1
|
Chen F, Guo S, Li Y, Lu Y, Liu L, Chen S, An J, Zhang G. Fusobacterium nucleatum-driven CX3CR1 + PD-L1 + phagocytes route to tumor tissues and reshape tumor microenvironment. Gut Microbes 2025; 17:2442037. [PMID: 39710592 DOI: 10.1080/19490976.2024.2442037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
The intracellular bacterium Fusobacterium nucleatum (Fn) mediates tumorigenesis and progression in colorectal cancer (CRC). However, the origin of intratumoral Fn and the role of Fn-infected immunocytes in the tumor microenvironment remain unclear. Here, we observed that Fn-infected neutrophils/macrophages (PMNs/MΦs), especially PMNs, accumulate in tumor tissues and fecal Fn abundance correlates positively with an abundance of blood PD-L1+ PMNs in CRC patients. Moreover, Fn accumulates in tumor tissues of tumor-bearing mice via intragingival infection and intravenous injection. Mechanistically, Fn can survive inside PMNs by reducing intracellular ROS levels and producing H2S. Specifically, the lysozyme inhibitor Fn1792 as a novel virulence factor of Fn suppressed apoptosis of phagocytes by inducing CX3CR1 expression. Furthermore, Fn-driven CX3CR1+PD-L1+ phagocytes transfer intracellular Fn to tumor cells, which recruit PMNs/MΦs through the CXCL2/8-CXCR2 and CCL5/CCR5 axes. Consequently, CX3CR1+PD-L1+ PMNs infiltration promotes CRC metastasis and weakens the efficacy of immunotherapy. Treatment with the doxycycline eradicated intracellular Fn, thereby reducing the CX3CR1+PD-L1+ PMNs populations and slowing Fn-promoted tumor growth and metastasis in mice. These results suggest phagocytes as Fn-presenting cells use mutualistic strategies to home to tumor tissues and induce immunosuppression, and treatment with ROS-enhanced antibiotics can inhibit Fn-positive tumor progression.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Songhe Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yiqiu Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yongfan Lu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Le Liu
- Department of Gastroenterology, Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Shengxin Chen
- Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Jun An
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Zepeda-Rivera MA, Eisele Y, Baryiames A, Wu H, Mengoni C, Piccinno G, McMahon EF, LaCourse KD, Jones DS, Hauner H, Minot SS, Segata N, Dewhirst FE, Johnston CD, Bullman S. Fusobacterium sphaericum sp. nov., isolated from a human colon tumor adheres to colonic epithelial cells and induces IL-8 secretion. Gut Microbes 2025; 17:2442522. [PMID: 39722539 DOI: 10.1080/19490976.2024.2442522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/20/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Cancerous tissue is a largely unexplored microbial niche that provides a unique environment for the colonization and growth of specific bacterial communities, and with it, the opportunity to identify novel bacterial species. Here, we report distinct features of a novel Fusobacterium species, F. sphaericum sp. nov. (Fs), isolated from primary colon adenocarcinoma tissue. We acquire the complete closed genome and associated methylome of this organism and phylogenetically confirm its classification into the Fusobacterium genus, with F. perfoetens as its closest neighbor. Fs is phenotypically and genetically distinct, with morphological analysis revealing its coccoid shape, that while similar to F. perfoetens is rare for most Fusobacterium members. Fs displays a metabolic profile and antibiotic resistance repertoire consistent with other Fusobacterium species. In vitro, Fs has adherent and immunomodulatory capabilities, as it intimately associates with human colon cancer epithelial cells and promotes IL-8 secretion. An analysis of the prevalence and abundance of Fs in > 20,000 human metagenomic samples shows that it is a rarely detected member within human stool with variable relative abundance, found in both healthy controls and patients with colorectal cancer (CRC). Our study sheds light on a novel bacterial species isolated directly from the human CRC tumor niche and given its in vitro interaction with cancer epithelial cells suggests that its role in human health and disease warrants further investigation.
Collapse
Affiliation(s)
- Martha A Zepeda-Rivera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, WA, USA
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yannick Eisele
- School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Nutritional Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | | | - Hanrui Wu
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Claudia Mengoni
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Gianmarco Piccinno
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Elsa F McMahon
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, WA, USA
| | | | - Dakota S Jones
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, WA, USA
| | - Hans Hauner
- Institute of Nutritional Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Samuel S Minot
- Data Core, Shared Resources, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Nicola Segata
- Department of Computational, Cellular and Integrative Biology, University of Trento, Trento, Italy
| | - Floyd E Dewhirst
- Department of Microbiology, ADA Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Christopher D Johnston
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, WA, USA
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Susan Bullman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunology, James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
3
|
Wang H, Han J, Zhang XA. Interplay of m6A RNA methylation and gut microbiota in modulating gut injury. Gut Microbes 2025; 17:2467213. [PMID: 39960310 PMCID: PMC11834532 DOI: 10.1080/19490976.2025.2467213] [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: 10/12/2024] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
The gut microbiota undergoes continuous variations among individuals and across their lifespan, shaped by diverse factors encompassing diet, age, lifestyle choices, medication intake, and disease states. These microbial inhabitants play a pivotal role in orchestrating physiological metabolic pathways through the production of metabolites like bile acids, choline, short-chain fatty acids, and neurotransmitters, thereby establishing a dynamic "gut-organ axis" with the host. The intricate interplay between the gut microbiota and the host is indispensable for gut health, and RNA N6-methyladenosine modification, a pivotal epigenetic mark on RNA, emerges as a key player in this process. M6A modification, the most prevalent internal modification of eukaryotic RNA, has garnered significant attention in the realm of RNA epigenetics. Recent findings underscore its potential to influence gut microbiota diversity and intestinal barrier function by modulating host gene expression patterns. Conversely, the gut microbiota, through its impact on the epigenetic landscape of host cells, may indirectly regulate the recruitment and activity of RNA m6A-modifying enzymes. This review endeavors to delve into the biological functions of m6A modification and its consequences on intestinal injury and disease pathogenesis, elucidating the partial possible mechanisms by which the gut microbiota and its metabolites maintain host intestinal health and homeostasis. Furthermore, it also explores the intricate crosstalk between them in intestinal injury, offering a novel perspective that deepens our understanding of the mechanisms underlying intestinal diseases.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
4
|
Moskal K, Khurana N, Siegert L, Lee YS, Clevers H, Elinav E, Puschhof J. Modeling cancer-microbiome interactions in vitro: A guide to co-culture platforms. Int J Cancer 2025; 156:2053-2067. [PMID: 39716471 PMCID: PMC11970552 DOI: 10.1002/ijc.35298] [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: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024]
Abstract
The biology of cancer is characterized by an intricate interplay of cells originating not only from the tumor mass, but also its surrounding environment. Different microbial species have been suggested to be enriched in tumors and the impacts of these on tumor phenotypes is subject to intensive investigation. For these efforts, model systems that accurately reflect human-microbe interactions are rapidly gaining importance. Here we present a guide for selecting a suitable in vitro co-culture platform used to model different cancer-microbiome interactions. Our discussion spans a variety of in vitro models, including 2D cultures, tumor spheroids, organoids, and organ-on-a-chip platforms, where we delineate their respective advantages, limitations, and applicability in cancer microbiome research. Particular focus is placed on methodologies that facilitate the exposure of cancer cells to microbes, such as organoid microinjections and co-culture on microfluidic devices. We highlight studies offering critical insights into possible cancer-microbe interactions and underscore the importance of in vitro models in those discoveries. We anticipate the integration of more complex microbial communities and the inclusion of immune cells into co-culture systems to more accurately simulate the tumor microenvironment. The advent of ever more sophisticated co-culture models will aid in unraveling the mechanisms of cancer-microbiome interplay and contribute to exploiting their potential in novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Kamil Moskal
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| | - Nimisha Khurana
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Luisa Siegert
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
| | - Ye Seul Lee
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Hans Clevers
- Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC UtrechtHubrecht InstituteUtrechtThe Netherlands
- Present address:
Roche Pharmaceutical Research and Early DevelopmentBaselSwitzerland
| | - Eran Elinav
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Systems Immunology DepartmentWeizmann Institute of ScienceRehovotIsrael
| | - Jens Puschhof
- Junior Research Group Epithelium Microbiome Interactions (EMIL), German Cancer Research CenterHeidelbergGermany
- Microbiome and Cancer Division, German Cancer Research CenterHeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
- DKFZ Hector Cancer Institute at the University Medical CenterMannheimGermany
| |
Collapse
|
5
|
Elkrief A, Routy B, Derosa L, Bolte L, Wargo JA, McQuade JL, Zitvogel L. Gut Microbiota in Immuno-Oncology: A Practical Guide for Medical Oncologists With a Focus on Antibiotics Stewardship. Am Soc Clin Oncol Educ Book 2025; 45:e472902. [PMID: 40262063 DOI: 10.1200/edbk-25-472902] [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: 04/24/2025]
Abstract
The gut microbiota has emerged as a critical determinant of immune checkpoint inhibitor (ICI) efficacy, resistance, and toxicity. Retrospective and prospective studies profiling the taxonomic composition of intestinal microbes of patients treated with ICI have revealed specific gut microbial signatures associated with response. By contrast, dysbiosis, which can be caused by chronic inflammatory processes (such as cancer) or comedications, is a risk factor of resistance to ICI. Recent large-scale meta-analyses have confirmed that antibiotic (ATB) use before or during ICI therapy alters the microbiota repertoire and significantly shortens overall survival, even after adjusting for prognostic factors. These results underscore the importance of implementing ATB stewardship recommendations in routine oncology practice. Microbiota-centered interventions are now being explored to treat gut dysbiosis and optimize ICI responses. Early-phase clinical trials evaluating fecal microbiota transplantation (FMT) from ICI responders or healthy donors have shown that this approach is safe and provided preliminary data on potential efficacy to overcome both primary and secondary resistance to ICI in melanoma, non-small cell lung cancer, and renal cell carcinoma. More targeted interventions including live bacterial products including Clostridium butyricum and Akkermansia massiliensis represent novel microbiome-based adjunct therapies. Likewise, dietary interventions, such as high-fiber diets, have shown promise in enhancing ICI activity. In this ASCO Educational Book, we summarize the current state-of-the-evidence of the clinical relevance of the intestinal microbiota in cancer immunotherapy and provide a practical guide for ATB stewardship.
Collapse
Affiliation(s)
- Arielle Elkrief
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Bertrand Routy
- University of Montreal Hospital Research Centre, Cancer Axis, Montreal, Canada
- University of Montreal Hospital Centre, Department of Hematology-Oncology, Montreal, Canada
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
| | - Laura Bolte
- Department of Medical Oncology, University Groningen and University Medical Center, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University Groningen and University Medical Center, Groningen, the Netherlands
| | | | | | - Laurence Zitvogel
- INSERM U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Gustave Roussy, ClinicObiome, Villejuif, France
- Université Paris-Saclay, Faculty of Medicine, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| |
Collapse
|
6
|
Wizenty J, Sigal M. Helicobacter pylori, microbiota and gastric cancer - principles of microorganism-driven carcinogenesis. Nat Rev Gastroenterol Hepatol 2025; 22:296-313. [PMID: 40011753 DOI: 10.1038/s41575-025-01042-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/28/2025]
Abstract
The demonstration that Helicobacter pylori is a pathogenic bacterium with marked carcinogenic potential has paved the way for new preventive approaches for gastric cancer. Although decades of research have uncovered complex interactions of H. pylori with epithelial cells, current insights have refined our view on H. pylori-associated carcinogenesis. Specifically, the cell-type-specific effects on gastric stem and progenitor cells deep in gastric glands provide a new view on the ability of the bacteria to colonize long-term, manipulate host responses and promote gastric pathology. Furthermore, new, large-scale epidemiological data have shed light on factors that determine why only a subset of carriers progress to gastric cancer. Currently, technological advances have brought yet another revelation: H. pylori is far from the only microorganism able to colonize the stomach. Instead, the stomach is colonized by a diverse gastric microbiota, and there is emerging evidence for the occurrence and pathological effect of dysbiosis resulting from an aberrant interplay between H. pylori and the gastric mucosa. With the weight of this evidence mounting, here we consider how the lessons learned from H. pylori research inform and synergize with this emerging field to bring a more comprehensive understanding of the role of microbes in gastric carcinogenesis.
Collapse
Affiliation(s)
- Jonas Wizenty
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy and BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Michael Sigal
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| |
Collapse
|
7
|
Andreu-Sánchez S, Blanco-Míguez A, Wang D, Golzato D, Manghi P, Heidrich V, Fackelmann G, Zhernakova DV, Kurilshikov A, Valles-Colomer M, Weersma RK, Zhernakova A, Fu J, Segata N. Global genetic structure of human gut microbiome species is related to geographic location and host health. Cell 2025:S0092-8674(25)00416-7. [PMID: 40311618 DOI: 10.1016/j.cell.2025.04.014] [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: 08/06/2024] [Revised: 01/23/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
The human gut harbors thousands of microbial species, each exhibiting significant inter-individual genetic variability. Although many studies have associated microbial relative abundances with human-health-related phenotypes, the substantial intraspecies genetic variability of gut microbes has not yet been comprehensively considered, limiting the potential of linking such genetic traits with host conditions. Here, we analyzed 32,152 metagenomes from 94 microbiome studies across the globe to investigate the human microbiome intraspecies genetic diversity. We reconstructed 583 species-specific phylogenies and linked them to geographic information and species' horizontal transmissibility. We identified 484 microbial-strain-level associations with 241 host phenotypes, encompassing human anthropometric factors, biochemical measurements, diseases, and lifestyle. We observed a higher prevalence of a Ruminococcus gnavus clade in nonagenarians correlated with distinct plasma bile acid profiles and a melanoma and prostate-cancer-associated Collinsella clade. Our large-scale intraspecies genetic analysis highlights the relevance of strain diversity as it relates to human health.
Collapse
Affiliation(s)
- Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Daoming Wang
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Davide Golzato
- Department of CIBIO, University of Trento, Trento, Italy
| | - Paolo Manghi
- Department of CIBIO, University of Trento, Trento, Italy
| | - Vitor Heidrich
- Department of CIBIO, University of Trento, Trento, Italy
| | | | - Daria V Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Mireia Valles-Colomer
- Department of CIBIO, University of Trento, Trento, Italy; MELIS Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands.
| | - Nicola Segata
- Department of CIBIO, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
8
|
Bhowmick K, Yang X, Mohammad T, Xiang X, Molmenti CL, Mishra B, Dasarathy S, Krainer AR, Hassan MI, Crandall KA, Mishra L. Microbial metabolite ammonia disrupts TGF-β signaling to promote colon cancer. J Biol Chem 2025:108559. [PMID: 40311681 DOI: 10.1016/j.jbc.2025.108559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Colorectal cancer (CRC) is rising alarmingly in younger populations, potentially arising from factors such as obesity, pro-inflammatory gut microbiome and the accumulation of toxic metabolites. However, how metabolites such as ammonia impact key signaling pathways to promote CRC remains unclear. Our study investigates a critical link between gut microbiome alterations, ammonia, and their toxic effects on the TGF-β signaling pathway, driving CRC progression. We observe altered microbial populations in an obesity-induced mouse model of cancer, where ammonia promotes caspase-3-mediated cleavage of the SMAD3 adaptor βII-spectrin (SPTBN1). Cleaved SPTBN1 fragments form adducts with ammonia that induce pro-inflammatory cytokine expression and disrupt TGF-β signaling. Extending from AlphaFold docking simulations, we identified that ammonia interacts with N-terminal SPTBN1 potentially through residues D81, Y556, S663, Y666, N986, and D1177 to form hydrogen bonds that disrupt downstream SMAD3 signaling, altering TGF-β signaling to a protumorigenic phenotype. Blocking SPTBN1, through an SPTBN1-specific siRNA blocks ammonia toxicity and restores normal SMAD3/TGF-β signaling by reducing the abundance of SPTBN1 cleaved fragments in SW480 and Caco-2 (CRC) cell lines. In addition, our research establishes crosstalk between TGF-β signaling and a microbial sensor, carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), which is significantly overexpressed in CRC patients. We identified CEACAM1-SPTBN1 interactions at specific residues (E517 and Y520) within the immunoreceptor tyrosine-based inhibitory motif (ITIM) of CEACAM1 cytoplasmic domain, identifying distinguishing a potential axis that is harnessed by the altered microbiome. Our study identifies mechanistic insights into how microbial metabolites target TGF-β as a major signaling pathway to promote CRC.
Collapse
Affiliation(s)
- Krishanu Bhowmick
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA.
| | - Xiaochun Yang
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Xiyan Xiang
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA
| | - Christine L Molmenti
- Department of Occupational Medicine, Epidemiology and Prevention, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Feinstein Institutes for Medical Research, Institute of Cancer Research, Manhasset, NY, USA; Department of Surgery, Northwell Health, Manhasset, NY, USA
| | - Bibhuti Mishra
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Department of Neurology, Hempstead, NY, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | | | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Keith A Crandall
- Computational Biology Institute and Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington DC, USA
| | - Lopa Mishra
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research; Divisions of Gastroenterology and Hepatology, Department of Medicine, Northwell Health, Manhasset, NY, USA; Cold Spring Harbor Laboratory; Cold Spring Harbor, NY, USA; Department of Surgery, George Washington University, Washington DC, USA.
| |
Collapse
|
9
|
Meyers AL, Dowty JG, Mahmood K, Macrae FA, Rosty C, Buchanan DD, Jenkins MA. Age-specific trends in colorectal, appendiceal, and anal tumour incidence by histological subtype in Australia from 1990 to 2020: a population-based time-series analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.21.25326138. [PMID: 40313308 PMCID: PMC12045447 DOI: 10.1101/2025.04.21.25326138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Background Early-onset bowel cancer incidence (age <50 years) has increased worldwide and is highest in Australia, but how this varies across histology and anatomical site remains unclear. We aimed to investigate appendiceal, proximal colon, distal colon, rectal, and anal cancer incidence trends by age and histology in Australia. Methods Cancer incidence rate data were obtained from all Australian cancer registries (1990-2020 period). Birth cohort-specific incidence rate ratios (IRRs) and annual percentage change in rates were estimated using age-period-cohort modelling and joinpoint regression. Findings After excluding neuroendocrine neoplasms, early-onset cancer incidence rose 5-9% annually, yielding 5,341 excess cases (2 per 100,000 person-years; 12% appendix, 45% colon, 36% rectum, 7% anus; 20-214% relative increase). Trends varied by site, period, and age: appendiceal cancer rose from 1990-2020 in 30-49-year-olds; colorectal cancers rose from around 1990-2010 in 20-29-year-olds and from 2010-2020 in 30-39-year-olds; anal cancer rose from 1990-2009 in 40-49-year-olds. Across all sites, IRRs increased with successive birth cohorts since 1960. Notably, adenocarcinoma incidence in the 1990s versus 1950s birth cohort was 2-3-fold for colorectum and 7-fold for appendix. The greatest subtype-specific increases occurred for appendiceal mucinous adenocarcinoma, colorectal non-mucinous adenocarcinoma, and anal squamous cell carcinoma. Only later-onset (age ≥50) colorectal and anal adenocarcinoma rates declined. Appendiceal tumours, neuroendocrine neoplasms (all sites), anorectal squamous cell carcinomas, and colon signet ring cell carcinomas rose across early-onset and later-onset strata. Interpretation Appendiceal, colorectal, and anal cancer incidence is rising in Australia with variation across age and histology, underscoring the need to identify factors driving these trends. Funding ALM is supported by an Australian Government Research Training Program Scholarship, Rowden White Scholarship, and WP Greene Scholarship. DDB is supported by a National Health and Medical Research Council of Australia (NHMRC) Investigator grant (GNT1194896), a University of Melbourne Dame Kate Campbell Fellowship, and by funding awarded to The Colon Cancer Family Registry (CCFR, www.coloncfr.org ) from the National Cancer Institute (NCI), National Institutes of Health (NIH) [award U01 CA167551]. MAJ is supported by an NHMRC Investigator grant (GNT1195099), a University of Melbourne Dame Kate Campbell Fellowship, and by funding awarded to the CCFR from NCI, NIH [award U01 CA167551].
Collapse
|
10
|
Bai H, Liu T, Wang H, Li Y, Wang Z. Regulating Effects of Three Polysaccharides on Gut Microbiota in Felines and Canines: Insights from In Vitro Digestion, Fecal Fermentation, and Lactic Acid Bacterial Fermentation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9618-9636. [PMID: 40211571 DOI: 10.1021/acs.jafc.4c12847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
This study evaluated the effects of laminaria polysaccharide (FU), aloe polysaccharide (AL), and yeast β-glucan (YG) on in vitro digestion, fecal fermentation, and lactic acid bacterial fermentation in felines and canines. FU (Mw 27 713 g/mol), AL (44 425 g/mol), and YG (29 296 g/mol) were primarily composed of glucose, galacturonic acid, and glucuronic acid. The FU strongly promoted the fermentation of Bifidobacterium, Lactobacillus, and Pediococcus. The AL slightly supported Lactobacillus. The YG showed minimal activity. Molecular weights decreased after digestion with negligible monosaccharide release. The FU and AL were almost completely metabolized during fecal fermentation, while YG had ∼50% utilization. The FU produced the highest lactate concentration. The YG significantly increased SCFA levels, especially in canines, surpassing fructooligosaccharides. The FU:AL:YG mixture (0.8:0.2:1, FAY) achieved the best overall effects. 16S rRNA analysis revealed the enrichment of SCFA-producing genera (e.g., Megamonas, Parabacteroides) and suppression of odor-related bacteria (e.g., Fusobacterium, Desulfovibrio). Metabolomics showed reduced levels of odor compounds (e.g., 2-oxindole, 5-hydroxyindole) and increased secondary bile acids (e.g., deoxycholic acid, glycocholic acid). The FU, AL, and FAY upregulated bile secretion, neuroactive pathways, and arachidonic acid metabolism, while YG activated the ABC transporter pathway.
Collapse
Affiliation(s)
- Huasong Bai
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| | - Tong Liu
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| | - Hengyan Wang
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| | - Yunliang Li
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, PR China
| | - Zhanzhong Wang
- Nourse Science Centre for Pet Nutrition, Wuhu 241200, PR China
| |
Collapse
|
11
|
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
|
12
|
Cho MY, Eom JH, Choi EM, Yang SJ, Lee D, Kim YY, Kim HS, Hwang I. Recent advances in therapeutic probiotics: insights from human trials. Clin Microbiol Rev 2025:e0024024. [PMID: 40261032 DOI: 10.1128/cmr.00240-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
SUMMARYRecent advances in therapeutic probiotics have shown promising results across various health conditions, reflecting a growing understanding of the human microbiome's role in health and disease. However, comprehensive reviews integrating the diverse therapeutic effects of probiotics in human subjects have been limited. By analyzing randomized controlled trials (RCTs) and meta-analyses, this review provides a comprehensive overview of key developments in probiotic interventions targeting gut, liver, skin, vaginal, mental, and oral health. Emerging evidence supports the efficacy of specific probiotic strains and combinations in treating a wide range of disorders, from gastrointestinal (GI) and liver diseases to dermatological conditions, bacterial vaginosis, mental disorders, and oral diseases. We discuss the expanding understanding of microbiome-organ connections underlying probiotic mechanisms of action. While many clinical trials demonstrate significant benefits, we acknowledge areas requiring further large-scale studies to establish definitive efficacy and optimal treatment protocols. The review addresses challenges in standardizing probiotic research methodologies and emphasizes the importance of considering individual variations in microbiome composition and host genetics. Additionally, we explore emerging concepts such as the oral-gut-brain axis and future directions, including high-resolution microbiome profiling, host-microbe interaction studies, organoid models, and artificial intelligence applications in probiotic research. Overall, this review offers a comprehensive update on the current state of therapeutic probiotics across multiple domains of human health, providing insights into future directions and the potential for probiotics to revolutionize preventive and therapeutic medicine.
Collapse
Affiliation(s)
- Mu-Yeol Cho
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | - Je-Hyun Eom
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | - Eun-Mi Choi
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| | | | - Dahye Lee
- Department of Orthodontics, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Young Youn Kim
- Department of Oral and Maxillofacial Surgery, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Hye-Sung Kim
- Department of Oral and Maxillofacial Surgery, Apple Tree Dental Hospital, Goyang-si, South Korea
| | - Inseong Hwang
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, South Korea
| |
Collapse
|
13
|
Yu J, Liang Y, Zhang Q, Ding H, Xie M, Zhang J, Hu W, Xu S, Xiao Y, Xu S, Na R, Wu B, Zhou J, Chen H. An interplay between human genetics and intratumoral microbiota in the progression of colorectal cancer. Cell Host Microbe 2025:S1931-3128(25)00136-2. [PMID: 40306270 DOI: 10.1016/j.chom.2025.04.003] [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: 09/25/2024] [Revised: 01/10/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Intratumoral microbiota plays a crucial role in cancer progression. However, the relationship between host genetics and intratumoral microbiota, as well as their interaction in colorectal cancer (CRC) progression, remains unclear. With 748 Chinese CRC patients enrolled from three cohorts, we find that the single nucleotide polymorphism (SNP) rs2355016, located in the intron of ATP-sensitive inward rectifier potassium channel 11 (KCNJ11), is significantly associated with the abundance of Fusobacterium. Compared with the rs2355016 GG genotype, patients carrying the A allele exhibit downregulation of KCNJ11 and enrichment of Fusobacterium, which corresponds to accelerated proliferation and progression. Low expression of KCNJ11 can increase the level of galactose-N-acetyl-d-galactosamine (Gal-GalNAc) on the surface of CRC cells, thereby facilitating the binding of the Fap2 protein from F. nucleatum to Gal-GalNAc. This further enhances the adhesion and invasion of F. nucleatum and promotes CRC growth. Our study explores the interaction between intratumoral microbiota and SNPs in CRC patients, which will enhance our understanding of CRC proliferation.
Collapse
Affiliation(s)
- Jing Yu
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Yuxuan Liang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Qingrong Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Hui Ding
- Department of General Surgery, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510655, China
| | - Minghao Xie
- Department of General Surgery, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330038, China
| | - Jingjing Zhang
- Cancer Hospital Chinese Academy of Medical Sciences Shenzhen Center, Shenzhen, Guangdong 518116, China
| | - Wenyan Hu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Sihua Xu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yiyuan Xiao
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Sha Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Rong Na
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China.
| | - Baixing Wu
- Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou 510530, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Jiaming Zhou
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Haitao Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510275, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
14
|
Di Pierro F, Sagheddu V, Galletti S, Casaroli A, Labrini E, Soldi S, Cazzaniga M, Bertuccioli A, Matera M, Cavecchia I, Palazzi CM, Tanda ML, Zerbinati N. Selection, Comparative Genomics, and Potential Probiotic Features of Escherichia coli 5C, a pks-Negative Strain Isolated from Healthy Infant Donor Feces. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10522-5. [PMID: 40238037 DOI: 10.1007/s12602-025-10522-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/18/2025]
Abstract
Among the emerging issues in probiotic safety, the possible presence of pks, a gene cluster synthetizing a genotoxin known as colibactin, is one of the most alarming. Indeed, indigenous E. coli strain pks-positive are found in 60% of patients with colorectal cancer, and the most widely used E. coli-based probiotic, known as E. coli Nissle 1917 (DSM 6601), is pks-positive. Starting from 25 potential candidates selected by screening 25 infant stool samples, we have selected an E. coli strain (named 5C, deposited as LMG S-33222) belonging to the phylotype A and having the serovar O173:H1. Having been previously completely sequenced by our group, we have further characterized this strain, demonstrating that it is (i) devoid of the most known potential pathogenic-related genes, (ii) devoid of possible plasmids, (iii) antibiotic-sensitive according to the EFSA panel, (iv) resistant in gastric and enteric juice, (v) significantly producing acetate, (vi) poorly producing histamine, (vii) endowed with a significant in vitro antipathogenic profile, (viii) promoting a significant in vitro immunological response based on IL-10 and IL-12, and (ix) devoid of the pks genes. A comparative genomics versus E. coli Nissle 1917 is also provided. Considering that the other two most commonly used E. coli-based probiotics (E. coli DSM 17252 and E. coli A0 34/86) are respectively pks-positive and alpha-hemolysin-(hly) and cytotoxic necrotizing factor-1-(cnf1) positive, this novel strain (E. coli 5C) is likely the probiotic E. coli strain with the best safety profile available to date for human use.
Collapse
Affiliation(s)
- Francesco Di Pierro
- Microbiota International Clinical Society, 10123, Turin, Italy
- Scientific & Research Department, Velleja Research, 20125, Milan, Italy
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| | - Valeria Sagheddu
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Serena Galletti
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Alice Casaroli
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Edoardo Labrini
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Sara Soldi
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | | | - Alexander Bertuccioli
- Microbiota International Clinical Society, 10123, Turin, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122, Urbino, Italy
| | - Mariarosaria Matera
- Microbiota International Clinical Society, 10123, Turin, Italy
- Department of Pediatric Emergencies, Misericordia Hospital, 58100, Grosseto, Italy
| | - Ilaria Cavecchia
- Microbiota International Clinical Society, 10123, Turin, Italy
- Microbiomic Department, Koelliker Hospital, 10134, Turin, Italy
| | | | - Maria Laura Tanda
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, 21100, Varese, Italy
| | - Nicola Zerbinati
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| |
Collapse
|
15
|
Wang SL, Chan TA. Navigating established and emerging biomarkers for immune checkpoint inhibitor therapy. Cancer Cell 2025; 43:641-664. [PMID: 40154483 DOI: 10.1016/j.ccell.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved outcomes of patients with many different cancers. These antibodies target molecules such as programmed cell death 1 (PD-1) or cytotoxic T lymphocyte associated protein 4 (CTLA-4) which normally function to limit immune activity. Treatment with ICIs reactivates T cells to destroy tumor cells in a highly specific manner, which in some patients, results in dramatic remissions and durable disease control. Over the last decade, much effort has been directed at characterizing factors that drive efficacy and resistance to ICI therapy. Food and Drug Administration (FDA)-approved biomarkers for ICI therapy have facilitated more judicious treatment of cancer patients and transformed the field of precision oncology. Yet, adaptive immunity against cancers is complex, and newer data have revealed the potential utility of other biomarkers. In this review, we discuss the utility of currently approved biomarkers and highlight how emerging biomarkers can further improve the identification of patients who benefit from ICIs.
Collapse
Affiliation(s)
- Stephen L Wang
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
16
|
Bommana S, Olagoke S, Hu Y, Wang R, Kama M, Dehdashti M, Kodimerla R, Read T, Dean D. Effect of Azithromycin treatment on the microbial composition, functional dynamics and resistomes of endocervical, vaginal and rectal microbiomes of women in Fiji with Chlamydia trachomatis infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646699. [PMID: 40236150 PMCID: PMC11996479 DOI: 10.1101/2025.04.02.646699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Antibiotics disrupt mucosal microbial communities, yet the effects on microbiomes with Chlamydia trachomatis (Ct) infection remain poorly understood. Some data exist on vaginal microbiomes pre- and post-treatment, but none are available for the endocervix or rectum that are primary sites of infection. We applied metagenomic shotgun sequencing to vaginal, endocervical and rectal samples from women who, overtime, had Ct persistence, clearance, or no infection to evaluate azithromycin-induced changes in microbial composition, function, and the resistome. Our results show a shift in composition and function that support Ct post-treatment with azithromycin resistance mutations in the Ct rplV gene and significant endocervical enrichment of azithromycin resistance genes in Lactobacillus iners and Gardnerella vaginalis, the strains of which have moderate/high potential for biofilm formation. These findings highlight the unintended ecological consequences of azithromycin, including resistance gene propagation and microbiome shifts that support persistent/recurrent Ct, emphasizing the need for novel treatment and microbiome-preserving strategies.
Collapse
|
17
|
Connolly JP, Kelly L. The physical biogeography of Fusobacterium nucleatum in health and disease. mBio 2025; 16:e0298924. [PMID: 40062772 PMCID: PMC11980382 DOI: 10.1128/mbio.02989-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/07/2025] [Indexed: 04/10/2025] Open
Abstract
Fusobacterium nucleatum (Fn) is an oral commensal inhabiting the human gingival plaque that is rarely found in the gut. However, in colorectal cancer (CRC), Fn can be isolated from stool samples and detected in metagenomes. We hypothesized that ecological characteristics of the gut are altered by disease, enabling Fn to colonize. Multiple genomically distinct populations of Fn exist, but their ecological preferences are unstudied. We identified six well-separated populations in 133 Fn genomes and used simulated metagenomes to demonstrate sensitive detection of populations in human oral and gut metagenomes. In 9,560 samples from 11 studies, Fn population C2 animalis is elevated in gut metagenomes from CRC and Crohn's disease patients and is observed more frequently in CRC stool samples than in the gingiva. Polymorphum, the most prevalent gingival Fn population, is significantly increased in Crohn's stool samples; this effect was significantly stronger in male hosts than in female. We find polymorphum genomes are enriched for biosynthetic gene clusters and fluoride exporters, while C2 animalis are high in iron transporters. Fn populations thus associate with specific clinical and demographic phenotypes and harbor distinct functional features. Ecological differences in closely related groups of bacteria inform microbiome impacts on human health. IMPORTANCE Fusobacterium nucleatum is a bacterium normally found in the gingiva. F. nucleatum generally does not colonize the healthy gut, but is observed in approximately a third of colorectal cancer (CRC) patient guts. F. nucleatum's presence in the gut during CRC has been linked to worse prognosis and increased tumor proliferation. Here, we describe the population structure of F. nucleatum in oral and gut microbiomes. We report substantial diversity in gene carriage among six distinct populations of F. nucleatum and identify population disease and body-site preferences. We find the C2 animalis population is more common in the CRC gut than in the gingiva and is enriched for iron transporters, which support gut colonization in known pathogens. We find that C2 animalis is also enriched in Crohn's disease and type 2 diabetes, suggesting ecological commonalities between the three diseases. Our work shows that closely related bacteria can have different associations with human physiology.
Collapse
Affiliation(s)
- John P. Connolly
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Libusha Kelly
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
18
|
Lu J, Wei W, Zheng D. Fusobacterium nucleatum in Colorectal Cancer: Ally Mechanism and Targeted Therapy Strategies. RESEARCH (WASHINGTON, D.C.) 2025; 8:0640. [PMID: 40207017 PMCID: PMC11979337 DOI: 10.34133/research.0640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Fusobacterium nucleatum (Fn), an oral anaerobic commensal, has recently been identified as a crucial oncogenic contributor to colorectal cancer pathogenesis through its ectopic colonization in the gastrointestinal tract. Accumulating evidence reveals its multifaceted involvement in colorectal cancer initiation, progression, metastasis, and therapeutic resistance to conventional treatments, including chemotherapy, radiotherapy, and immunotherapy. This perspective highlights recent advances in anti-Fn strategies, including small-molecule inhibitors, nanomedicines, and biopharmaceuticals, while critically analyzing the translational barriers in developing targeted antimicrobial interventions. We further propose potential strategies to overcome current challenges in Fn modulation, aiming to pave the way for more effective therapeutic interventions and better clinical outcomes.
Collapse
Affiliation(s)
- Junna Lu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery,
Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Wei Wei
- State Key Laboratory of Biopharmaceutical Preparation and Delivery,
Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering,
University of Chinese Academy of Sciences, Beijing 100049, China
| | - Diwei Zheng
- State Key Laboratory of Biopharmaceutical Preparation and Delivery,
Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering,
University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
19
|
Greco L, Rubbino F, Ferrari C, Cameletti M, Grizzi F, Bonelli F, Malesci A, Mazzone M, Ricciardiello L, Laghi L. Association of Fusobacterium nucleatum with colorectal cancer molecular subtypes and its outcome: a systematic review. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2025; 6:e5. [PMID: 40297307 PMCID: PMC12035788 DOI: 10.1017/gmb.2025.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025]
Abstract
Colorectal cancer (CRC) represents a relevant public health problem, with high incidence and mortality in Western countries. CRC can occur as sporadic (65%-75%), common familial (25%), or as a consequence of an inherited predisposition (up to 10%). While unravelling its genetic basis has been a long trip leading to relevant clinical implementation over more than 30 years, other contributing factors remain to be clarified. Among these, micro-organisms have emerged as critical players in the development and progression of the disease, as well as for CRC treatment response. Fusobacterium nucleatum (Fn) has been associated with CRC development in both pre-clinical models and clinical settings. Fusobacteria are core members of the human oral microbiome, while being less prevalent in the healthy gut, prompting questions about their localization in CRC and its precursor lesions. This review aims to critically discuss the evidence connecting Fn with CRC pathogenesis, its molecular subtypes and clinical outcomes.
Collapse
Affiliation(s)
- Luana Greco
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Federica Rubbino
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Clarissa Ferrari
- Research and Clinical Trials Office, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | | | - Fabio Grizzi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
| | | | | | - Massimiliano Mazzone
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Macrophage Dynamics Lab, IRCCS Humanitas Research Hospital, Milan, Italy
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Luigi Ricciardiello
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
20
|
Zhang Y, Song Q, Meng Q, Zhao T, Wang X, Meng X, Cong J. Size-dependent ecotoxicological impacts of tire wear particles on zebrafish physiology and gut microbiota: Implications for aquatic ecosystem health. JOURNAL OF HAZARDOUS MATERIALS 2025; 487:137215. [PMID: 39842107 DOI: 10.1016/j.jhazmat.2025.137215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/07/2025] [Accepted: 01/12/2025] [Indexed: 01/24/2025]
Abstract
The ecological impact of tire wear particles (TWP), a significant source of microplastics pollution, is increasingly concerning, especially given their potential effects on the health of aquatic ecosystems. This study investigates the size-dependent ecotoxicological responses of zebrafish (Danio rerio) to TWP exposure, focusing on physiological, metabolic, and microbial community impacts over a 15-day exposure period followed by a 15-day excretion period. Through integrated analysis of gut microbiome composition, liver transcriptomics, and host physiological markers, we found that smaller TWP particles (< 120 μm) induced oxidative stress, evidenced by increased SOD and MDA levels, and inhibited growth by reducing body mass and gut length. In contrast, larger TWP particles (250-380 μm) caused more substantial disruptions in lipid and xenobiotic metabolic pathways, as shown by significant downregulation of key metabolic genes (acads, cpt2_1, hadhaa), and alterations in the gut microbiome, including the enrichment of pathogenic genera, such as Enterococcus and Fusobacterium, while depleting beneficial microbes like Acinetobacter and Methyloversatilis. These microbiome shifts led to a more complex and potentially pathogenic gut microbiome. Notably, zebrafish displayed adaptive resilience during the excretion period, with significant recovery in body mass and microbial composition, emphasizing the adaptive capacity of aquatic organisms to pollutants. Our findings underscore the broader ecological risks posed by TWP, the pivotal role of gut microbiota in host resilience to pollutants, and the need for comprehensive management strategies addressing emerging contaminants in aquatic ecosystems.
Collapse
Affiliation(s)
- Yun Zhang
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Qianqian Song
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Qingxuan Meng
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Tianyu Zhao
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Xiaolong Wang
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Xinrui Meng
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China
| | - Jing Cong
- College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266000, China.
| |
Collapse
|
21
|
Birg A, Lin HC. The Role of Bacteria-Derived Hydrogen Sulfide in Multiple Axes of Disease. Int J Mol Sci 2025; 26:3340. [PMID: 40244174 PMCID: PMC11990059 DOI: 10.3390/ijms26073340] [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: 12/15/2024] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
In this review article, we discuss and explore the role of bacteria-derived hydrogen sulfide. Hydrogen sulfide is a signaling molecule produced endogenously that plays an important role in health and disease. It is also produced by the gut microbiome. In the setting of microbial disturbances leading to disruption of intestinal homeostasis (dysbiosis), the concentration of available hydrogen sulfide can also vary leading to pathologic sequelae. The brain-gut axis is the original studied paradigm of gut microbiome and host interaction. In recent years, our understanding of microbial and host interaction has expanded greatly to include specific pathways that have branched into their own axes. These axes share a principal concept of microbiota changes, intestinal permeability, and an inflammatory response, some of which are modulated by hydrogen sulfide (H2S). In this review, we will discuss multiple axes including the gut-immune, gut-heart, and gut-endocrine axes. We will evaluate the role of H2S in modulation of intestinal barrier, mucosal healing in intestinal inflammation and tumor genesis. We will also explore the role of H2S in alpha-synuclein aggregation and ischemic injury. Finally, we will discuss H2S in the setting of metabolic syndrome as int pertains to hypertension, atherosclerosis and glucose-like peptide-1 activity. Majority of studies that evaluate hydrogen sulfide focus on endogenous production; the role of this review is to examine the lesser-known bacteria-derived source of hydrogen sulfide in the progression of diseases as it relates to these axes.
Collapse
Affiliation(s)
- Aleksandr Birg
- Medicine Service, New Mexico VA Health Care System, Albuquerque, NM 87108, USA;
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87106, USA
| | - Henry C. Lin
- Medicine Service, New Mexico VA Health Care System, Albuquerque, NM 87108, USA;
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87106, USA
| |
Collapse
|
22
|
Ding YP, Liu CC, Yu KD. RNA modifications in the tumor microenvironment: insights into the cancer-immunity cycle and beyond. Exp Hematol Oncol 2025; 14:48. [PMID: 40176140 PMCID: PMC11963313 DOI: 10.1186/s40164-025-00648-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
The chemical modification of biological molecules is a critical regulatory mechanism for controlling molecular functions. Although research has long focused on DNA and proteins, RNA modifications have recently attracted substantial interest with the advancement in detection technologies. In oncology, many studies have identified dysregulated RNA modifications including m6A, m1A, m5C, m7G, pseudouridylation and A to I editing, leading to disrupted downstream pathways. As the concept of the tumor microenvironment has gained prominence, studies have increasingly examined the role of RNA modifications in this context, focusing on interactions among cancer cells, immune cells, stromal cells, and other components. Here we review the RNA modifications in the tumor microenvironment through the perspective of the Cancer-Immunity Cycle. The extracellular RNA modifications including exosomes and influence of microbiome in RNA modifications are potential research questions. Additionally, RNA modifying enzymes including FTO, ALKBH5, METTL3, PUS7 are under investigation as potential biomarkers and targets for combination with immunotherapies. ADCs and mimetics of modified RNA could be potential novel drugs. This review discusses the regulatory roles of RNA modifications within the tumor microenvironment.
Collapse
Affiliation(s)
- You-Peng Ding
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
23
|
Gilbert JA, Azad MB, Bäckhed F, Blaser MJ, Byndloss M, Chiu CY, Chu H, Dugas LR, Elinav E, Gibbons SM, Gilbert KE, Henn MR, Ishaq SL, Ley RE, Lynch SV, Segal E, Spector TD, Strandwitz P, Suez J, Tropini C, Whiteson K, Knight R. Clinical translation of microbiome research. Nat Med 2025; 31:1099-1113. [PMID: 40217076 DOI: 10.1038/s41591-025-03615-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/26/2025] [Indexed: 04/18/2025]
Abstract
The landscape of clinical microbiome research has dramatically evolved over the past decade. By leveraging in vivo and in vitro experimentation, multiomic approaches and computational biology, we have uncovered mechanisms of action and microbial metrics of association and identified effective ways to modify the microbiome in many diseases and treatment modalities. This Review explores recent advances in the clinical application of microbiome research over the past 5 years, while acknowledging existing barriers and highlighting opportunities. We focus on the translation of microbiome research into clinical practice, spearheaded by Food and Drug Administration (FDA)-approved microbiome therapies for recurrent Clostridioides difficile infections and the emerging fields of microbiome-based diagnostics and therapeutics. We highlight key examples of studies demonstrating how microbiome mechanisms, metrics and modifiers can advance clinical practice. We also discuss forward-looking perspectives on key challenges and opportunities toward integrating microbiome data into routine clinical practice, precision medicine and personalized healthcare and nutrition.
Collapse
Affiliation(s)
- Jack A Gilbert
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA.
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA.
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin J Blaser
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Mariana Byndloss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Charles Y Chiu
- Department of Laboratory Medicine, University of California, San Fransisco, San Francisco, CA, USA
- Department of Medicine, Division of Infectious Diseases, University of California, San Fransisco, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Hiutung Chu
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines, La Jolla, CA, USA
| | - Lara R Dugas
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Microbiome and Cancer Division, DKFZ, Heidelberg, Germany
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - Katharine E Gilbert
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | | | - Suzanne L Ishaq
- School of Food and Agriculture, University of Maine, Orono, ME, USA
- Microbes and Social Equity working group, Orono, ME, USA
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - Susan V Lynch
- Benioff Center for Microbiome Medicine, Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- ZOE Ltd, London, UK
| | | | - Jotham Suez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Carolina Tropini
- CIFAR Humans & the Microbiome Program, CIFAR, Toronto, Ontario, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, San Diego, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
24
|
Burke C, Glynn T, Jahangir C, Murphy C, Buckley N, Tangney M, Rahman A, Gallagher WM. Exploring the prognostic and predictive potential of bacterial biomarkers in non-gastrointestinal solid tumors. Expert Rev Mol Diagn 2025; 25:117-128. [PMID: 39973615 DOI: 10.1080/14737159.2025.2465743] [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: 08/19/2024] [Revised: 01/06/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Standard clinical parameters like tumor size, age, lymph node status, and molecular markers are used to predict progression risk and treatment response. However, exploring additional markers that reflect underlying biology could offer a more comprehensive understanding of the tumor microenvironment (TME). The TME influences tumor development, progression, disease severity, and survival, with tumor-associated bacteria posited to play significant roles. Studies on tumor-associated microbiota have focused on high bacterial-load sites such as the gut, oral cavity, and stomach, but interest is growing in non-gastrointestinal (GI) solid tumors, such as breast, lung, and pancreas. Microbe-based biomarkers, including Helicobacter pylori, human papillomavirus (HPV), and hepatitis B and C viruses, have proven valuable in predicting gastric, cervical, and renal cancers. AREAS COVERED Potential of prognostic and predictive bacterial biomarkers in non-GI solid tumors and the methodologies used. EXPERT OPINION Advances in techniques like 16S rRNA gene sequencing, qPCR, immunostaining, and in situ hybridization have enabled detailed analysis of difficult-to-culture microbes in solid tumors. However, to ensure reliable results, it is critical to standardize protocols, accurately align reads, address contamination, and maintain proper sample handling. This will pave the way for developing reliable bacterial markers that enhance prognosis, prediction, and personalized treatment planning.
Collapse
Affiliation(s)
- Caoimbhe Burke
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Thomas Glynn
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Chowdhury Jahangir
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Clodagh Murphy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Niamh Buckley
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland
| | - Mark Tangney
- Cancer Research@UCC, University College Cork, Cork, Ireland
| | - Arman Rahman
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
25
|
Vargas‐Castellanos E, Rincón‐Riveros A. Microsatellite Instability in the Tumor Microenvironment: The Role of Inflammation and the Microbiome. Cancer Med 2025; 14:e70603. [PMID: 40231893 PMCID: PMC11998172 DOI: 10.1002/cam4.70603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/13/2024] [Accepted: 01/03/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Microsatellite instability (MSI) is a hallmark of DNA mismatch repair (MMR) deficiency that leads to genomic instability and increased cancer risk. The tumor microenvironment (TME) significantly influences MSI-driven tumorigenesis, and emerging evidence points to a critical role of the microbiome in shaping this complex interplay. METHODS This review comprehensively examines the existing literature on the intricate relationship between MSI, microbiome, and cancer development, with a particular focus on the impact of microbial dysbiosis on the TME. RESULTS MSI-high tumors exhibited increased immune cell infiltration owing to the generation of neoantigens. However, immune evasion mechanisms such as PD-1/CTLA-4 upregulation limit the efficacy of immune checkpoint inhibitors (ICIs) in a subset of patients. Pathobionts, such as Fusobacterium nucleatum and Bacteroides fragilis, contribute to MSI through the production of genotoxins, further promoting inflammation and oxidative stress within the TME. CONCLUSIONS The microbiome profoundly affects MSI-driven tumorigenesis. Modulation of the gut microbiota through interventions such as fecal microbiota transplantation, probiotics, and dietary changes holds promise for improving ICI response rates. Further research into cancer pharmacomicrobiomics, investigating the interplay between microbial metabolites and anticancer therapies, is crucial for developing personalized treatment strategies.
Collapse
Affiliation(s)
| | - Andrés Rincón‐Riveros
- Facultad de Ciencias de la SaludUniversidad Colegio Mayor de CundinamarcaBogotáColombia
| |
Collapse
|
26
|
Zitvogel L, Derosa L, Routy B, Loibl S, Heinzerling L, de Vries IJM, Engstrand L, Segata N, Kroemer G. Impact of the ONCOBIOME network in cancer microbiome research. Nat Med 2025; 31:1085-1098. [PMID: 40217075 DOI: 10.1038/s41591-025-03608-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
The European Union-sponsored ONCOBIOME network has spurred an international effort to identify and validate relevant gut microbiota-related biomarkers in oncology, generating a unique and publicly available microbiome resource. ONCOBIOME explores the effects of the microbiota on gut permeability and metabolism as well as on antimicrobial and antitumor immune responses. Methods for the diagnosis of gut dysbiosis have been developed based on oncomicrobiome signatures associated with the diagnosis, prognosis and treatment responses in patients with cancer. The mechanisms explaining how dysbiosis compromises natural or therapy-induced immunosurveillance have been explored. Through its integrative approach of leveraging multiple cohorts across populations, cancer types and stages, ONCOBIOME has laid the theoretical and practical foundations for the recognition of microbiota alterations as a hallmark of cancer. ONCOBIOME has launched microbiota-centered interventions and lobbies in favor of official guidelines for avoiding diet-induced or iatrogenic (for example, antibiotic- or proton pump inhibitor-induced) dysbiosis. Here, we review the key advances of the ONCOBIOME network and discuss the progress toward translating these into oncology clinical practice.
Collapse
Affiliation(s)
- Laurence Zitvogel
- INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
- Clinicobiome, Gustave Roussy, Villejuif, France.
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.
| | - Lisa Derosa
- INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Clinicobiome, Gustave Roussy, Villejuif, France
| | - Bertrand Routy
- University of Montreal Research Center (CR-CHUM), Montreal, Quebec, Canada
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Sibylle Loibl
- German Breast Group c/ GBG Forschungs GmbH, Neu-Isenburg, Goethe University, Frankfurt, Germany
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - I Jolanda M de Vries
- Medical Biosciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lars Engstrand
- Department of Microbiology Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology IRCCS, Milan, Italy
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
27
|
Sousa MGC, Brasino DSK, Krieger M, Dindar DA, Duhen R, Zhang Z, Franca CM, Bertassoni LE. Host-microbe-cancer interactions on-a-chip. Front Bioeng Biotechnol 2025; 13:1505963. [PMID: 40230461 PMCID: PMC11994592 DOI: 10.3389/fbioe.2025.1505963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
The tumor microbiota has emerged as a pivotal contributor to a variety of cancers, impacting disease development, progression, and therapeutic resistance. Due to the complexity of the tumor microenvironment, reproducing the interactions between the microbes, tumor cells, and the immune system remains a great challenge for both in vitro and in vivo studies. To this end, significant progress has been made toward leveraging tumor-on-a-chip model systems to replicate critical hallmarks of the native disease in vitro. These microfluidic platforms offer the ability to mimic essential components of the tumor microenvironment, including controllable fluid flow conditions, manipulable extracellular matrix dynamics, and intricate 3D multi-cellular communication. The primary objective of this review is to discuss recent challenges and advances in engineering host-microbiota and tumor interactions on-a-chip. Ultimately, overcoming these obstacles will help us gain deeper insights into tumor-microbe interactions and enhance avenues for developing more effective cancer therapies.
Collapse
Affiliation(s)
- Mauricio G. C. Sousa
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Danielle S. K. Brasino
- Department of Microbiology and Molecular Genetics, Robert Larner College of Medicine at the University of Vermont, Burlington, VT, United States
| | - Madeline Krieger
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Duygu A. Dindar
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Rebekka Duhen
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Zhenzhen Zhang
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Cristiane Miranda Franca
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Luiz E. Bertassoni
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, United States
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
28
|
Alves Costa Silva C, Almonte AA, Zitvogel L. Oncobiomics: Leveraging Microbiome Translational Research in Immuno-Oncology for Clinical-Practice Changes. Biomolecules 2025; 15:504. [PMID: 40305219 PMCID: PMC12024955 DOI: 10.3390/biom15040504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/16/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Growing evidence suggests that cancer should not be viewed solely as a genetic disease but also as the result of functional defects in the metaorganism, including disturbances in the gut microbiota (i.e., gut dysbiosis). The human microbiota plays a critical role in regulating epithelial barrier function in the gut, airways, and skin, along with host metabolism and systemic immune responses against microbes and cancer. Collaborative international networks, such as ONCOBIOME, are essential in advancing research equity and building microbiome resources to identify and validate microbiota-related biomarkers and therapies. In this review, we explore the intricate relationship between the microbiome, metabolism, and cancer immunity, and we propose microbiota-based strategies to improve outcomes for individuals at risk of developing cancer or living with the disease.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
| | - Andrew A. Almonte
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Clinicobiome, 94805 Villejuif, France; (C.A.C.S.); (A.A.A.)
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, 94800 Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, 94270 Kremlin-Bicêtre, France
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, 94805 Villejuif, France
| |
Collapse
|
29
|
Zepeda-Rivera MA, Ponath F, Lewis KN, Gavate RP, Dewhirst FE, Tomida J, Kawamura Y, Tanaka K, Bullman S, Johnston CD. Complete genome sequences of Fusobacterium watanabei sp. isolates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645266. [PMID: 40196618 PMCID: PMC11974791 DOI: 10.1101/2025.03.25.645266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
We report the complete genome sequences of eight Fusobacterium watanabei clinical isolates, ranging from 1.95 to 2.09 Mbp. Analysis against the Genome Taxonomy Database (GTDB) indicates that Fusobacterium watanabei genomes are part of the " Fusobacterium nucleatum_J " group, which also encompasses the previously published FNU strain and Fna C1 isolates.
Collapse
|
30
|
Robinson AV, Vancuren SJ, Marcone M, Allen-Vercoe E. Characterization of diet-linked amino acid pool influence on Fusobacterium spp. growth and metabolism. mSphere 2025; 10:e0078924. [PMID: 39945521 PMCID: PMC11934328 DOI: 10.1128/msphere.00789-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/14/2025] [Indexed: 03/26/2025] Open
Abstract
The genus Fusobacterium contains multiple proteolytic opportunistic pathogens that have been increasingly linked to colorectal cancer (CRC). "Oncomicrobes" such as these fusobacterial species within the gut microbiota may contribute to CRC onset and/or progression. Protein-rich diets may both directly increase CRC risk and enrich for proteolytic oncomicrobes, including Fusobacterium spp. Individual food substrates vary in amino acid content, and released amino acid content that is not absorbed in the small intestine may influence the growth of colonic proteolytic fermenters. Fusobacteria such as Fusobacterium spp. are known to preferentially metabolize certain amino acids. As such, some foods may better support the growth of these species within the colonic environment than others. To explore this, in this study, we created free amino acid pools (FAAPs) to represent proportions of amino acids in major proteins of three common dietary protein sources (soy, beef, and bovine milk). Growth curves were generated for 39 Fusobacterium spp. strains cultured in a dilute medium supplemented with each of the three FAAPs. Thereafter, amino acid use by 31 of the 39 Fusobacterium spp. strains in each FAAP treatment was assessed. FAAP supplementation increased growth metrics of all Fusobacterium spp. strains tested; however, the strains varied greatly in terms of the FAAP(s) generating the greatest increase in growth. Furthermore, the amino acid utilization strategy was highly variable between strains of Fusobacterium spp. Neither growth metrics nor amino acid utilization could be explained by species classification of Fusobacterium spp. strains. This report expands upon the previous knowledge of fusobacterial amino acid metabolism and indicates that proteolytic oncomicrobial activity should be assessed in the context of available protein sources.IMPORTANCEFusobacterium spp. including F. animalis, F. nucleatum, F. vincentii, and F. polymorphum are common oral commensals with emerging importance in diseases across multiple body sites, including CRC. CRC lesions associated with fusobacteria tend to result in poorer prognosis and increased disease recurrence. While Fusobacterium spp. are thought to colonize after tumorigenesis, little is known about the factors that facilitate this colonization. Protein-rich diets yielding readily metabolized free amino acids within the colon may promote the growth of proteolytic fermenters such as fusobacteria. Here, we show that variable concentrations of free amino acids within pools that represent different dietary protein sources differentially influence fusobacterial growth, including CRC-relevant strains of Fusobacterium spp. This work highlights the high degree of variation in fusobacterial amino acid utilization patterns and suggests differing proportions of dietary amino acids that reach the colon could influence fusobacterial growth.
Collapse
Affiliation(s)
- Avery V. Robinson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Sarah J. Vancuren
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| | - Massimo Marcone
- Department of Food Science, University of Guelph, Guelph, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada
| |
Collapse
|
31
|
Shastry RP, Hameed A, Banerjee S, Prabhu A, Bajire SK, Pavan SR, Dhanyashree HR, Kotimoole CN, Stothard P, Surya S, Keshava Prasad TS, Shetty R, Shen FT, Bhandary YP. Tryptic oncopeptide secreted from the gut bacterium Cronobacter malonaticus PO3 promotes colorectal cancer. Sci Rep 2025; 15:9958. [PMID: 40121280 PMCID: PMC11929907 DOI: 10.1038/s41598-025-94666-y] [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: 01/24/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
The involvement of Cronobacter, which is frequently associated with meningitis and necrotizing enterocolitis, in human colorectal cancer remains unexplored. In this study, we isolate and characterize a novel strain of C. malonaticus designated PO3 from a fecal sample of a colon cancer patient and demonstrate its proliferative effects on colorectal cancer both in vitro and in vivo. The secretome of PO3 significantly promoted cell proliferation, as evidenced by increased cell viability, fluorescence intensity, and Ki-67 expression, without inducing cell death. Furthermore, using high-resolution mass spectrometry (HRMS), we identified a novel tryptic oncopeptide designated P506, in the PO3 secretome that promotes colorectal cancer. Synthetic P506 further stimulated human colorectal adenocarcinoma cell line HT-29 cell proliferation in a dose-dependent manner. Experiments with the BALB/c mouse model in vivo revealed that both the PO3 secretome and P506 contributed to the development of colorectal polyps and associated histological changes, including dysplasia and altered colonic architecture. These findings suggest that P506, a potent peptide from the PO3 secretome, may have oncogenic potential, promoting colorectal cancer progression.
Collapse
Affiliation(s)
- Rajesh Padumane Shastry
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India.
| | - Asif Hameed
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India.
| | - Shukla Banerjee
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Ashwini Prabhu
- Division of Cancer Research and Therapeutics, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Sukesh Kumar Bajire
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Sonnenahalli Rudramurthy Pavan
- Division of Cancer Research and Therapeutics, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Honagodu Ravichandra Dhanyashree
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Chinmaya Narayana Kotimoole
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Paul Stothard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Suprith Surya
- Advanced Surgical Skill ENhancement Division (ASSEND), Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | | | - Rohan Shetty
- Department of Surgical Oncology, Yenepoya Medical College Hospital, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Fo-Ting Shen
- Department of Soil and Environmental Sciences, National Chung Hsing University, Taichung, 40227, Taiwan.
- Innovation and Development Center of Sustainable Agriculture (IDCSA), National Chung Hsing University, Taichung, 40227, Taiwan.
| | - Yashodhar Prabhakar Bhandary
- Division of Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| |
Collapse
|
32
|
Zhang X, Chen Y, Xia Y, Lin S, Zhou X, Pang X, Yu J, Sun L. Oral microbiota in colorectal cancer: Unraveling mechanisms and application potential. Life Sci 2025; 365:123462. [PMID: 39947314 DOI: 10.1016/j.lfs.2025.123462] [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: 10/22/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Colorectal cancer (CRC), with a rising prevalence, is the third most commonly diagnosed cancer and the third leading cause of cancer-related death. Studies have shown that a complex interplay between the development of CRC and alterations in the oral microbiome. Recent advancements in genomics and metagenomics have highlighted the significant roles of certain oral microbes, particularly Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum), in the progression of CRC. However, the detailed mechanisms by which the oral microbiota influence CRC development remain unclear. This review aims to elucidate the role of oral microbiota in CRC progression, evaluate their potential as biomarkers, and explore therapeutic strategies targeting these microbes. This review offers insights into the mechanisms underlying the interaction between oral microbiota and CRC, underscoring the potential of oral microbes as diagnostic and prognostic biomarkers, as well as therapeutic targets. Future research should focus on clarifying the exact pathways and developing innovative therapeutic strategies to enhance the diagnosis and treatment.
Collapse
Affiliation(s)
- Xinran Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Yixin Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Yuwei Xia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Shenghao Lin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Xinlei Zhou
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xi Pang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Jieru Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Leitao Sun
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
33
|
Wang K, Han L, Ma J, Fang Y, He Y, Liu X, Wen S, Zhuang J, Yuan H. Gut microbiota community and metabolic profiles in direct total cavopulmonary connection and Fontan circulation: a cross-sectional study in the single center. Front Microbiol 2025; 16:1539046. [PMID: 40160266 PMCID: PMC11952763 DOI: 10.3389/fmicb.2025.1539046] [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: 12/03/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Background This study aimed to evaluate the impact of the direct total cavopulmonary connection (dTCPC) procedure on the gut microbiome and metabolome. It also sought to elucidate the features of the gut microbiota community and metabolic profiles in Fontan circulation. Methods We randomly recruited 45 participants above 14 years old undergoing Fontan procedure by typical extracardiac conduit (TCPC group, n = 15), direct total cavopulmonary connection (dTCPC group, n = 16) procedure and healthy control (control group, n = 14) in our institution between May 2023 and October 2023. 16S rRNA amplicon sequencing and untargeted metabolites measurement were performed on their fecal sample. Results The four alpha diversity indexes showed no statistical significance between the dTCPC and TCPC groups (p > 0.05). Orthogonal partial least squares discriminant analysis (OPLS-DA) followed by permutation testing indicated an overfitting effect in the model between the dTCPC and TCPC groups. We observed significant differences in the Chao1 index (p = 0.0236), the ACE index (p = 0.0236), and the unweighted beta diversity (p = 0.0099) between the Fontan group and healthy control group. Strains of Fusobacterium were significantly overrepresented in the Fontan group [with linear discriminant analysis (LDA) scores exceeding (log10) >3]. Functional enrichment analysis revealed a significant overrepresentation of several metabolic pathways. These pathways predominantly included those related to amino acid metabolism, such as histidine metabolism, glycine, serine, and threonine metabolism, and cysteine and methionine metabolism. Additionally, the biosynthesis of unsaturated fatty acids was also notably enriched. Conclusion The dTCPC procedure demonstrated similar gut microbiota composition and metabolic profiles to the traditional ECC procedure in Fontan patients. Notably, the increased abundance of Fusobacterium, reduced microbial biodiversity, and altered metabolic profiles of amino acids and unsaturated fatty acids in the alimentary canal may serve as distinctive characteristics of patients who have undergone Fontan circulation. These findings provide valuable insights into the long-term physiological consequences of Fontan procedure and may inform future clinical management strategies.
Collapse
Affiliation(s)
- Kaiyu Wang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Linjiang Han
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Jianrui Ma
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Yushen Fang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yinru He
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaobing Liu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shusheng Wen
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jian Zhuang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| | - Haiyun Yuan
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, China
| |
Collapse
|
34
|
Liu QL, Zhou H, Wang Z, Chen Y. Exploring the role of gut microbiota in colorectal liver metastasis through the gut-liver axis. Front Cell Dev Biol 2025; 13:1563184. [PMID: 40181829 PMCID: PMC11965903 DOI: 10.3389/fcell.2025.1563184] [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: 01/19/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Colorectal liver metastasis (CRLM) represents a major therapeutic challenge in colorectal cancer (CRC), with complex interactions between the gut microbiota and the liver tumor microenvironment (TME) playing a crucial role in disease progression via the gut-liver axis. The gut barrier serves as a gatekeeper, regulating microbial translocation, which influences liver colonization and metastasis. Through the gut-liver axis, the microbiota actively shapes the TME, where specific microbial species and their metabolites exert dual roles in immune modulation. The immunologically "cold" nature of the liver, combined with the influence of the gut microbiota on liver immunity, complicates effective immunotherapy. However, microbiota-targeted interventions present promising strategies to enhance immunotherapy outcomes by modulating the gut-liver axis. Overall, this review highlights the emerging evidence on the role of the gut microbiota in CRLM and provides insights into the molecular mechanisms driving the dynamic interactions within the gut-liver axis.
Collapse
Affiliation(s)
- Qiu-Luo Liu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Institute of Digestive Surgery, Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Huijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Health Management Center, General Practice Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ziqiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Chen
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
35
|
Zong G, Deng R, Pan Y, Liu M, Zhu H, Tao R, Shan Y, Wei Z, Lu Y. Ginseng polysaccharides ameliorate colorectal tumorigenesis through Lachnospiraceae-mediated immune modulation. Int J Biol Macromol 2025; 307:142015. [PMID: 40081698 DOI: 10.1016/j.ijbiomac.2025.142015] [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: 12/08/2024] [Revised: 02/24/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Probiotics and their metabolites play a critical role in immunotherapy for colorectal cancer (CRC) and intestinal damage. Identifying specific probiotics from natural products and elucidating the underlying mechanisms represent promising strategies for CRC research. This study investigated the structural characterization and therapeutic potential of ginseng polysaccharides (GPS) in inhibiting tumor growth. The results showed that the molecular weight of GPS was 2425.512 kDa, which was mainly composed of Man, GluA, Gal, Glc, Xyl, and Ara contained in its structure. GPS (100, 200, and 400 mg/kg) significantly ameliorates colorectal tumorigenesis in AOM/DSS-induced and MC38-induced CRC models. 16S rRNA shows that GPS supplementation significantly increased the abundance of Lachnospiraceae compared to the model group. Mechanistically, GPS supplementation promoted the proliferation of beneficial Lachnospiraceae bacterium (L.B.), leading to increased short-chain fatty acids (SCFAs) production. The effective anti-CRC effects of key probiotics were further substantiated by their ability to inhibit myeloid-derived suppressor cells (MDSCs) and enhance the infiltration and activation of CD8+ T cells. These findings highlight the pivotal role of GPS-induced alterations in the potential probiotics L.B. production in CRC suppression, emphasizing the potential of GPS in immune regulation for microbiome-targeted cancer therapies.
Collapse
Affiliation(s)
- Gangfan Zong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated with Nanjing University of Chinese Medicine, Nanjing 210014, China
| | - Yehua Pan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mingyuan Liu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hongzheng Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruizhi Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
36
|
Scano A, Fais S, Ciappina G, Genovese M, Granata B, Montopoli M, Consolo P, Carroccio P, Muscolino P, Ottaiano A, Bignucolo A, Picone A, Toscano E, Orrù G, Berretta M. Oxidative Stress by H 2O 2 as a Potential Inductor in the Switch from Commensal to Pathogen in Oncogenic Bacterium Fusobacterium nucleatum. Antioxidants (Basel) 2025; 14:323. [PMID: 40227274 PMCID: PMC11939671 DOI: 10.3390/antiox14030323] [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: 01/12/2025] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Fusobacterium nucleatum is a pathobiont that plays a dual role as both a commensal and a pathogen. The oral cavity typically harbors this anaerobic, Gram-negative bacterium. At the same time, it is closely linked to colorectal cancer due to its potential involvement in tumor progression and resistance to chemotherapy. The mechanism by which it transforms from a commensal to a pathogen remains unknown. For this reason, we investigated the role of oxidative status as an initiatory factor in changing the bacterium's pathogenicity profile. METHODS A clinical strain of F. nucleatum subsp. animalis biofilm was exposed to different oxidative stress levels through varying subinhibitory amounts of H2O2. Subsequently, we investigated the bacterium's behavior in vitro by infecting the HT-29 cell line. We evaluated bacterial colonization, volatile sulfur compounds production, and the infected cell's oxidative status by analyzing HMOX1, pri-miRNA 155, and 146a gene expression. RESULTS The bacterial colonization rate, dimethyl sulfide production, and pri-miRNA 155 levels all increased when stressed bacteria were used, suggesting a predominant pathogenic function of these strains. CONCLUSIONS The response of F. nucleatum to different oxidative conditions could potentially explain the increase in its pathogenic traits and the existence of environmental factors that may trigger the bacterium's pathogenicity and virulence.
Collapse
Affiliation(s)
- Alessandra Scano
- Oral Biotechnology Laboratory (OBL), Department of Surgical Science, University of Cagliari, 09124 Cagliari, Italy; (A.S.); (S.F.)
| | - Sara Fais
- Oral Biotechnology Laboratory (OBL), Department of Surgical Science, University of Cagliari, 09124 Cagliari, Italy; (A.S.); (S.F.)
- Molecular Biology Laboratory, Azienda Ospedaliera Universitaria di Cagliari (AOU), 09124 Cagliari, Italy
| | - Giuliana Ciappina
- Division of Medical Oncology, AOU “G. Martino” Hospital, University of Messina, 98124 Messina, Italy; (G.C.); (A.P.)
| | - Martina Genovese
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.G.); (P.C.); (A.B.)
| | - Barbara Granata
- School of Specialization in Medical Oncology, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (B.G.); (P.C.); (P.M.); (E.T.)
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy;
| | - Pierluigi Consolo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.G.); (P.C.); (A.B.)
| | - Patrizia Carroccio
- School of Specialization in Medical Oncology, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (B.G.); (P.C.); (P.M.); (E.T.)
| | - Paola Muscolino
- School of Specialization in Medical Oncology, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (B.G.); (P.C.); (P.M.); (E.T.)
| | - Alessandro Ottaiano
- Division of Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy;
| | - Alessia Bignucolo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.G.); (P.C.); (A.B.)
| | - Antonio Picone
- Division of Medical Oncology, AOU “G. Martino” Hospital, University of Messina, 98124 Messina, Italy; (G.C.); (A.P.)
| | - Enrica Toscano
- School of Specialization in Medical Oncology, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy; (B.G.); (P.C.); (P.M.); (E.T.)
| | - Germano Orrù
- Oral Biotechnology Laboratory (OBL), Department of Surgical Science, University of Cagliari, 09124 Cagliari, Italy; (A.S.); (S.F.)
- Molecular Biology Laboratory, Azienda Ospedaliera Universitaria di Cagliari (AOU), 09124 Cagliari, Italy
| | - Massimiliano Berretta
- Division of Medical Oncology, AOU “G. Martino” Hospital, University of Messina, 98124 Messina, Italy; (G.C.); (A.P.)
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.G.); (P.C.); (A.B.)
| |
Collapse
|
37
|
Chen Y, Zhang Y, Dai M, Qiu C, Sun Q, Fan T, Guo Y, Zhao L, Jiang Y. γ-Linolenic acid derived from Lactobacillus plantarum MM89 induces ferroptosis in colorectal cancer. Food Funct 2025; 16:1760-1771. [PMID: 39924991 DOI: 10.1039/d4fo04790g] [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: 02/11/2025]
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers worldwide; however, current treatment options are inadequate, necessitating the exploration of new therapeutic strategies. The microbiota significantly influences the tumor microenvironment, suggesting that probiotics may serve as promising candidates for cancer treatment. We previously identified a novel probiotic, Lactobacillus plantarum MM89 (L. plantarum MM89), which was found to regulate the immune microenvironment. However, its specific role in CRC remained unclear. In this study, we employed an azoxymethane/dextran sodium sulfate-induced carcinogenesis mouse model to evaluate the therapeutic effects of L. plantarum MM89 in vivo. Transcriptome analysis was conducted to elucidate the mechanisms of action of L. plantarum MM89. Ferroptosis induction in tumor cells was assessed through cell viability assays and C11-BODIPY staining. Liquid chromatography/mass spectrometry was used to identify metabolites derived from L. plantarum MM89. MitoTracker and MitoTracker CMXRos staining and ATP content measurements were performed to assess mitochondrial damage. L. plantarum MM89 significantly inhibited tumor growth in vivo and alleviated intestinal inflammation at non-tumor foci. Transcriptome analysis and immunohistochemistry revealed that L. plantarum MM89 enhanced arachidonic acid metabolism. Small molecules present in the L. plantarum MM89 supernatant induced ferroptosis in cancer cells, as indicated by cell viability and C11-BODIPY assays. Furthermore, γ-linolenic acid (γ-LA) derived from L. plantarum MM89 was shown to induce ferroptosis via mitochondrial damage. In conclusion, γ-LA derived from L. plantarum MM89 triggers ferroptosis in tumor cells by inducing mitochondrial damage, highlighting its potential as a novel therapeutic agent for CRC treatment.
Collapse
Affiliation(s)
- Yan Chen
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Yijie Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
| | - Mengmeng Dai
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Cheng Qiu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Qinsheng Sun
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
| | - Tingting Fan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yuan Guo
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Liqing Zhao
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China.
| | - Yuyang Jiang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
38
|
Ang TL, Koo SH, Ang D, Tan CK, Wang LM, Wong SH, Chow PKH. Postcholecystectomy Gut Microbiome Changes and the Clinical Impact: A Systematic Review With Narrative Synthesis. J Gastroenterol Hepatol 2025; 40:574-583. [PMID: 39675817 DOI: 10.1111/jgh.16846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Cholecystectomy (CCE) can affect the enterohepatic circulation of bile acids and result in gut microbiome changes. This systematic review aimed to clarify the effect of CCE on gut microbiome composition and its clinical impact. METHOD A systematic search was conducted in PubMed, Web of Science, and Scopus, combining keywords such as "cholecystectomy" or "post-cholecystectomy" with "gut microbiome," "stool microbiome," or "gut dysbiosis." Data were extracted and synthesized using narrative review. Study quality was assessed using the Newcastle-Ottawa scale. RESULTS A total of 1373 articles were screened and 14 studies were selected. Significant but inconsistent microbiome changes were reported. Changes were observed in alpha and beta diversity. At phylum level, an increase in Bacteroides and Ascomycota, decrease in Firmicutes, Actinomycetes, and Basidiomycota, and both increase and decrease in Fusobacteria were reported. At genus level, an increase in Prevotella and a decrease in Faecalibacterium were reported. In post-CCE diarrhea, decreased beta diversity, a decreased F/B ratio, an increase in Prevotella, an increase in Phocaeicola vulgatus, and a decrease in Prevotella copri were noted. For post-CCE syndrome, a higher abundance of Proteobacteria and decreased Firmicutes/Bacteroides (F/B) ratio were reported. A decreased relative abundance of Bifidobacterium longum subsp. longum from controls to CCE without colonic neoplasia to CCE with colonic neoplasia, and an increased abundance of Candida glabrata from controls, to CCE without colonic neoplasia and CCE with colonic neoplasia, were reported. CONCLUSION Patients who underwent CCE had significant gut dysbiosis. However, current studies could not clarify the detailed gut microbial structural and functional changes associated with CCE.
Collapse
Affiliation(s)
- Tiing Leong Ang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-NUS Medical School, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Seok Hwee Koo
- Research Laboratory, Clinical Trials and Research Unit, Changi General Hospital, Singapore
| | - Daphne Ang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Duke-NUS Medical School, Singapore
| | - Chin Kimg Tan
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lai Mun Wang
- Department of Gastroenterology and Hepatology, Changi General Hospital, Singapore
- Department of Pathology, Parkway Laboratory Services Ltd, Singapore
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore
| | - Pierce K H Chow
- Duke-NUS Medical School, Singapore
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, National Cancer Center and Singapore General Hospital, Singapore
- Program in Translational and Clinical Research in Liver Cancer, National Cancer Centre Singapore, Singapore
| |
Collapse
|
39
|
Zhou P, G C B, Wu C. Development of a conditional plasmid for gene deletion in non-model Fusobacterium nucleatum strains. Appl Environ Microbiol 2025; 91:e0181624. [PMID: 39853127 PMCID: PMC11837546 DOI: 10.1128/aem.01816-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025] Open
Abstract
Fusobacterium nucleatum is an opportunistic pathogen with four subspecies: nucleatum (FNN), vincentii (FNV), polymorphum (FNP), and animalis (FNA), each with distinct disease potentials. Research on fusobacterial pathogenesis has mainly focused on the model strain ATCC 23726 from FNN. However, this narrow focus may overlook significant behaviors of other FNN strains and those from other subspecies, given the genetic and phenotypic diversity within F. nucleatum. While ATCC 23726 is highly transformable, most other Fusobacterium strains exhibit low transformation efficiency, complicating traditional gene deletion methods that rely on non-replicating plasmids. To address this, we developed a conditional plasmid system in which the RepA protein, essential for replication of a pCWU6-based shuttle plasmid, is controlled by an inducible system combining an fdx promoter with a theophylline-responsive riboswitch. This system allows plasmid replication in host cells upon induction and plasmid loss when the inducer is removed, forcing chromosomal integration via homologous recombination in the presence of the antibiotic thiamphenicol. We validated this approach by targeting the galK gene, successfully generating mutants in FNN (ATCC 23726, CTI-2), FNP (ATCC 10953), FNA (21_1A), and the closely related species Fusobacterium periodonticum. Incorporating a sacB counterselection marker in this conditional plasmid enabled the deletion of the radD gene in non-model strains. Interestingly, while radD deletion in 23726, 10953, and 21_1A abolished coaggregation with Actinomyces oris, the CTI-2 mutant retained this ability, suggesting the involvement of other unknown adhesins. This work significantly advances gene deletion in genetically recalcitrant F. nucleatum strains, enhancing our understanding of this pathogen.IMPORTANCEFusobacterium nucleatum is implicated in various human diseases, including periodontal disease, preterm birth, and colorectal cancer, often linked to specific strains and reflecting the species' genetic and phenotypic diversity. Despite this diversity, most genetic research has centered on the model strain ATCC 23726, potentially missing key aspects of other strains' pathogenic potential. This study addresses a critical gap by developing a novel conditional plasmid system that enables gene deletion in genetically recalcitrant strains of F. nucleatum. We successfully deleted genes in the FNN clinical strain CTI-2, the FNA strain 21_1A, and F. periodonticum for the first time. Our findings, particularly the varying behavior of the radD gene production in coaggregation across strains, underscore the complexity of F. nucleatum and the need for broader genetic studies. This work advances our understanding of F. nucleatum virulence at the strain level and provides a valuable tool for future bacterial genetics research.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Microbiology & Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Bibek G C
- Department of Microbiology & Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Chenggang Wu
- Department of Microbiology & Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
40
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [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: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
41
|
Qu EB, Baker JS, Markey L, Khadka V, Mancuso C, Tripp D, Lieberman TD. Intraspecies associations from strain-rich metagenome samples. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636498. [PMID: 39974997 PMCID: PMC11839054 DOI: 10.1101/2025.02.07.636498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Genetically distinct strains of a species can vary widely in phenotype, reducing the utility of species-resolved microbiome measurements for detecting associations with health or disease. While metagenomics theoretically provides information on all strains in a sample, current strain-resolved analysis methods face a tradeoff: de novo genotyping approaches can detect novel strains but struggle when applied to strain-rich or low-coverage samples, while reference database methods work robustly across sample types but are insensitive to novel diversity. We present PHLAME, a method that bridges this divide by combining the advantages of reference-based approaches with novelty awareness. PHLAME explicitly defines clades at multiple phylogenetic levels and introduces a probabilistic, mutation-based, framework to accurately quantify novelty from the nearest reference. By applying PHLAME to publicly available human skin and vaginal metagenomes, we uncover previously undetected clade associations with coexisting species, geography, and host age. The ability to characterize intraspecies associations and dynamics in previously inaccessible environments will propel new mechanistic insights from accumulating metagenomic data.
Collapse
Affiliation(s)
- Evan B. Qu
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| | - Jacob S. Baker
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| | - Laura Markey
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| | - Veda Khadka
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| | - Chris Mancuso
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| | - Delphine Tripp
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Systems Biology, Harvard University, Cambridge, MA 02138, USA
| | - Tami D. Lieberman
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA 02139, USA
| |
Collapse
|
42
|
Berg SZ, Berg J. Microbes, macrophages, and melanin: a unifying theory of disease as exemplified by cancer. Front Immunol 2025; 15:1493978. [PMID: 39981299 PMCID: PMC11840190 DOI: 10.3389/fimmu.2024.1493978] [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: 09/10/2024] [Accepted: 12/03/2024] [Indexed: 02/22/2025] Open
Abstract
It is widely accepted that cancer mostly arises from random spontaneous mutations triggered by environmental factors. Our theory challenges the idea of the random somatic mutation theory (SMT). The SMT does not fit well with Charles Darwin's theory of evolution in that the same relatively few mutations would occur so frequently and that these mutations would lead to death rather than survival of the fittest. However, it would fit well under the theory of evolution, if we were to look at it from the vantage point of pathogens and their supporting microbial communities colonizing humans and mutating host cells for their own benefit, as it does give them an evolutionary advantage and they are capable of selecting genes to mutate and of inserting their own DNA or RNA into hosts. In this article, we provide evidence that tumors are actually complex microbial communities composed of various microorganisms living within biofilms encapsulated by a hard matrix; that these microorganisms are what cause the genetic mutations seen in cancer and control angiogenesis; that these pathogens spread by hiding in tumor cells and M2 or M2-like macrophages and other phagocytic immune cells and traveling inside them to distant sites camouflaged by platelets, which they also reprogram, and prepare the distant site for metastasis; that risk factors for cancer are sources of energy that pathogens are able to utilize; and that, in accordance with our previous unifying theory of disease, pathogens utilize melanin for energy for building and sustaining tumors and metastasis. We propose a paradigm shift in our understanding of what cancer is, and, thereby, a different trajectory for avenues of treatment and prevention.
Collapse
Affiliation(s)
- Stacie Z. Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| | - Jonathan Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| |
Collapse
|
43
|
Franklin DS, Chen YW, Chen Y, Wittchen M, Agnew A, Luu A, Whitelegge JP, Zhou ZH, Tauch A, Das A, Ton-That H. Ethanolamine-induced assembly of microcompartments is required for Fusobacterium nucleatum virulence. mBio 2025; 16:e0340524. [PMID: 39714188 PMCID: PMC11796344 DOI: 10.1128/mbio.03405-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
Many bacteria metabolize ethanolamine as a nutrient source through cytoplasmic organelles named bacterial microcompartments (BMCs). Here we investigated the molecular assembly, regulation, and function of BMCs in Fusobacterium nucleatum-a Gram-negative oral pathobiont that is associated with adverse pregnancy outcomes. The F. nucleatum genome harbors a conserved ethanolamine utilization (eut) locus with 21 genes that encode several putative BMC shell proteins and a two-component signal transduction system (TCS), in addition to the enzymes for ethanolamine transport and catabolism. We show that the expression of most of these genes and BMC formation are highly increased in wild-type fusobacteria when cultured in the presence of ethanolamine as a nutrient source. Deletion of the response regulator EutV eliminated this induction of eut mRNAs and BMCs, thus demonstrating that BMC formation is transcriptionally regulated by the TCS EutV-EutW in response to ethanolamine. Mass spectrometry of isolated BMCs unveiled the identity of the constituent proteins EutL, EutM1, EutM2, and EutN. Consistent with the role of these proteins in BMC assembly and metabolism, deletion of eutN, eutL/eutM1/eutM2, or eutL/eutM1/eutM2/eutN not only affected BMC formation but also ethanolamine utilization, causing cell growth defects with ethanolamine as a nutrient. BMCs are also assembled in fusobacteria cultured with placental cells or the culture media, a process that is dependent on the BMC shell proteins. Significantly, we show that the eutN mutant is defective in inducing preterm birth in a mouse model. Together, these results establish that the BMC-mediated metabolism of ethanolamine is critical for fusobacterial virulence. IMPORTANCE The oral anaerobe Fusobacterium nucleatum can spread to distal internal organs, such as the colon and placenta, thereby promoting the development of colorectal cancer and inducing preterm birth, respectively. Yet, how this opportunistic pathogen adapts to the various metabolically distinct host cellular niches remains poorly understood. We demonstrated here that this microbe assembles specialized metabolic organelles, termed bacterial microcompartments (BMCs), to utilize environmental ethanolamine (EA) as a key environmental nutrient source. The formation of F. nucleatum BMCs, containing BMC shell proteins EutLM1M2N, is controlled by a two-component system, EutV-EutW, responsive to EA. Significantly, this ability of F. nucleatum to form BMCs in response to EA is crucial for its pathogenicity evidenced by the fact that the genetic disruption of BMC formation reduces fusobacterial virulence in a mouse model of preterm birth.
Collapse
Affiliation(s)
- Dana S. Franklin
- Molecular Biology Institute, University of California, Los Angeles, California, USA
| | - Yi-Wei Chen
- Division of Oral & Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, California, USA
| | - Yimin Chen
- Division of Oral & Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, California, USA
| | - Manuel Wittchen
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Angela Agnew
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Alexis Luu
- Division of Oral & Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, California, USA
| | - Julian P. Whitelegge
- Pasarow Mass Spectrometry Laboratory, NPI-Semel Institute, University of California Los Angeles, Los Angeles, California, USA
| | - Z. Hong Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Andreas Tauch
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Asis Das
- Department of Medicine, Neag Comprehensive Cancer Center, School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Hung Ton-That
- Molecular Biology Institute, University of California, Los Angeles, California, USA
- Division of Oral & Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, California, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
44
|
Nakatsu G, Ko D, Michaud M, Franzosa EA, Morgan XC, Huttenhower C, Garrett WS. Virulence factor discovery identifies associations between the Fic gene family and Fap2 + fusobacteria in colorectal cancer microbiomes. mBio 2025; 16:e0373224. [PMID: 39807864 PMCID: PMC11796403 DOI: 10.1128/mbio.03732-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Fusobacterium is a bacterium associated with colorectal cancer (CRC) tumorigenesis, progression, and metastasis. Fap2 is a fusobacteria-specific outer membrane galactose-binding lectin that mediates Fusobacterium adherence to and invasion of CRC tumors. Advances in omics analyses provide an opportunity to profile and identify microbial genomic features that correlate with the cancer-associated bacterial virulence factor Fap2. Here, we analyze genomes of Fusobacterium colon tumor isolates and find that a family of post-translational modification enzymes containing Fic domains is associated with Fap2 positivity in these strains. We demonstrate that Fic family genes expand with the presence of Fap2 in the fusobacterial pangenome. Through comparative genomic analysis, we find that Fap2+ Fusobacteriota are highly enriched with Fic gene families compared to other cancer-associated and human gut microbiome bacterial taxa. Using a global data set of CRC shotgun metagenomes, we show that fusobacterial Fic and Fap2 genes frequently co-occur in the fecal microbiomes of individuals with late-stage CRC. We further characterize specific Fic gene families harbored by Fap2+ Fusobacterium animalis genomes and detect recombination events and elements of horizontal gene transfer via synteny analysis of Fic gene loci. Exposure of a F. animalis strain to a colon adenocarcinoma cell line increases gene expression of fusobacterial Fic and virulence-associated adhesins. Finally, we demonstrate that Fic proteins are synthesized by F. animalis as Fic peptides are detectable in F. animalis monoculture supernatants. Taken together, our study uncovers Fic genes as potential virulence factors in Fap2+ fusobacterial genomes.IMPORTANCEAccumulating data support that bacterial members of the intra-tumoral microbiota critically influence colorectal cancer progression. Yet, relatively little is known about non-adhesin fusobacterial virulence factors that may influence carcinogenesis. Our genomic analysis and expression assays in fusobacteria identify Fic domain-containing genes, well-studied virulence factors in pathogenic bacteria, as potential fusobacterial virulence features. The Fic family proteins that we find are encoded by fusobacteria and expressed by Fusobacterium animalis merit future investigation to assess their roles in colorectal cancer development and progression.
Collapse
Affiliation(s)
- Geicho Nakatsu
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
| | - Duhyun Ko
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
| | - Monia Michaud
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
| | - Eric A. Franzosa
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Xochitl C. Morgan
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Curtis Huttenhower
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Wendy S. Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Turocy T, Crawford JM. Bacterial small molecule metabolites implicated in gastrointestinal cancer development. Nat Rev Microbiol 2025; 23:106-121. [PMID: 39375475 DOI: 10.1038/s41579-024-01103-4] [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: 08/28/2024] [Indexed: 10/09/2024]
Abstract
Numerous associations have been identified between cancer and the composition and function of the human microbiome. As cancer remains the second leading global cause of mortality, investigating the carcinogenic contributions of microbiome members could advance our understanding of cancer risk and support potential therapeutic interventions. Although fluctuations in bacterial species have been associated with cancer progression, studying their small molecule metabolites offers one avenue to establish support for causal relationships and the molecular mechanisms governing host-microorganism interactions. In this Review, we explore the expanding repertoire of small molecule metabolites and their mechanisms implicated in the risk of developing gastrointestinal cancers.
Collapse
Affiliation(s)
- Tayah Turocy
- Department of Chemistry, Yale University, New Haven, CT, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA
| | - Jason M Crawford
- Department of Chemistry, Yale University, New Haven, CT, USA.
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA.
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
46
|
Ghaddar BC, Blaser MJ, De S. Revisiting the cancer microbiome using PRISM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634087. [PMID: 39896561 PMCID: PMC11785023 DOI: 10.1101/2025.01.21.634087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Recent controversy around the cancer microbiome highlights the need for improved microbial analysis methods for human genomics data. We developed PRISM, a computational approach for precise microorganism identification and decontamination from low-biomass sequencing data. PRISM removes spurious signals and achieves excellent performance when benchmarked on a curated dataset of 62,006 known true- and false-positive taxa. We then use PRISM to detect microbes in 8 cancer types from the CPTAC and TCGA datasets. We identify rich microbiomes in gastrointestinal tract tumors in CPTAC and identify bacteria in a subset of pancreatic tumors that are associated with altered glycoproteomes, more extensive smoking histories, and higher tumor recurrence risk. We find relatively sparse microbes in other cancer types and in TCGA, which we demonstrate may reflect differing sequencing parameters. Overall, PRISM does not replace gold-standard controls, but it enables higher-confidence analyses and reveals tumor-associated microorganisms with potential molecular and clinical significance.
Collapse
Affiliation(s)
- Bassel C. Ghaddar
- Center for Systems and Computational Biology, Rutgers Cancer Institute, Rutgers University; 195 Albany St., New Brunswick, New Jersey 08901
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University; 679 Hoes Lane West, Piscataway, New Jersey 08854
| | - Subhajyoti De
- Center for Systems and Computational Biology, Rutgers Cancer Institute, Rutgers University; 195 Albany St., New Brunswick, New Jersey 08901
| |
Collapse
|
47
|
Li X, Zhang S, Sheng H, Zhen Y, Wu B, Li Z, Chen D, Zhou H. Oral Fusobacterium nucleatum resists the acidic pH of the stomach due to membrane erucic acid synthesized via enoyl-CoA hydratase-related protein FnFabM. J Oral Microbiol 2025; 17:2453964. [PMID: 39845704 PMCID: PMC11753016 DOI: 10.1080/20002297.2025.2453964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/11/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
Background and Objective Oral bacteria can translocate to the intestine, and their colonization efficiency is influenced by the gastrointestinal tract pH. Understanding how oral bacteria resist acidic environments is crucial for elucidating their role in gut health and disease. Methods To investigate the mechanisms of acid resistance in oral bacteria, an in vitro gastrointestinal tract Dynamic pH Model was established. This model was used to simulate the acidic conditions encountered by bacteria during their translocation from the mouth to the intestine. Results Fusobacterium nucleatum exhibited the highest survival rate in an acidified fluid mimicking the stomach pH (pH 1.5). The survival was significantly increased in the presence of erucic acid C22:1(n9) in cell membranes. Phylogenetic tree analysis revealed that C22:1(n9) synthesis was significantly associated with FnFabM gene expression in F. nucleatum at pH 1.5. Inhibition of FnFabM expression by cerulenin reduced the C22:1(n9) content and decreased the colonization efficiency of F. nucleatum in the stomach and jejunum of mice. Conclusions Oral F. nucleatum translocate to the intestine by resisting the acidic environment owing to the presence of erucic acid in its cell membrane, which is regulated by FnFabM. These results provide novel insights into the mechanisms underlying the oral bacteria survival in acidic environments and their potential to colonize the intestine; thus, shedding light on the oral-gut axis and its implications on human health.
Collapse
Affiliation(s)
- Xiaocong Li
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen City, Guangdong, China
| | - Shipeng Zhang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong, China
| | - Huafang Sheng
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong, China
| | - Yan Zhen
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen City, Guangdong, China
| | - Buling Wu
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen City, Guangdong, China
| | - Zhuang Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong, China
| | - Dingqiang Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong, China
| | - Hongwei Zhou
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen City, Guangdong, China
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou City, Guangdong, China
| |
Collapse
|
48
|
Wong CC, Yu J. pks +E. coli adhesins-The fine line between good and evil. Cell Host Microbe 2025; 33:1-3. [PMID: 39788090 DOI: 10.1016/j.chom.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Gut bacteria could promote colorectal cancer by generating genotoxins. In a recent issue of Nature, Jans et al. identified bacterial adhesion as an additional determinant for the genotoxic activity of colibactin-producing E. coli on the colon epithelium, which could be targeted to mitigate bacteria-associated DNA damage.
Collapse
Affiliation(s)
- Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
49
|
Chiscuzzu F, Crescio C, Varrucciu S, Rizzo D, Sali M, Delogu G, Bussu F. Current Evidence on the Relation Between Microbiota and Oral Cancer-The Role of Fusobacterium nucleatum-A Narrative Review. Cancers (Basel) 2025; 17:171. [PMID: 39857953 PMCID: PMC11763498 DOI: 10.3390/cancers17020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one the most prevalent head and neck cancers and represents a major cause of morbidity and mortality worldwide. The main established risk factors for OSCC include tobacco and alcohol consumption and betel quid chewing, which may contribute alone or in combination with other environmental factors to carcinogenesis. The oral microbiota is emerging as a key player in the establishment of the molecular and cellular mechanisms that may trigger or promote carcinogenesis, including in the oral cavity. Among the bacterial species found in the oral microbiota, Fusobacterium nucleatum, an anaerobic bacterium commonly found in oral biofilms and a periodontal pathogen, has gained attention due to solid evidence implicating F. nucleatum in colorectal cancer (CRC). F. nucleatum has been shown to induce chronic inflammation, promote cell proliferation and trigger cellular invasion while deploying immune evasion mechanisms. These experimental findings were first obtained in in vitro and in vivo models of CRC and are being confirmed in studies on OSCC. In this review, we summarize the most recent findings on the role of F. nucleatum in OSCC, discuss the clinical implications in terms of prognosis and provide an overview of the key mechanisms involved. Moreover, we identify research questions and aspects that require investigations to clarify the role of F. nucleatum in OSCC. We anticipate that studies in this emerging field may have a significant clinical impact on the diagnosis, prognosis and management of OSCC.
Collapse
Affiliation(s)
| | - Claudia Crescio
- Otolaryngology Division, Azienda Ospedaliera Universitaria di Sassari, 07100 Sassari, Italy; (D.R.); (F.B.)
| | - Simona Varrucciu
- Department of Medicine Surgery and Pharmacy, Sassari University, 07100 Sassari, Italy;
| | - Davide Rizzo
- Otolaryngology Division, Azienda Ospedaliera Universitaria di Sassari, 07100 Sassari, Italy; (D.R.); (F.B.)
- Department of Medicine Surgery and Pharmacy, Sassari University, 07100 Sassari, Italy;
| | - Michela Sali
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Department of Laboratory and Infectivology Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Delogu
- Mater Olbia Hospital, 07026 Olbia, Italy; (F.C.); (G.D.)
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Francesco Bussu
- Otolaryngology Division, Azienda Ospedaliera Universitaria di Sassari, 07100 Sassari, Italy; (D.R.); (F.B.)
- Department of Medicine Surgery and Pharmacy, Sassari University, 07100 Sassari, Italy;
| |
Collapse
|
50
|
Lam HYP, Lai MJ, Wang PC, Wu WJ, Chen LK, Fan HW, Tseng CC, Peng SY, Chang KC. A Novel Bacteriophage with the Potential to Inhibit Fusobacterium nucleatum-Induced Proliferation of Colorectal Cancer Cells. Antibiotics (Basel) 2025; 14:45. [PMID: 39858331 PMCID: PMC11760851 DOI: 10.3390/antibiotics14010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/26/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Increasing evidence shows that Fusobacterium nucleatum (F. nucleatum) largely affects colorectal cancer (CRC) growth and progression; therefore, the inhibition of intratumoral F. nucleatum may be one realistic approach to combat CRC. Although antibiotics are helpful in eliminating bacteria, the major problem remains the rise of potential antibiotic-resistant strains and antibiotic-associated adverse effects. Currently, bacteriophage therapy has gained interest because of its high selectivity to bacterial hosts and may become a realistic approach in treating bacteria-associated cancers. METHODS In this study, a new F. nucleatum bacteriophage, ØTCUFN3, was isolated and its biological characteristics were identified. In vitro and in vivo studies were performed to investigate the effect of ØTCUFN3 in combating F. nucleatum-induced CRC growth. RESULTS By applying ØTCUFN3 to F. nucleatum-induced CRC cell lines, p53+/+, and p53-/- isogenic HCT116 cells, our results revealed an inhibition of CRC proliferation and the expression of epithelial-to-mesenchymal transition (EMT) markers. ØTCUFN3 injection also reduced the growth of F. nucleatum-induced mouse xenografts. CONCLUSIONS Our results demonstrated the use of F. nucleatum bacteriophage against CRC, laying the foundation for the future usage of bacteriophage in cancer treatment.
Collapse
Affiliation(s)
- Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan;
- Institute of Medical Science, College of Medicine, Tzu Chi University, Hualien 970374, Taiwan;
| | - Meng-Jiun Lai
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien 970374, Taiwan; (M.-J.L.); (P.-C.W.); (W.-J.W.)
| | - Pin-Chun Wang
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien 970374, Taiwan; (M.-J.L.); (P.-C.W.); (W.-J.W.)
| | - Wen-Jui Wu
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien 970374, Taiwan; (M.-J.L.); (P.-C.W.); (W.-J.W.)
| | - Li-Kuang Chen
- Institute of Medical Science, College of Medicine, Tzu Chi University, Hualien 970374, Taiwan;
- Branch of Clinical Pathology, Department of Laboratory Medicine, Buddhist Tzu Chi General Hospital, Hualien 970473, Taiwan
| | - Hsiang-Wei Fan
- Master Program in Biomedical Science, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan;
| | - Chun-Chieh Tseng
- Department and Graduate Institute of Public Health, Tzu Chi University, Hualien 970374, Taiwan;
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan;
| | - Kai-Chih Chang
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien 970374, Taiwan; (M.-J.L.); (P.-C.W.); (W.-J.W.)
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan
| |
Collapse
|