Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Dietary fibre counters the oncogenic potential of colibactin-producing Escherichia coli in colorectal cancer

Abstract

Diet, microbiome, inflammation and host genetics have been linked to colorectal cancer development; however, it is not clear whether and how these factors interact to promote carcinogenesis. Here we used Il10/ mice colonized with bacteria previously associated with colorectal cancer: enterotoxigenic Bacteroides fragilis, Helicobacter hepaticus or colibactin-producing (polyketide synthase-positive (pks+)) Escherichia coli and fed either a low-carbohydrate (LC) diet deficient in soluble fibre, a high-fat and high-sugar diet, or a normal chow diet. Colonic polyposis was increased in mice colonized with pks+ E. coli and fed the LC diet. Mechanistically, mucosal inflammation was increased in the LC-diet-fed mice, leading to diminished colonic PPAR-γ signalling and increased luminal nitrate levels. This promoted both pks+ E. coli growth and colibactin-induced DNA damage. PPAR-γ agonists or supplementation with dietary soluble fibre in the form of inulin reverted inflammatory and polyposis phenotypes. The pks+ E. coli also induced more polyps in mismatch-repair-deficient mice by inducing a senescence-associated secretory phenotype. Moreover, oncogenic effects were further potentiated by inflammatory triggers in the mismatch-repair-deficient model. These data reveal that diet and host genetics influence the oncogenic potential of a common bacterium.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: LC diet augments colibactin-mediated colon polyposis.
Fig. 2: LC diet augments NC101 colonization and colibactin-mediated DNA damage in the colon.
Fig. 3: The LC diet modulates the composition of faecal microbiota to enrich Enterobacteriaceae in Il10/ mice.
Fig. 4: The effect of the LC diet on NC101 colonization and colonic tumorigenesis is dependent on colonic iNOS activity.
Fig. 5: Soluble-fibre-induced colonic PPAR-γ signalling limits NC101 colonization and tumorigenesis.
Fig. 6: MMR deficiency sensitizes mice to colibactin-induced senescence and CRC development.

Similar content being viewed by others

Data availability

The raw 16S sequencing data generated from mouse stool samples in this study have been submitted to the National Center for Biotechnology Information under BioProject ID PRJNA1198898. Silva database version 138.1 (https://github.com/BenKaehler/readytowear) was used for taxonomy assignment in microbiome analysis. The raw 16S genomic data from the GEM human cohort are publicly accessible under accession PRJNA685746. Requests for additional raw and analysed data should follow the instructions given at http://www.gemproject.ca/data-access/. All submissions will be reviewed by the CCC-GEM Project Operating Committee to ensure that the requested samples and data will not interfere in any way with the intended GEM Project analysis of the nested cohort as per the original GEM Project Study Design and is not a duplication of analysis already ongoing. Those proposals meeting this evaluation will be distributed to all members of the GEM Project Steering Committee (GPSC) for review and open discussion. This review will focus on the global scientific merit of the proposal and will assess the basic scientific merit and the availability of requested samples and data, ensuring that there is no compromise of the original intent of the GEM Project. It would be of value to contact a member of the GPSC who could help sponsor your application. Those projects achieving majority vote of approval at the GPSC will be informed that the GEM Project will provide a letter of support stating that the requested samples or data will be made available to the applicants once the applicant receives funding from a granting agency that applies an independent peer review process to the proposal. The criteria to be used for review of all submissions will include the ‘scientific relevance’ of the proposal and the judged availability of biological material requested. The budget to be requested from a funding agency must allow for any expenses in processing samples or in setting up the appropriate queries of the database. The intent is to allow sufficient time for applicants to consider submission for funding opportunities. Source data are provided with this paper.

References

  1. Morgan, E. et al. Global burden of colorectal cancer in 2020 and 2040: incidence and mortality estimates from GLOBOCAN. Gut 72, 338–344 (2023).

    Article  PubMed  Google Scholar 

  2. Kong, C. et al. Ketogenic diet alleviates colitis by reduction of colonic group 3 innate lymphoid cells through altering gut microbiome. Signal Transduct. Target. Ther. 6, 154 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Kopp, W. How western diet and lifestyle drive the pandemic of obesity and civilization diseases. Diabetes Metab. Syndr. Obes. 12, 2221–2236 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. O’Keefe, S. J. et al. Fat, fibre and cancer risk in African Americans and rural Africans. Nat. Commun. 6, 6342 (2015).

    Article  PubMed  Google Scholar 

  5. Feng, Y. L. et al. Dietary patterns and colorectal cancer risk: a meta-analysis. Eur. J. Cancer Prev. 26, 201–211 (2017).

    Article  PubMed  Google Scholar 

  6. Gaines, S. et al. Western diet promotes intestinal colonization by collagenolytic microbes and promotes tumor formation after colorectal surgery. Gastroenterology 158, 958–970.e2 (2020).

    Article  PubMed  CAS  Google Scholar 

  7. Sonnenburg, J. L. & Backhed, F. Diet–microbiota interactions as moderators of human metabolism. Nature 535, 56–64 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Lee, J. Y. et al. High-fat diet and antibiotics cooperatively impair mitochondrial bioenergetics to trigger dysbiosis that exacerbates pre-inflammatory bowel disease. Cell Host Microbe 28, 273–284.e6 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Seidelmann, S. B. et al. Dietary carbohydrate intake and mortality: a prospective cohort study and meta-analysis. Lancet Public Health 3, e419–e428 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mazidi, M., Katsiki, N., Mikhailidis, D. P., Sattar, N. & Banach, M. Lower carbohydrate diets and all-cause and cause-specific mortality: a population-based cohort study and pooling of prospective studies. Eur. Heart J. 40, 2870–2879 (2019).

    Article  PubMed  CAS  Google Scholar 

  11. Dejea, C. M. et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science 359, 592–597 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Li, S., Konstantinov, S. R., Smits, R. & Peppelenbosch, M. P. Bacterial biofilms in colorectal cancer initiation and progression. Trends Mol. Med. 23, 18–30 (2017).

    Article  PubMed  Google Scholar 

  13. Irrazabal, T. et al. Limiting oxidative DNA damage reduces microbe-induced colitis-associated colorectal cancer. Nat. Commun. 11, 1802 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Arthur, J. C. et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 338, 120–123 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Kostic, A. D. et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 14, 207–215 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Tilg, H., Adolph, T. E., Gerner, R. R. & Moschen, A. R. The intestinal microbiota in colorectal cancer. Cancer Cell 33, 954–964 (2018).

    Article  PubMed  CAS  Google Scholar 

  17. Auvray, F. et al. Insights into the acquisition of the pks island and production of colibactin in the Escherichia coli population. Microb. Genom. 7, 000579 (2021).

    PubMed  PubMed Central  CAS  Google Scholar 

  18. Buc, E. et al. High prevalence of mucosa-associated E. coli producing cyclomodulin and genotoxin in colon cancer. PloS ONE 8, e56964 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Dubinsky, V., Dotan, I. & Gophna, U. Carriage of colibactin-producing bacteria and colorectal cancer risk. Trends Microbiol. 28, 874–876 (2020).

    Article  PubMed  CAS  Google Scholar 

  20. Arthur, J. C. et al. Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer. Nat. Commun. 5, 4724 (2014).

    Article  PubMed  CAS  Google Scholar 

  21. Wilson, M. R. et al. The human gut bacterial genotoxin colibactin alkylates DNA. Science 363, eaar7785 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Cuevas-Ramos, G. et al. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc. Natl Acad. Sci. USA 107, 11537–11542 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Pleguezuelos-Manzano, C. et al. Mutational signature in colorectal cancer caused by genotoxic pks+ E. coli. Nature 580, 269–273 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Dziubanska-Kusibab, P. J. et al. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat. Med. 26, 1063–1069 (2020).

    Article  PubMed  CAS  Google Scholar 

  25. Fearon, E. R. Molecular genetics of colorectal cancer. Annu. Rev. Pathol. 6, 479–507 (2011).

    Article  PubMed  CAS  Google Scholar 

  26. Li, S. K. H. & Martin, A. Mismatch repair and colon cancer: mechanisms and therapies explored. Trends Mol. Med. 22, 274–289 (2016).

    Article  PubMed  CAS  Google Scholar 

  27. Belcheva, A. et al. Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells. Cell 158, 288–299 (2014).

    Article  PubMed  CAS  Google Scholar 

  28. Nougayrede, J. P. et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science 313, 848–851 (2006).

    Article  PubMed  CAS  Google Scholar 

  29. Riva, A. et al. A fiber-deprived diet disturbs the fine-scale spatial architecture of the murine colon microbiome. Nat. Commun. 10, 4366 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Desai, M. S. et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 167, 1339–1353.e21 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Silpe, J. E., Wong, J. W. H., Owen, S. V., Baym, M. & Balskus, E. P. The bacterial toxin colibactin triggers prophage induction. Nature 603, 315–320 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Buffie, C. G. & Pamer, E. G. Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol. 13, 790–801 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Fukuda, S. et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 469, 543–547 (2011).

    Article  PubMed  CAS  Google Scholar 

  34. Hu, Y. et al. Manipulation of the gut microbiota using resistant starch is associated with protection against colitis-associated colorectal cancer in rats. Carcinogenesis 37, 366–375 (2016).

    Article  PubMed  CAS  Google Scholar 

  35. Lu, D. et al. Consumption of dietary fiber with different physicochemical properties during late pregnancy alters the gut microbiota and relieves constipation in sow model. Nutrients 14, 2511 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Greten, F. R. & Grivennikov, S. I. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity 51, 27–41 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Yang, Y., Gharaibeh, R. Z., Newsome, R. C. & Jobin, C. Amending microbiota by targeting intestinal inflammation with TNF blockade attenuates development of colorectal cancer. Nat. Cancer 1, 723–734 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Byndloss, M. X. et al. Microbiota-activated PPAR-gamma signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 357, 570–575 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. McCafferty, D. M. et al. Spontaneously developing chronic colitis in IL-10/iNOS double-deficient mice. Am. J. Physiol. Gastrointest. Liver Physiol. 279, G90–G99 (2000).

    Article  PubMed  CAS  Google Scholar 

  40. Caioni, G. et al. Inflammatory bowel disease: new insights into the interplay between environmental factors and PPARγ. Int. J. Mol. Sci. 22, 985 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Lange, K. et al. Comparison of the effects of five dietary fibers on mucosal transcriptional profiles, and luminal microbiota composition and SCFA concentrations in murine colon. Mol. Nutr. Food Res. 59, 1590–1602 (2015).

    Article  PubMed  CAS  Google Scholar 

  42. Li, M., Pascual, G. & Glass, C. K. Peroxisome proliferator-activated receptor gamma-dependent repression of the inducible nitric oxide synthase gene. Mol. Cell. Biol. 20, 4699–4707 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Stanton, M. V. et al. DIETFITS study (diet intervention examining the factors interacting with treatment success)—study design and methods. Contemp. Clin. Trials 53, 151–161 (2017).

    Article  PubMed  Google Scholar 

  44. Fragiadakis, G. K. et al. Long-term dietary intervention reveals resilience of the gut microbiota despite changes in diet and weight. Am. J. Clin. Nutr. 111, 1127–1136 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lin, Y. J. et al. High citrulline-to-arginine ratio associated with blood pressure abnormalities in children with early chronic kidney disease. Circ. J. 77, 181–187 (2013).

    Article  PubMed  CAS  Google Scholar 

  46. Kato, N. et al. Sensing and processing of DNA interstrand crosslinks by the mismatch repair pathway. Cell Rep. 21, 1375–1385 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Cougnoux, A. et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 63, 1932–1942 (2014).

    Article  PubMed  CAS  Google Scholar 

  48. Wang, B., Kohli, J. & Demaria, M. Senescent cells in cancer therapy: friends or foes?. Trends Cancer 6, 838–857 (2020).

    Article  PubMed  CAS  Google Scholar 

  49. Yousefzadeh, M. J. et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 36, 18–28 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Zhu, W. et al. Editing of the gut microbiota reduces carcinogenesis in mouse models of colitis-associated colorectal cancer. J. Exp. Med. 216, 2378–2393 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Arima, K. et al. Western-style diet, pks island-carrying Escherichia coli, and colorectal cancer: analyses from two large prospective cohort studies. Gastroenterology 163, 862–874 (2022).

    Article  PubMed  CAS  Google Scholar 

  52. Dougherty, M. W. et al. The microbial genotoxin colibactin exacerbates mismatch repair mutations in colorectal tumors. Neoplasia 43, 100918 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Nieminen, U., Jussila, A., Nordling, S., Mustonen, H. & Farkkila, M. A. Inflammation and disease duration have a cumulative effect on the risk of dysplasia and carcinoma in IBD: a case-control observational study based on registry data. Int. J. Cancer 134, 189–196 (2014).

    Article  PubMed  Google Scholar 

  54. Fantini, M. C. & Guadagni, I. From inflammation to colitis-associated colorectal cancer in inflammatory bowel disease: pathogenesis and impact of current therapies. Dig. Liver Dis. 53, 558–565 (2021).

    Article  PubMed  Google Scholar 

  55. Arnott, I. D. et al. Sero-reactivity to microbial components in Crohn’s disease is associated with disease severity and progression, but not NOD2/CARD15 genotype. Am. J. Gastroenterol. 99, 2376–2384 (2004).

    Article  PubMed  Google Scholar 

  56. Kimura, H. et al. Increased expression of an inducible isoform of nitric oxide synthase and the formation of peroxynitrite in colonic mucosa of patients with active ulcerative colitis. Gut 42, 180–187 (1998).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Renouf, M. J., Cho, Y. H. & McPhee, J. B. Emergent behavior of IBD-associated Escherichia coli during disease. Inflamm. Bowel Dis. 25, 33–44 (2019).

    Article  PubMed  Google Scholar 

  58. Khan, A. A. et al. Colorectal cancer–inflammatory bowel disease nexus and felony of Escherichia coli. Life Sci. 180, 60–67 (2017).

    Article  PubMed  CAS  Google Scholar 

  59. Massip, C. et al. Deciphering the interplay between the genotoxic and probiotic activities of Escherichia coli Nissle 1917. PLoS Pathog. 15, e1008029 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Jans, M. et al. Colibactin-driven colon cancer requires adhesin-mediated epithelial binding. Nature 635, 472–480 (2024).

    Article  PubMed  CAS  Google Scholar 

  61. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37, 852–857 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Ewels, P., Magnusson, M., Lundin, S. & Kaller, M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics 32, 3047–3048 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Article  Google Scholar 

  65. Edgar, R. C. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461 (2010).

    Article  PubMed  CAS  Google Scholar 

  66. Rognes, T., Flouri, T., Nichols, B., Quince, C. & Mahe, F. VSEARCH: a versatile open source tool for metagenomics. PeerJ 4, e2584 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Bokulich, N. A. et al. Optimizing taxonomic classification of marker-gene amplicon sequences with QIIME 2’s q2-feature-classifier plugin. Microbiome 6, 90 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Kaehler, B. D. et al. Species abundance information improves sequence taxonomy classification accuracy. Nat. Commun. 10, 4643 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Janssen, S. et al. Phylogenetic placement of exact amplicon sequences improves associations with clinical information. mSystems 3, e00021-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Turpin, W. et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat. Genet. 48, 1413–1417 (2016).

    Article  PubMed  CAS  Google Scholar 

  71. Shatenstein, B., Amre, D., Jabbour, M. & Feguery, H. Examining the relative validity of an adult food frequency questionnaire in children and adolescents. J. Pediatr. Gastroenterol. Nutr. 51, 645–652 (2010).

    Article  PubMed  Google Scholar 

  72. Shatenstein, B., Nadon, S., Godin, C. & Ferland, G. Development and validation of a food frequency questionnaire. Can. J. Diet. Practice Res. 66, 67–75 (2005).

    Article  Google Scholar 

  73. Tavasoli, F. et al. Increased arginase expression and decreased nitric oxide in pig donor lungs after normothermic ex vivo lung perfusion. Biomolecules 10, 300 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Johansson, M. E. & Hansson, G. C. Preservation of mucus in histological sections, immunostaining of mucins in fixed tissue, and localization of bacteria with FISH. Methods Mol. Biol. 842, 229–235 (2012).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Philpott and P. Poussier and the Martin laboratory for their helpful comments. We thank C. Sears (Johns Hopkins University, USA) for providing us the bacterial strain ETBF and A. Baumler (University of California, Davis, USA) for providing indicator strains EcN wild type and cydA napA narG narZ mutant, and plasmids pWSK29 and pWSK129. We also thank C. Gardner, J. Sonnenburg and E. Sonnenburg (Stanford University, USA) for providing us with the human stool samples from their DIETFITS study. This work was funded by the Canadian Institute of Health Research (PJT-173501). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the paper.

Author information

Authors and Affiliations

Authors

Contributions

B.K.T. designed the experiments, performed the research and analysed the data. Y.M., S.R.C., A.N. and W.T. performed the research and analysed the data. C.S., E.O.Y.W. and J.C. performed the research. D.S.G., W.W.N., C.J., K.C. and A.M. supervised. B.K.T., C.J. and A.M. wrote the paper.

Corresponding author

Correspondence to Alberto Martin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Omer Yilmaz and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 The LC diet potentiates NC101-mediated colon carcinogenesis in Il10/ or C57BL/6J WT mice.

(a) Representative H&E-stained microscopic sections, displaying adenomas with dysplastic changes in the colon of mice infected with NC101 and fed LC diet (Related to Fig. 1f). Scale bar = 400 μm. The dashed line outline the adenoma with dysplastic lesions. (b) (left panels) Representative immunohistochemistry sections, displaying nuclear localization of β-catenin in dysplastic lesions and normal colonic tissues of mice infected with NC101 and fed the LC diet. Scale bar = 100 μm. The black dashed line indicates the adenoma with dysplastic lesions and the red arrows indicate the nuclear staining of β-catenin. (right panel) Nuclear β-catenin were quantified based on data in b. Percent of cells positive for nuclear β-catenin were calculated in four randomly chosen fields of view from both dysplastic lesions and normal regions of the tissue per mice. N = 6 in each group; **P = 0.0022. (c) Representative macroscopic colonic image displaying polyps from C57BL/6J wild-type mice orally inoculated with either PBS or the indicated NC101 strains, and fed with either NCD or LC diet for 16 weeks. (d, e) Four-week-old Il10/ mice were orally inoculated with enterotoxigenic Bacteroides fragilis (ETBF) or H. hepaticus (H. hep) and fed with either NCD or LC diet. N = 6 (ETBF groups) and N = 5 (H. hep groups). Colonic polyp numbers were measured 8-week post-infection (d). Colonic polyps counted in d were graded according to their size (e). (f) Schematic illustrating the experimental design and timeline of infection and dietary intervention. Four-week-old Il10/ mice were fed with NCD or LC diet for 4 weeks, followed by treatment with or without cefoxitin before infecting with NC101 or NC101ΔclbP, and fed NCD or LC diet for another 8 weeks. (g) Colonic polyp numbers were measured 8-week post-infection in mice treated as indicated in f. (+)Cef: N = 6 (NCD/ΔclbP and NCD/NC101) and N = 8 (LC diet/ΔclbP and LC diet/NC101); *P = 0.0308, ****P < 0.0001. (-)Cef: N = 7 (LC diet/ΔclbP) and N = 8 (LC diet/NC101); ***P = 0.0005. (h) Colonic polyps counted in g were graded according to their size. (i) Number of adenomas with different grades of dysplasia were assessed based on histopathological features in H&E-stained colonic sections of mice used in Fig. 1j. (j, k) Four-week-old Il10/ mice were orally inoculated with NC101 or NC101ΔclbP, and fed either NCD or Low-Kcal LC diet for 8 weeks. Colonic polyp numbers were measured 8-weeks post-infection (j). NCD: N = 4 (PBS), N = 4 (NC101) and N = 3 (ΔclbP); Low-Kcal LC diet: N = 7 (PBS), N = 8 (NC101) and N = 8 (ΔclbP). **P = 0.0023, ***P = 0.0002, ****P < 0.0001. Colonic polyps counted in j were graded according to their size (k). Data are presented as mean values ± s.e.m. of one to three independent experiments and were analyzed with one-way ANOVA with Sidak’s multiple comparison test (d,g,j) or two-sided Mann-Whitney test (b,right panel of g,i). ns, not significant.

Source data

Extended Data Fig. 2 Effect of specific diets on the gut colonization of CRC-associated bacteria, mucus, and DNA damage in the colon of Il10/ or C57BL/6J WT mice.

(a, b) Four-week-old Il10/ mice were infected orally with ETBF or H. hep and fed with either the NCD or LC diet. B. fragilis (a) and H. hep (b) ΔCt relative to total bacteria was calculated by 16S rRNA qPCR using genomic DNA extracted from faeces collected at 6 weeks post-infection from these mice. N = 5-6 mice in each group. (c) Gut colonization of NC101 was determined by quantifying their numbers in faeces collected at indicated times from C57BL/6J WT mice indicated in Fig. 1g. Data are presented as CFU/g faeces after considering their dilution factors and samples with undetected CFU count are plotted as detection limit. ***P = 0.0004. (d) Transcript level of Muc2 in colonic tissues of mice from Fig. 1b was quantified by qPCR. N = 6 in each group. ***P = 0.0008 (NCD vs. LC diet), ***P = 0.0010 (NCD/NC101 vs. WSD/NC101), ****P < 0.0001. (e) Representative images displaying γH2AX immunofluorescence in colonic sections from Il10/ mice inoculated with either PBS or NC101 and fed with WSD for 8 weeks. Scale bar = 20 μm. (f) Number of γH2AX-positive cells in the colon of mice indicated in e were counted in two crypts per FOV and four FOV per mice. One dot represents the mean γH2AX-positive cells from eight crypts per mice. N = 3 in each group. Data are presented as mean values ± s.e.m. of one to three independent experiments and were analyzed with either one-way ANOVA with Sidak’s multiple comparison test (d) or two-way ANOVA with Sidak’s multiple comparison test (c) or two-sided Mann-Whitney test (a,b,f). ns, not significant.

Source data

Extended Data Fig. 3 Mice fed the LC diet develop signs of chronic mucosal inflammation.

(a, b) Four-week-old Il10/ mice were inoculated with either PBS, NC101 or NC101ΔclbP, and fed with either the NCD, LC diet or WSD for 8 weeks. Caecum weight (a) and colon length (b) were measured 8-week post-infection from mice on indicated treatments. NCD: N = 6 (PBS), N = 6 (NC101) and N = 4 (ΔclbP); LC diet: N = 10 (PBS), N = 13 (NC101) and N = 9 (ΔclbP); WSD: N = 9 (PBS), N = 9 (NC101) and N = 5 (ΔclbP). ****P < 0.0001. (c) Colon image displaying the gross macroscopic appearance (Red circle highlights the inflamed proximal colon). (d, e) Four-week-old C57BL/6J WT mice were orally inoculated with either PBS or the indicated NC101 strains, and fed with either NCD or LC diet for 16 weeks. Caecum weight (d) and colon length (e) were measured 16-week post-infection from mice on indicated treatments. NCD: N = 4 (PBS), N = 5 (NC101) and N = 5 (ΔclbP); LC diet: N = 8 (PBS), N = 10 (NC101) and N = 8 (ΔclbP). *P = 0.0338 (NCD/PBS vs. LC diet/PBS), *P = 0.0297 (NCD/PBS vs. LC diet/NC101). (f) Representative H&E-stained colonic sections displaying microscopic features. Scale bar = 200 μm. (g) Transcript level of indicated inflammatory cytokines were quantified by qPCR in the colon of Il10/ mice orally inoculated with either PBS or NC101 and fed the indicated diets. Relative mRNA expression in comparison to PBS-treated, NCD-fed control mice is presented (set as 1). N = 6 in each group. TNFα: *P = 0.0241 (NCD vs LC diet), **P = 0.0056 (NCD vs. WSD), *P = 0.0361 (NCD vs LC diet/NC101), *P = 0.0371 (NCD vs. WSD/NC101), *P = 0.0357 (NCD/NC101 vs LC diet/NC101), *P = 0.0366 (NCD/NC101 vs. WSD/NC101); IL-1β: **P = 0.0060 (NCD vs. WSD), *P = 0.0486 (NCD vs LC diet/NC101), *P = 0.0423 (NCD vs. WSD/NC101), *P = 0.0377 (NCD/NC101 vs LC diet/NC101), *P = 0.0327 (NCD/NC101 vs. WSD/NC101); IFNγ: *P = 0.0184 (NCD vs LC diet), **P = 0.0048 (NCD vs. WSD), **P = 0.0069 (NCD vs LC diet/NC101), **P = 0.0082 (NCD vs. WSD/NC101), **P = 0.0042 (NCD/NC101 vs LC diet/NC101), **P = 0.0050 (NCD/NC101 vs. WSD/NC101). (h) (left panels) Macrophage (F4/80) in colonic sections of NC101-infected mice fed with the indicated diets. Scale bar = 200 μm. (right panel) Macrophage (F4/80) are quantified based on data in h. N = 3 (NCD), N = 5 (LC diet) and N = 4 (WSD); P = 0.529 (NCD/NC101 vs.LC diet/NC101), P = 0.7871 (NCD/NC101 vs. WSD/NC101). Data are presented as mean values ± s.e.m. of one to three independent experiments and were analyzed with one-way ANOVA with Sidak’s multiple comparison test (a,b,d,e,g,h). ns, not significant.

Source data

Extended Data Fig. 4 LC diet-induced iNOS regulates colon carcinogenesis but not mucosal inflammation.

(a) Nos2 mRNA expression was quantified by qPCR in the colonic epithelial cells (CECs) of mice inoculated with a single dose of either PBS or NC101 and fed the NCD or LC diet for 8 weeks. N = 3 in each group; **P = 0.0030, ***P = 0.0005. (b) Immunofluorescence for iNOS and Epithelial Cell Adhesion Molecule (EpCAM) in colonic sections of mice fed the indicated diets. Scale bar = 50 μm. (c, d) Caecum weight (c) and colon length (d) were measured from mice orally inoculated with either PBS or NC101 and fed on LC diet with or without L-Nil for 8 weeks. N = 7 (LC diet), N = 6 (LC diet/L-Nil), N = 9 (LC diet/NC101) and N = 5 (LC diet/NC101/L-Nil). (e) Transcript level of indicated inflammatory cytokines were quantified by qPCR in the colon of mice treated as indicated above. Relative mRNA expression in comparison to PBS-treated, NCD-fed control mice is presented (set as 1).TNFα: *P = 0.0395 (NCD vs. LC diet), *P = 0.0198 (NCD vs. LC diet/NC101); IL-1β: **P = 0.0030 (NCD vs. LC diet), *P = 0.0429 (NCD vs. LC diet/NC101); IFNγ: P = 0.0521 (NCD vs. LC diet), P = 0.0248 (NCD vs. LC diet/NC101); IL-6: *P = 0.0457 (NCD vs. LC diet), *P = 0.0350 (NCD vs. LC diet/NC101); IL-17: **P = 0.0089 (NCD vs. LC diet), **P = 0.0029 (NCD vs. LC diet/NC101); IL-22: ***P = 0.0004 (NCD vs. LC diet), ***P = 0.0004 (NCD vs. LC diet/NC101). (f) Colonic polyps counted in Fig. 4k were graded according to their size. Data are presented as mean values ± s.e.m. from two independent experiments and were analyzed with one-way ANOVA with Sidak’s multiple comparison test (a,c,d,e). ns, not significant.

Source data

Extended Data Fig. 5 LC diet abrogates epithelial PPAR-γ signaling to induce colonic nitrates and tumorigenesis.

(a) PPAR-γ and Angptl4 mRNA expression as quantified by qPCR in colonic epithelial cells (CECs) of mice orally inoculated with either PBS or NC101 and fed with NCD or LC diet for 8 weeks. N = 3 in each group. PPAR-γ: **P = 0.0022 (NCD vs. LC diet), **P = 0.0036 (NCD vs. LC diet/NC101), *P = 0.0118 (NCD/NC101 vs. LC diet/NC101); Angptl4: **P = 0.0037 (NCD vs. LC diet), **P = 0.0079 (NCD vs. LC diet/NC101), *P = 0.0153 (NCD/NC101 vs. LC diet/NC101). (b) PPAR-γ and Nos2 mRNA expression quantified by qPCR in colon of mice inoculated with either PBS or NC101 and fed the LC diet with or without rosiglitazone (Rosi) for 8 weeks. PPAR-γ: N = 6 (NCD and LC diet) and N = 8 (LC diet/Rosi, LC diet/NC101 and LC diet/NC101/Rosi); P < 0.0001. Nos2: N = 6 (NCD and LC diet), N = 8 (LC diet/Rosi and LC diet/NC101) and N = 7 (LC diet/NC101/Rosi); ***P = 0.0002, P < 0.0001, **P = 0.0019 (LC diet vs. LC diet/Rosi), **P = 0.0037 (LC diet/NC101 vs. LC diet/NC101/Rosi). (c) Colonic transcript levels of indicated inflammatory cytokines from indicated treatment groups were quantified by qPCR. Relative mRNA expression in comparison to PBS-treated, NCD-fed control mice (set as 1) is presented. TNFα: ****P < 0.0001, **P = 0.0035 (LC diet vs. LC diet/Rosi), ***P = 0.0008 (LC diet/NC101 vs. LC diet/NC101/Rosi); IL-1β: **P = 0.0011 (NCD vs. LC diet), ***P = 0.0007 (NCD vs. LC diet/NC101), **P = 0.0061 (LC diet vs. LC diet/Rosi), **P = 0.0100 (LC diet/NC101 vs. LC diet/NC101/Rosi); IFNγ: **P = 0.0019 (NCD vs. LC diet), *P = 0.0121 (NCD vs. LC diet/NC101), *P = 0.0239 (LC diet vs. LC diet/Rosi), P = 0.0584 (LC diet/NC101 vs. LC diet/NC101/Rosi); IL-6: ***P = 0.0010 (NCD vs. LC diet), **P = 0.0100 (NCD vs. LC diet/NC101), **P = 0.0012 (LC diet vs. LC diet/Rosi), P = 0.0552 (LC diet/NC101 vs. LC diet/NC101/Rosi); IL-17: **P = 0.0062 (NCD vs. LC diet), ****P < 0.0001 (NCD vs. LC diet/NC101), *P = 0.0462 (LC diet vs. LC diet/Rosi), **P = 0.0031 (LC diet/NC101 vs. LC diet/NC101/Rosi); IL-22: ****P < 0.0001, ***P = 0.0005 (LC diet vs. LC diet/Rosi), **P = 0.0024 (LC diet/NC101 vs. LC diet/NC101/Rosi). (d) Colonic polyps counted in Fig. 5e were graded according to their size. Data are presented as mean values ± s.e.m. of two independent experiments and were analyzed with one-way ANOVA with Sidak’s multiple comparison test (a,b,c). ns, not significant.

Source data

Extended Data Fig. 6 Inulin supplementation rescues LC diet-mediated effects on colonic inflammation and CRC development.

(a) Transcript level of Nos2 was quantified by qPCR in the colonic tissues of mice fed either the NCD or LC diet with or without inulin for 8 weeks. Relative mRNA expression in comparison to PBS-treated, NCD-fed control mice (set as 1) is presented. N = 6 (NCD), N = 4 (NCD/inulin), N = 5 (LC diet), N = 6 (LC diet/inulin); ***P = 0.0004, **P = 0.0076. (b, c) Caecum weight (b) and colon length (c) were measured in mice orally inoculated with PBS or NC101 and fed either the NCD or LC diet with or without inulin for 8 weeks. N = 6 (NCD), N = 4 (NCD/inulin), N = 5 (LC diet), N = 11 (LC diet/inulin), N = 6 (LC diet/NC101) and N = 10 (LC diet/NC101/inulin). **P = 0.0041, *P = 0.0280 (b) and *P = 0.0141 (c). (d) Colonic transcript levels of indicated inflammatory cytokines from indicated treatment groups in a were quantified by qPCR. Relative mRNA expression in comparison to PBS-treated, NCD-fed control mice (set as 1) is presented. TNFα: *P = 0.0173; IL-1β: **P = 0.0023; IFNγ: ***P = 0.0005; IL-6: **P = 0.0075; IL-17: *P = 0.0402; IL-22: ****P < 0.0001, **P = 0.0010. (e, f) Faecal samples at 6-weeks from indicated group of mice used in Fig. 5i were collected and processed for microbiota analysis by 16S rRNA gene sequencing. N = 5 (LC diet) and N = 6 (LC diet/inulin, LC diet/NC101, LC diet/NC101/inulin). Bray-Curtis PCoA plot for beta diversity (e) and taxonomic composition (f) of faecal microbiota of mice with indicated treatments were shown. (g) Colonic polyps counted in Fig. 5i were graded according to their size. Data are presented as mean values ± s.e.m. of two independent experiments and were analyzed with one-way ANOVA with Sidak’s multiple comparison test (a,b,c,d) or ADONIS2 (e). ns, not significant.

Source data

Extended Data Fig. 7 Dietary pattern low in carbohydrates promotes colonic inflammation and E. coli abundance in human cohort.

Total dietary energy (a), total dietary fat (b), total dietary protein (c), total dietary carbohydrate (d), and total dietary fiber (e) intake was calculated by food-intake assessment of subjects on ad libitum diet lower in carbohydrate in the DIETFITS study at baseline, 3 months, and 12 months. N = 20. ***P = 0.0007, **P = 0.0056 (a); ****P < 0.0001 (d); ***P = 0.0001, *P = 0.0484 (e). (f, g) Faecal calprotectin was quantified by ELISA (f), and E. coli enrichment was measured by qPCR (g) in stool samples of these subjects. **P = 0.0083, *P = 0.0296 (f); **P = 0.0073 (g). Data were analyzed using two-tailed Wilcoxon matched-pairs signed rank test. (h) Fiber intake was obtained from StatsCan data of items within a food frequency questionnaire, and intestinal inflammation was measured via stool-derived faecal calprotectin (log10 transformation). N = 4,143 CCC-GEM participants. A Pearson correlation was performed, R represents the correlation coefficient, and the P value is for the statistical test to reject the hypothesis of lack of correlation (R = 0). A generalized estimating equation general linear model was also performed, the effect (beta) of dietary fiber was -0.0186, P = 0.00036, while the effect (beta) of dietary fiber was -0.0193, P = 0.000022 when adjusted for age, sex, and familial cluster. The Pearson coefficient of determination (R2) was -0.002809. (i) The relative abundance of E. coli (cube root transformed) was obtained from shotgun sequencing of stool microbiome, and intestinal inflammation was measured via stool-derived faecal calprotectin (log10 transformation). N = 286 CCC-GEM participants. A Pearson correlation was performed, R represents the correlation coefficient, and the P value is for the statistical test to reject the hypothesis of lack of correlation (R = 0). A generalized estimating equation general linear model was also performed, the effect (beta) of faecal calprotectin was 0.167, P = 0.015, while the effect (beta) of faecal calprotectin was 0.167, P = 0.015, when adjusted for age, sex, and familial cluster. The Pearson coefficient of determination (R2) was 0.042436. (j) The ratio of arginine to citrulline was derived from stool-based metabolomics data, and intestinal inflammation was measured via stool-derived faecal calprotectin (log10 transformation). N = 83 CCC-GEM participants. A Pearson correlation was performed, R represents the correlation coefficient, and the P value is for the statistical test to reject the hypothesis of lack of correlation (R = 0). A beta regression was also performed, the effect (beta) of faecal calprotectin was -0.435, P = 0.0379, while the effect (beta) of faecal calprotectin was -0.414, P = 0.0454 when adjusted for age and sex. The Pearson coefficient of determination (R2) was -0.047961. ns, not significant.

Source data

Extended Data Fig. 8 Specific DNA repair-deficiencies in susceptibility to colibactin-induced CRC development.

(a-c) Four-week-old APCmin mice with Msh2-proficiency or -deficiency in their intestinal epithelial cells were orally inoculated with either PBS, H. hepaticus or CDT-mutant H. hepaticus (ΔCDT). Colonic (a) and small intestinal (SI) (b) polyp numbers were measured 8-week post-infection. Msh2fl/fl/Msh2fl/+ Villin-Cre APCmin mice: N = 5 (PBS), N = 5 (NC101) and N = 3 (ΔclbP); Msh2fl/fl Villin-Cre APCmin mice: N = 8 (PBS), N = 9 (NC101) and N = 6 (ΔclbP). (c) Colonic polyps counted in a were graded according to their size. (d-f) Same as (a-c), except that four-week-old 53BP1-proficient or -deficient mice with APCmin background were used, and polyp numbers were measured 8-week post-infection. 53BP+/+ APCmin mice: N = 7 (PBS), N = 10 (NC101) and N = 8 (ΔclbP); 53BP/ APCmin mice: N = 8 (PBS), N = 10 (NC101) and N = 8 (ΔclbP). (g-i) Same as (a-c), except that four-week-old FancC-proficient or -deficient mice with APCmin background were used. FancC+/ APCmin mice: N = 10 (PBS), N = 8 (NC101) and N = 9 (ΔclbP); FancC/ APCmin mice: N = 4 (PBS), N = 4 (NC101) and N = 4 (ΔclbP). Data are presented as mean values from at least two independent experiments and were analyzed with two-way ANOVA with Dunnett’s multiple comparison test (a,b,d,e,g,h). ns, not significant.

Source data

Extended Data Fig. 9 MMR-deficiency increases susceptibility to colibactin-induced CRC development.

(a) Relative γH2AX intensity is presented for colonic sections counted for γH2AX-positive cells in Fig. 6d. N = 4 in each group. The mean fluorescence intensity (MFI) of γH2AX and the number of cells per FOV was calculated using ImageJ. MFI of γH2AX/cell was calculated by dividing MFI with number of cells/FOV. Relative γH2AX intensity in comparison to PBS-treated, Msh2-proficient control mice is presented (set as 1). *P = 0.0117. (b, c) Four-week-old APCmin mice with Msh2-proficiency or -deficiency in their intestinal epithelial cells were orally inoculated with either PBS, NC101 or NC101ΔclbP (indicated groups from mice used in Fig. 6a). Colon (b) and caecum weight (c) were measured 8-week post-infection. Msh2fl/fl APCmin mice: N = 5 (PBS), N = 7 (NC101) and N = 4 (ΔclbP); Msh2fl/fl Villin-Cre APCmin mice: N = 8 (PBS), N = 11 (NC101) and N = 7 (ΔclbP). (d, e) Colonic transcript levels of indicated inflammatory cytokines 2-week (d) and 8-week (e) post-infection were quantified by qPCR in mice as indicated. Relative mRNA expression in comparison to PBS-treated, Msh2-proficient control mice is presented (set as 1). 2-week: N = 4 in each group; TNFα: *P = 0.0168 (PBS vs. NC101), *P = 0.0150 (PBS vs. ΔclbP); IL-6: *P = 0.0231 (PBS vs. NC101), *P = 0.0283 (PBS vs. ΔclbP); CXCL1: Msh2fl/fl APCmin mice ***P = 0.0008 (PBS vs. NC101), ***P = 0.0003 (PBS vs. ΔclbP); CXCL1: Msh2fl/fl Villin-Cre APCmin mice *P = 0.0156 (PBS vs. NC101), **P = 0.0031 (PBS vs. ΔclbP). 8-week: Msh2fl/fl APCmin mice: N = 5 (PBS), N = 5 (NC101) and N = 4 (ΔclbP); Msh2fl/fl Villin-Cre APCmin mice: N = 4 (PBS), N = 7 (NC101) and N = 7 (ΔclbP); TNFα: *P = 0.0196; IL-6: *P = 0.0155; CXCL1: *P = 0.0170; IL-1β: *P = 0.0478. (f) Representative images for cleaved caspase-3 immunofluorescence in colonic sections of indicated groups of mice. Scale bar = 50 μm. (g) Colonic transcript levels of p16INK4A 8-week post-infection were quantified by qPCR. Relative mRNA expression in comparison to PBS-treated, Msh2-proficient control mice is presented (set as 1). Msh2fl/fl APCmin mice: N = 5 (PBS), N = 5 (NC101) and N = 4 (ΔclbP); Msh2fl/fl Villin-Cre APCmin mice: N = 4 (PBS), N = 7 (NC101) and N = 6 (ΔclbP); **P = 0.0012. (h) Schematic illustrating the experimental design and timeline of infection and senolytic (Fisetin) treatment. (i) Representative macroscopic colonic image displaying polyps. (j) Colonic polyps counted in Fig. 6g were graded according to their size. (k) Colonic polyps counted in Fig. 6h were graded according to their size. (l) Colonic polyps counted in Fig. 6i were graded according to their size. Data are presented as mean values ± s.e.m. from at least two independent experiments and were analyzed with two-way ANOVA with Tukey’s multiple comparison test (a) or Dunnett’s multiple comparison test (b,c,d,e,g). ns, not significant.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thakur, B.K., Malaise, Y., Choudhury, S.R. et al. Dietary fibre counters the oncogenic potential of colibactin-producing Escherichia coli in colorectal cancer. Nat Microbiol 10, 855–870 (2025). https://doi.org/10.1038/s41564-025-01938-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-025-01938-4

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer