes (WMD = 0.00 95 % CI [0.00,0.01], P = 0.032) and Streptococcus (WMD = 0.00 95 % CI [0.00,0.00], P = 0.001) was increased in the AD vs HC. The relative abundance of Firmicutes (WMD = -0.07 95 % CI [-0.12, -0.03], P = 0.003), Bifidobacteria (WMD = -0.03 95 % CI [-0.05, -0.01], P = 0.016), and Klebsiella (WMD = -0.01 95 % CI [-0.01, -0.00], P = 0.001) was decreased in the CRC vs HC. Compared with AD, the relative abundance of Firmicutes (WMD = -0.04 95 % CI [-0.07, -0.02], P = 0.002), Peptostreptococcaceae (WMD = -0.03 95 % CI [-0.05, -0.00], P = 0.021), Lachnospiraceae (WMD = -0.04 95 % CI [-0.08,-0.00], P = 0.037), Ruminococcaceae (WMD = -0.06 95 % CI [-0.09,-0.03], P < 0.001), Faecalibacterium (WMD = -0.01 95 % CI [-0.02, -0.01], P = 0.001), and Lachnoclostridium (WMD = -0.02 95 % CI [-0.03, -0.00], P = 0.040) was decreased in the CRC group, while Proteobacteria (WMD = 0.04 95 % CI [0.02,0.05], P < 0.001) was increased. CONCLUSIONS The dysbiosis characterized by reduced levels of short-chain fatty acid (SCFA)-producing bacteria, decreased anti-inflammatory bacteria, increased pro-inflammatory bacteria, and an elevation of bacteria with cytotoxic effects damaging to DNA may represent the specific microbial signature of colorectal adenoma/carcinoma. Further research is required to elucidate the mechanisms by which gut dysbiosis leads to the progression from AD to CRC and to explore the potential of specific microbiota markers in clinical treatment and non-invasive screening.
Collapse
Affiliation(s)
- Yi-Jun Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing-Fang Xiong
- Department of Geriatrics, Hangzhou Red Cross Hospital, Hangzhou, China
| | - Cheng-Nan Zhan
- Medical Service Community, Hangzhou Xiaoshan Hospital of TCM, Hangzhou, China
| | - Hong Xu
- Department of Gastroenterology and Hepatology, Hangzhou Red Cross Hospital, Hangzhou, China.
| |
Collapse
|
40
|
Arrè V, Balestra F, Scialpi R, Dituri F, Donghia R, Coletta S, Stabile D, Bianco A, Vincenti L, Fedele S, Shen C, Pettinato G, Scavo MP, Giannelli G, Negro R. Inorganic Polyphosphate Promotes Colorectal Cancer Growth via TRPM8 Receptor Signaling Pathway. Cancers (Basel) 2024; 16:3326. [PMID: 39409946 PMCID: PMC11476407 DOI: 10.3390/cancers16193326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is characterized by a pro-inflammatory microenvironment and features high-energy-supply molecules that assure tumor growth. A still less studied macromolecule is inorganic polyphosphate (iPolyP), a high-energy linear polymer that is ubiquitous in all forms of life. Made up of hundreds of repeated orthophosphate units, iPolyP is essential for a wide variety of functions in mammalian cells, including the regulation of proliferative signaling pathways. Some evidence has suggested its involvement in carcinogenesis, although more studies need to be pursued. Moreover, iPolyP regulates several homeostatic processes in animals, spanning from energy metabolism to blood coagulation and tissue regeneration. RESULTS In this study, we tested the role of iPolyP on CRC proliferation, using in vitro and ex vivo approaches, in order to evaluate its effect on tumor growth. We found that iPolyP is significantly increased in tumor tissues, derived from affected individuals enrolled in this study, compared to the corresponding peritumoral counterparts. In addition, iPolyP signaling occurs through the TRPM8 receptor, a well-characterized Na+ and Ca2+ ion channel often overexpressed in CRC and linked with poor prognosis, thus promoting CRC cell proliferation. The pharmacological inhibition of TRPM8 or RNA interference experiments performed in established CRC cell lines, such as Caco-2 and SW620, showed that the involvement of TRPM8 is essential, greater than that of the other two known iPolyP receptors, P2Y1 and RAGE. The presence of iPolyP drives cancer cells towards the mitotic phase of the cell cycle by enhancing the expression of ccnb1, which encodes the Cyclin B protein. In vitro 2D and 3D data reflected the ex vivo results, obtained by the generation of CRC-derived organoids, which increased in size. CONCLUSIONS These results indicate that iPolyP may be considered a novel and unexpected early biomarker supporting colorectal cancer cell proliferation.
Collapse
Affiliation(s)
- Valentina Arrè
- Personalized Medicine Laboratory, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (F.B.); (R.S.); (F.D.); (M.P.S.)
| | - Francesco Balestra
- Personalized Medicine Laboratory, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (F.B.); (R.S.); (F.D.); (M.P.S.)
| | - Rosanna Scialpi
- Personalized Medicine Laboratory, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (F.B.); (R.S.); (F.D.); (M.P.S.)
| | - Francesco Dituri
- Personalized Medicine Laboratory, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (F.B.); (R.S.); (F.D.); (M.P.S.)
| | - Rossella Donghia
- Data Science, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy;
| | - Sergio Coletta
- Core Facility Biobank, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (S.C.); (D.S.); (A.B.)
| | - Dolores Stabile
- Core Facility Biobank, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (S.C.); (D.S.); (A.B.)
| | - Antonia Bianco
- Core Facility Biobank, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (S.C.); (D.S.); (A.B.)
| | - Leonardo Vincenti
- Unit of Surgery, Department of Surgery Sciences, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (L.V.); (S.F.)
| | - Salvatore Fedele
- Unit of Surgery, Department of Surgery Sciences, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (L.V.); (S.F.)
| | - Chen Shen
- Division of Infectious Diseases, Washington University School in Medicine in St. Louis, 660 S Euclid Ave., St. Louis, MO 63110, USA;
| | - Giuseppe Pettinato
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA;
| | - Maria Principia Scavo
- Personalized Medicine Laboratory, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (F.B.); (R.S.); (F.D.); (M.P.S.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy;
| | - Roberto Negro
- Personalized Medicine Laboratory, National Institute of Gastroenterology “S. de Bellis”, IRCCS Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (F.B.); (R.S.); (F.D.); (M.P.S.)
| |
Collapse
|
41
|
Long J, Wang J, Xiao C, You F, Jiang Y, Li X. Intratumoral microbiota in colorectal cancer: focus on specific distribution and potential mechanisms. Cell Commun Signal 2024; 22:455. [PMID: 39327582 PMCID: PMC11426098 DOI: 10.1186/s12964-024-01831-3] [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: 06/04/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal malignant tumors globally, posing significant health risks and societal burdens. Recently, advancements in next-generation sequencing technology have identified CRC intratumoral microbiota, thereby opening up novel avenues for further research. This review synthesizes the current advancements in CRC intratumoral microbiota and their impact on CRC progression and discusses the disparities in the relative abundance and community composition of CRC intratumoral microbiota across various colorectal tumors based on their anatomical location and molecular subtypes, as well as the tumor stages, and spatial tumor distribution. Intratumoral microbiota predominantly influence CRC development by modulating colonic epithelial cells, tumor cells, and the tumor microenvironment. Mechanistically, they can cause DNA damage, apoptosis and epithelial-mesenchymal transition. The effects of different intratumoral microbiota on CRC have been shown to be two-fold. In the future, to address the limitations of existing studies, it is important to develop comprehensive experimental protocols and suitable in vitro models for elucidating more mechanisms of intratumoral microbiota on CRC, which will facilitate the clinical application of microbe-related therapeutic strategies in CRC and potentially other tumors.
Collapse
Affiliation(s)
- Jing Long
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
| | - Jiamei Wang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
| | - Chong Xiao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
- Oncology Teaching and Research Department, Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
- Institute of Oncology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China
| | - Yifang Jiang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China.
| | - Xueke Li
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China.
- Oncology Teaching and Research Department, Chengdu University of Traditional Chinese Medicine, Chengdu, 610036, China.
| |
Collapse
|
42
|
Tîrziu AT, Susan M, Susan R, Sonia T, Harich OO, Tudora A, Varga NI, Tiberiu-Liviu D, Avram CR, Boru C, Munteanu M, Horhat FG. From Gut to Eye: Exploring the Role of Microbiome Imbalance in Ocular Diseases. J Clin Med 2024; 13:5611. [PMID: 39337098 PMCID: PMC11432523 DOI: 10.3390/jcm13185611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The gut microbiome plays a crucial role in human health, and recent research has highlighted its potential impact on ocular health through the gut-eye axis. Dysbiosis, or an imbalance in the gut microbiota, has been implicated in various ocular diseases. Methods: A comprehensive literature search was conducted using relevant keywords in major electronic databases, prioritizing recent peer-reviewed articles published in English. Results: The gut microbiota influences ocular health through immune modulation, maintenance of the blood-retinal barrier, and production of beneficial metabolites. Dysbiosis can disrupt these mechanisms, contributing to ocular inflammation, tissue damage, and disease progression in conditions such as uveitis, age-related macular degeneration, diabetic retinopathy, dry eye disease, and glaucoma. Therapeutic modulation of the gut microbiome through probiotics, prebiotics, synbiotics, and fecal microbiota transplantation shows promise in preclinical and preliminary human studies. Conclusions: The gut-eye axis represents a dynamic and complex interplay between the gut microbiome and ocular health. Targeting the gut microbiome through innovative therapeutic strategies holds potential for improving the prevention and management of various ocular diseases.
Collapse
Affiliation(s)
- Andreea-Talida Tîrziu
- Department of General Medicine, Doctoral School, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Ophthalmology, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Monica Susan
- Centre for Preventive Medicine, Department of Internal Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Razvan Susan
- Centre for Preventive Medicine, Department of Family Medicine, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Tanasescu Sonia
- Department of Pediatrics, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Octavia Oana Harich
- Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Adelina Tudora
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, Strada Liviu Rebreanu 86, 310419 Arad, Romania
| | - Norberth-Istvan Varga
- Department of General Medicine, Doctoral School, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Dragomir Tiberiu-Liviu
- Medical Semiology II Discipline, Internal Medicine Department, "Victor Babes" University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Cecilia Roberta Avram
- Department of Residential Training and Post-University Courses, "Vasile Goldis" Western University, 310414 Arad, Romania
| | - Casiana Boru
- Department of Medicine, "Vasile Goldis" University of Medicine and Pharmacy, 310414 Arad, Romania
| | - Mihnea Munteanu
- Department of Ophthalmology, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Florin George Horhat
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
43
|
Zrelli M, Ferjani A, Nouira M, Hammami S, Ghithia N, Mouelhi L, Debbeche R, Raoult D, Boutiba Ben Boubaker I. Diversity in gut microbiota among colorectal cancer patients: findings from a case-control study conducted at a Tunisian University Hospital. Discov Oncol 2024; 15:402. [PMID: 39225843 PMCID: PMC11372012 DOI: 10.1007/s12672-024-01232-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
PURPOSE Globally, colorectal cancer (CRC) is among the most prevalent cancers. One distinctive feature of colorectal cancer is its close relationship to the gut microbiota, which is a crucial component of the tumor microenvironment. Over the last ten years, research has demonstrated that colorectal cancer is accompanied with dysbiosis of gut bacteria, fungi, viruses, and Archaea, and that these alterations may be causal. OBJECTIVES This study aimed to evaluate the disruption of the microorganism composition in the intestine, especially bacteria and to determine their relationship with colorectal cancer. METHODS An evaluation system for determining colorectal cancer (CRC) risk and prognosis can be established more easily with the help of accurate gut microbiota profiling. Stool samples from 14 CRC patients and 13 controls were collected and the flora relative abundance was measured using targeted quantitative PCR (qPCR) assays to evaluate diagnostic potential of selected biomarkers: Streptococcus gallolyticus and Enterococcus faecalis. Culture and MALDI-TOF mass spectrometry were coupled to identify the gut microbiota in both colorectal cancer and control groups. RESULTS Compared with controls, the gut microbiota of CRC patients showed an increase in the abundance of Enterococcus, Fusobacterium and Streptococcus. At the species level, the CRC enriched bacterium including Escherichia coli, Enterococcus faecalis, Fusobacterium nucleatum, Streptococcus gallolyticus, Flavoni fractorplautii and Eggerthella lenta acted as promising biomarkers for early detection of CRC. CONCLUSION This study highlights the potential of gut microbiota biomarkers as a promising non-invasive tool for the accurate detection and distinction of individuals with CRC.
Collapse
Affiliation(s)
- Mariem Zrelli
- Faculty of Medicine of Tunis, Research Laboratory ''Antimicrobial Resistance'' LR99ES09, University of Tunis El Manar, 1007, Tunis, Tunisia.
- Laboratory of Microbiology, Charles Nicolle Hospital, 1006, Tunis, Tunisia.
| | - Asma Ferjani
- Faculty of Medicine of Tunis, Research Laboratory ''Antimicrobial Resistance'' LR99ES09, University of Tunis El Manar, 1007, Tunis, Tunisia
- Laboratory of Microbiology, Charles Nicolle Hospital, 1006, Tunis, Tunisia
| | - Mariem Nouira
- Epidemiology Department, Charles Nicolle Hospital, Faculty of Medicine of Tunis, University of Tunis El Manar, 1006, Tunis, Tunisia
| | - Sirine Hammami
- Department of Gastroenterology, Charles Nicolle Hospital, 1006, Tunis, Tunisia
| | - Nadine Ghithia
- Department of Gastroenterology, Charles Nicolle Hospital, 1006, Tunis, Tunisia
| | - Leila Mouelhi
- Department of Gastroenterology, Charles Nicolle Hospital, 1006, Tunis, Tunisia
| | - Radhouane Debbeche
- Department of Gastroenterology, Charles Nicolle Hospital, 1006, Tunis, Tunisia
| | - Didier Raoult
- IRD, APHM, MEPHI, Aix Marseille Univ, 19-21 Boulevard Jean Moulin, 13005, Marseille, France.
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005, Marseille, France.
| | - Ilhem Boutiba Ben Boubaker
- Faculty of Medicine of Tunis, Research Laboratory ''Antimicrobial Resistance'' LR99ES09, University of Tunis El Manar, 1007, Tunis, Tunisia
- Laboratory of Microbiology, Charles Nicolle Hospital, 1006, Tunis, Tunisia
| |
Collapse
|
44
|
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
|
45
|
Parveen S, Alqahtani AS, Aljabri MY, Dawood T, Khan SS, Gupta B, Vempalli S, Hassan AAHAA, Elamin NMH. Exploring the Interplay: Oral–Gut Microbiome Connection and the Impact of Diet and Nutrition. EUROPEAN JOURNAL OF GENERAL DENTISTRY 2024; 13:165-176. [DOI: 10.1055/s-0044-1786154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
AbstractThe intricate interplay between the oral and intestinal microbiota holds increasing fascination within the context of health and nutrition. Serving as the gateway to the gastrointestinal tract, the oral microbiota hosts a diverse array of microbial species that significantly influence well-being or contribute to various diseases. Dysbiosis in the oral microbiota has been linked to conditions such as dental caries, periodontal diseases, and systemic disorders, including diabetes, cardiovascular disease, obesity, rheumatoid arthritis, Alzheimer's disease, and colorectal cancer. This review aims to comprehend the nuanced relationship between oral and intestinal microbiotas, exploring the pivotal role of diet in developing strategies for wellness promotion and disease prevention. Drawing insights from a myriad of studies encompassing both animals and humans, we examine the implications of microbial dysbiosis and its impact on health. A bibliographic search of 78 scientific articles was conducted across PubMed Central, Web of Science, Scopus, Google Scholar, and the Saudi digital library from January 2000 to August 2023. Following a rigorous screening process, the full texts of selected articles were critically reviewed to extract relevant information. Articles not meeting the inclusion criteria—specifically focused on oral–intestinal microbiota interaction and diet and nutrition—were meticulously excluded. Diet emerges as a key player in influencing both oral and intestinal microbiotas. Various dietary components, such as fiber, prebiotics, probiotics, and bioactive compounds, have demonstrated significant effects on the diversity and function of microorganisms in these ecosystems. Conversely, diets high in processed foods, added sugars, and saturated fats correlate with dysbiosis and an elevated risk of oral and gastrointestinal diseases. Understanding the intricacies of this interaction is paramount for the development of innovative approaches fostering a balanced oral–gut microbiota axis and improving overall human health. The implications extend to preventive and therapeutic interventions, emphasizing the practical importance of unraveling these complexities for public health and clinical practice. This comprehensive review delves into the intricate relationship between gut and oral microbiota, shedding light on their roles in various diseases, particularly focusing on oral diseases. Key findings are summarized, and implications for future research and clinical practice are discussed. In conclusion, the review underscores the urgent need for special attention to key microbiota in developing targeted interventions for promoting oral and gut health.
Collapse
Affiliation(s)
- Sameena Parveen
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Ahmed Shaher Alqahtani
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Mohammed Y. Aljabri
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Tazeen Dawood
- Department of Preventive Dental Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Samar Saeed Khan
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Bharti Gupta
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Swetha Vempalli
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | | | - Nahid Mahmoud Hassan Elamin
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
46
|
Parveen S, Alqahtani AS, Aljabri MY, Bajonaid A, Khan SS, Hassan AAHAA, Dawood T. Nationwide exploration: assessing oral microbiome knowledge among dental professionals in Saudi Arabia and its implications for oral health care. BMC Oral Health 2024; 24:1028. [PMID: 39217310 PMCID: PMC11366131 DOI: 10.1186/s12903-024-04770-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The relationship between the microbiome and oral health is intricate, yet there is a lack of comprehensive knowledge regarding the microbiome's impact on oral health. Integrating knowledge regarding the oral microbiome and its significance in oral and systemic diseases holds profound implications for dental professionals in patient care and professional development. This study assessed dental professionals' oral microbiome comprehension and knowledge levels in Saudi Arabia and its implications for oral healthcare. METHODS Data were gathered using a cross-sectional design by administering a comprehensive online questionnaire to 253 dental professionals from diverse demographic backgrounds. The questionnaire, administered in English, was divided into four sections: (1) Microbiome awareness and understanding, (2) Diet, nutrition, and microbiome relationship, (3) Microbiome and oral and systemic diseases, and (4) Counselling, education, and implications. Statistical analyses were used to identify and understand underlying patterns, including descriptive statistics, chi-squared tests, ANOVA, and post hoc tests. The Spearman rank correlation coefficient was applied to assess self-rated knowledge. RESULTS Of the 253 participants, 94.6% were familiar with the term "microbiome." Merely 13% of participants considered the oral microbiome to be the second most diverse, following the gut microbiome. About 39.9% of participants knew the connection between oral mucosal diseases and the oral microbiome. Furthermore, only 6.7% thought there was a connection between systemic diseases and the oral microbiome. Participant comprehension of oral microbiome questions averaged 9.19 out of 13, with 83.7% scoring "good". There were significant differences in knowledge scores among dental specializations (F = 7.082, P < 0.001) and years of professional experience (F = 4.755, P = 0.003). Significantly, 53.8% of participants had uncertain self-perceptions of their knowledge of the oral microbiome, while only 0.8% rated their understanding as 'very good'. CONCLUSION Our findings reveal that dental professionals have varying levels of awareness and comprehension of the oral microbiome. Despite widespread awareness, understanding its diversity and implications for oral and systemic health remains limited. It is essential to address these gaps in knowledge through future research and educational interventions, considering the vital part that dental professionals play in promoting oral health through personalised dietary recommendations, lifestyle changes, and hygiene practices. These initiatives may promote a robust oral microbial community, enhance patient outcomes, and advance oral healthcare locally and globally.
Collapse
Affiliation(s)
- Sameena Parveen
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | - Ahmed Shaher Alqahtani
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Mohammed Y Aljabri
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Amal Bajonaid
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Samar Saeed Khan
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | | | - Tazeen Dawood
- Department of Preventive Dental Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
47
|
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
|
48
|
Tanwar H, Gnanasekaran JM, Allison D, Chuang LS, He X, Aimetti M, Baima G, Costalonga M, Cross RK, Sears C, Mehandru S, Cho J, Colombel JF, Raufman JP, Thumbigere-Math V. Unravelling the Oral-Gut Axis: Interconnection Between Periodontitis and Inflammatory Bowel Disease, Current Challenges, and Future Perspective. J Crohns Colitis 2024; 18:1319-1341. [PMID: 38417137 PMCID: PMC11324343 DOI: 10.1093/ecco-jcc/jjae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/04/2023] [Accepted: 02/27/2024] [Indexed: 03/01/2024]
Abstract
As the opposite ends of the orodigestive tract, the oral cavity and the intestine share anatomical, microbial, and immunological ties that have bidirectional health implications. A growing body of evidence suggests an interconnection between oral pathologies and inflammatory bowel disease [IBD], implying a shift from the traditional concept of independent diseases to a complex, reciprocal cycle. This review outlines the evidence supporting an 'oral-gut' axis, marked by a higher prevalence of periodontitis and other oral conditions in IBD patients and vice versa. We present an in-depth examination of the interconnection between oral pathologies and IBD, highlighting the shared microbiological and immunological pathways, and proposing a 'multi-hit' hypothesis in the pathogenesis of periodontitis-mediated intestinal inflammation. Furthermore, the review underscores the critical need for a collaborative approach between dentists and gastroenterologists to provide holistic oral-systemic healthcare.
Collapse
Affiliation(s)
- Himanshi Tanwar
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | | | - Devon Allison
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Ling-shiang Chuang
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Mario Aimetti
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Giacomo Baima
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Massimo Costalonga
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Raymond K Cross
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cynthia Sears
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Saurabh Mehandru
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judy Cho
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vivek Thumbigere-Math
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| |
Collapse
|
49
|
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
|
50
|
Zhao Z, Zhang X, Zhao W, Wang J, Peng Y, Liu X, Liu N, Liu Q. Effect of chronic alcohol consumption on oral microbiota in rats with periodontitis. PeerJ 2024; 12:e17795. [PMID: 39148678 PMCID: PMC11326440 DOI: 10.7717/peerj.17795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Background The imbalance of oral microbiota can contribute to various oral disorders and potentially impact general health. Chronic alcohol consumption beyond a certain threshold has been implicated in influencing both the onset and progression of periodontitis. However, the mechanism by which chronic alcohol consumption affects periodontitis and its association with changes in the oral microbial community remains unclear. Objective This study used 16S rRNA gene amplicon sequencing to examine the dynamic changes in the oral microbial community of rats with periodontitis influenced by chronic alcohol consumption. Methods Twenty-four male Wistar rats were randomly allocated to either a periodontitis (P) or periodontitis + alcohol (PA) group. The PA group had unrestricted access to alcohol for 10 weeks, while the P group had access to water only. Four weeks later, both groups developed periodontitis. After 10 weeks, serum levels of alanine aminotransferase and aspartate aminotransferase in the rats' serum were measured. The oral swabs were obtained from rats, and 16S rRNA gene sequencing was conducted. Alveolar bone status was assessed using hematoxylin and eosin staining and micro-computed tomography. Results Rats in the PA group exhibited more severe periodontal tissue damage compared to those in the periodontitis group. Although oral microbial diversity remained stable, the relative abundance of certain microbial communities differed significantly between the two groups. Actinobacteriota and Desulfobacterota were more prevalent at the phylum level in the PA group. At the genus level, Cutibacterium, Tissierella, Romboutsia, Actinomyces, Lawsonella, Anaerococcus, and Clostridium_sensu_stricto_1 were significantly more abundant in the PA group, while Haemophilus was significantly less abundant. Additionally, functional prediction using Tax4Fun revealed a significant enrichment of carbohydrate metabolism in the PA group. Conclusion Chronic alcohol consumption exacerbated periodontitis in rats and influenced the composition and functional characteristics of their oral microbiota, as indicated by 16S rRNA gene sequencing results. These microbial alterations may contribute to the exacerbation of periodontitis in rats due to chronic alcohol consumption.
Collapse
Affiliation(s)
- Zirui Zhao
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiao Zhang
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wanqing Zhao
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianing Wang
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanhui Peng
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xuanning Liu
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Na Liu
- Department of Preventive Dentistry, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qing Liu
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|