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:

Microbiome dysbiosis, neutrophil recruitment and mesenchymal transition of mesothelial cells promotes peritoneal metastasis of colorectal cancer

Abstract

Peritoneal metastasis (PM) is common in colorectal cancer (CRC), yet its underlying mechanisms are poorly understood. Here, we explored the transcriptional profile of CRC, PM and adjacent tissues revealing key players that facilitate PM. Single-cell analysis of 48 matched samples from 12 patients revealed that remodeling of malignant cells and the tumor microenvironment promotes CRC progression and metastasis. Multiplexed imaging confirmed depletion in PM by enrichment in CRC tissues of neutrophils associated with mucosal immunity disruption, intestinal microbiota dysbiosis and mesenchymal transition of both cancerous and mesothelial cells. Functional analyses in cell lines, organoids and in vivo models demonstrated that dysbiosis promoted inflammation and protumor neutrophil recruitment, while coupled mesenchymal transition of malignant and mesothelial cells disrupted the stromal structure and increased cancer cell invasiveness. Our findings suggest that targeting mesothelial cells and tumor microenvironment remodeling may offer therapeutic strategies for CRC-PM.

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: Study design, single-cell atlas and compositional changes in immune and stromal cell subclusters.
Fig. 2: Transcriptomic differences between cancer cells in peritoneal metastases and those in colorectal cancer.
Fig. 3: The enrichment of neutrophils in CRC is associated with dysbiosis.
Fig. 4: The depletion of neutrophils in PM is attributed to the loss of CXCL14 expression and the diversity of the neutrophils.
Fig. 5: Mesothelial-to-mesenchymal transition.
Fig. 6: Anti-MSLN treatment strategy to inhibit peritoneal metastasis of colorectal cancer.

Similar content being viewed by others

Data availability

All raw and processed sequencing data generated as part of this study have been deposited at the Genome Sequence Archive of the BIG Data Center at the Beijing Institution of Genomics, Chinese Academy of Sciences (accession no. HRA003293) and are publicly accessible. The published data used for validation or comparison in this study were retrieved from the NCBI Gene Expression Omnibus database under accession codes GSE17834, GSE132257, GSE132465 and GSE144735. In this study, we used the GRCh38 reference genome for data analysis (https://www.10xgenomics.com/support/software/cell-ranger/downloads). Source data for Figs. 16 and Extended Data Figs. 16 have been provided as Source Data files. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

No algorithm or software was generated for this study. The code for reproducing major figures is available on GitHub (https://github.com/cubense/PM-in-CRC-PROJECT). Any additional information required to reanalyze the data reported in this article is available from the corresponding author upon request.

References

  1. Sung, H. et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 71, 209–249 (2021).

    Article  PubMed  Google Scholar 

  2. Biller, L. H. & Schrag, D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA 325, 669–685 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Segelman, J. et al. Incidence, prevalence and risk factors for peritoneal carcinomatosis from colorectal cancer. Br. J. Surg. 99, 699–705 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Enblad, M., Graf, W. & Birgisson, H. Risk factors for appendiceal and colorectal peritoneal metastases. Eur. J. Surg. Oncol. 44, 997–1005 (2018).

    Article  PubMed  Google Scholar 

  5. van Gestel, Y. R. et al. Metachronous peritoneal carcinomatosis after curative treatment of colorectal cancer. Eur. J. Surg. Oncol. 40, 963–969 (2014).

    Article  PubMed  Google Scholar 

  6. Kranenburg, O., van der Speeten, K. & de Hingh, I. Peritoneal metastases from colorectal cancer: defining and addressing the challenges. Front. Oncol. 11, 650098 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Klaver, C. E. et al. Recommendations and consensus on the treatment of peritoneal metastases of colorectal origin: a systematic review of national and international guidelines. Colorectal Dis. 19, 224–236 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Elias, D. et al. Complete cytoreductive surgery plus intraperitoneal chemohyperthermia with oxaliplatin for peritoneal carcinomatosis of colorectal origin. J. Clin. Oncol. 27, 681–685 (2009).

    Article  PubMed  Google Scholar 

  9. Sluiter, N. R. et al. Metachronous peritoneal metastases after adjuvant chemotherapy are associated with poor outcome after cytoreduction and HIPEC. Ann. Surg. Oncol. 25, 2347–2356 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Lenos, K. J. et al. Molecular characterization of colorectal cancer related peritoneal metastatic disease. Nat. Commun. 13, 4443 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, B. et al. Differential pre-malignant programs and microenvironment chart distinct paths to malignancy in human colorectal polyps. Cell 184, 6262–6280 e6226 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Liu, Y. et al. Immune phenotypic linkage between colorectal cancer and liver metastasis. Cancer Cell 40, 424–437 e425 (2022).

    Article  PubMed  Google Scholar 

  13. Pelka, K. et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell 184, 4734–4752 e4720 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhang, L. et al. Single-cell analyses inform mechanisms of myeloid-targeted therapies in colon cancer. Cell 181, 442–459 e429 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  CAS  PubMed  Google Scholar 

  16. Lee, H. O. et al. Lineage-dependent gene expression programs influence the immune landscape of colorectal cancer. Nat. Genet. 52, 594–603 (2020).

    Article  CAS  PubMed  Google Scholar 

  17. Kienes, I., Johnston, E. L., Bitto, N. J., Kaparakis-Liaskos, M. & Kufer, T. A. Bacterial subversion of NLR-mediated immune responses. Front. Immunol. 13, 930882 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Brubaker, S. W., Bonham, K. S., Zanoni, I. & Kagan, J. C. Innate immune pattern recognition: a cell biological perspective. Annu. Rev. Immunol. 33, 257–290 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Galeano Nino, J. L. et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature 611, 810–817 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Chen, K., Magri, G., Grasset, E. K. & Cerutti, A. Rethinking mucosal antibody responses: IgM, IgG and IgD join IgA. Nat. Rev. Immunol. 20, 427–441 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. McDonald, B. D., Jabri, B. & Bendelac, A. Diverse developmental pathways of intestinal intraepithelial lymphocytes. Nat. Rev. Immunol. 18, 514–525 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Huus, K. E., Petersen, C. & Finlay, B. B. Diversity and dynamism of IgA-microbiota interactions. Nat. Rev. Immunol. 21, 514–525 (2021).

    Article  CAS  PubMed  Google Scholar 

  23. Martens, E. C., Neumann, M. & Desai, M. S. Interactions of commensal and pathogenic microorganisms with the intestinal mucosal barrier. Nat. Rev. Microbiol. 16, 457–470 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Perez-Lopez, A., Behnsen, J., Nuccio, S. P. & Raffatellu, M. Mucosal immunity to pathogenic intestinal bacteria. Nat. Rev. Immunol. 16, 135–148 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Janney, A., Powrie, F. & Mann, E. H. Host-microbiota maladaptation in colorectal cancer. Nature 585, 509–517 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Cao, X. et al. Molecular cloning and characterization of a novel CXC chemokine macrophage inflammatory protein-2γ chemoattractant for human neutrophils and dendritic cells. J. Immunol. 165, 2588–2595 (2000).

    Article  CAS  PubMed  Google Scholar 

  27. Jaillon, S. et al. Neutrophil diversity and plasticity in tumour progression and therapy. Nat. Rev. Cancer 20, 485–503 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Nieto, M. A., Huang, R. Y., Jackson, R. A. & Thiery, J. P. EMT: 2016. Cell 166, 21–45 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Lambert, A. W. & Weinberg, R. A. Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat. Rev. Cancer 21, 325–338 (2021).

    Article  CAS  PubMed  Google Scholar 

  30. Williams, E. D., Gao, D., Redfern, A. & Thompson, E. W. Controversies around epithelial-mesenchymal plasticity in cancer metastasis. Nat. Rev. Cancer 19, 716–732 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yanez-Mo et al. Peritoneal dialysis and epithelial-to-mesenchymal transition of mesothelial cells. N. Engl. J. Med. 348, 403–413 (2003).

    Article  PubMed  Google Scholar 

  32. Si, M. et al. Inhibition of hyperglycolysis in mesothelial cells prevents peritoneal fibrosis. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aav5341 (2019).

  33. Kent, L. N. & Leone, G. The broken cycle: E2F dysfunction in cancer. Nat. Rev. Cancer 19, 326–338 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Hirata, H. et al. Actomyosin bundles serve as a tension sensor and a platform for ERK activation. EMBO Rep. 16, 250–257 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Aragona, M. et al. Mechanisms of stretch-mediated skin expansion at single-cell resolution. Nature 584, 268–273 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tsai, J. M. et al. Surgical adhesions in mice are derived from mesothelial cells and can be targeted by antibodies against mesothelial markers. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aan6735 (2018).

  37. Cerise, A., Bera, T. K., Liu, X., Wei, J. & Pastan, I. Anti-mesothelin recombinant immunotoxin therapy for colorectal cancer. Clin. Colorectal Cancer 18, 192–199 e191 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Hassan, R. et al. Mesothelin immunotherapy for cancer: ready for prime time? J. Clin. Oncol. 34, 4171–4179 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Beumer, J. & Clevers, H. Cell fate specification and differentiation in the adult mammalian intestine. Nat. Rev. Mol. Cell Biol. 22, 39–53 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Schmitt, M. & Greten, F. R. The inflammatory pathogenesis of colorectal cancer. Nat. Rev. Immunol. 21, 653–667 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. Huang, H. et al. Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell 40, 656–673 e657 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Koyama, Y. et al. Mesothelin/mucin 16 signaling in activated portal fibroblasts regulates cholestatic liver fibrosis. J. Clin. Invest. 127, 1254–1270 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Xue, R. et al. Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature 612, 141–147 (2022).

    Article  CAS  PubMed  Google Scholar 

  44. Salcher, S. et al. High-resolution single-cell atlas reveals diversity and plasticity of tissue-resident neutrophils in non-small cell lung cancer. Cancer Cell 40, 1503–1520 e1508 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, L. et al. Single-cell RNA-seq analysis reveals BHLHE40-driven pro-tumour neutrophils with hyperactivated glycolysis in pancreatic tumour microenvironment. Gut https://doi.org/10.1136/gutjnl-2021-326070 (2022).

    Article  PubMed  Google Scholar 

  46. Chang, Y. et al. Engineering chimeric antigen receptor neutrophils from human pluripotent stem cells for targeted cancer immunotherapy. Cell Rep. 40, 111128 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kalafati, L. et al. Innate immune training of granulopoiesis promotes anti-tumor activity. Cell 183, 771–785 e712 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Mo, S. et al. Patient-derived organoids from colorectal cancer with paired liver metastasis reveal tumor heterogeneity and predict response to chemotherapy. Adv. Sci. 9, e2204097 (2022).

    Article  Google Scholar 

  49. Mayakonda, A., Lin, D. C., Assenov, Y., Plass, C. & Koeffler, H. P. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 28, 1747–1756 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Roth, A. et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods 11, 396–398 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Liu, M. et al. MesKit: a tool kit for dissecting cancer evolution of multi-region tumor biopsies through somatic alterations. Gigascience https://doi.org/10.1093/gigascience/giab036 (2021).

  52. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinform. 14, 7 (2013).

    Article  Google Scholar 

  54. Wagner, A. et al. Metabolic modeling of single Th17 cells reveals regulators of autoimmunity. Cell 184, 4168–4185 e4121 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jin, S. et al. Inference and analysis of cell–cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lange, M. et al. CellRank for directed single-cell fate mapping. Nat. Methods 19, 159–170 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the grants of Science and Technology Commission of Shanghai Municipality (no. 23XD1400700), the National Natural Science Foundation of China (nos. 82241230, T2125002, 82173133, 82341007, 82103154, 82373090, 82003317, 82103554, 92259303), the National Key Research and Development Program of China (2022YFC2504602), Beijing Natural Science Foundation (Z220014), Shanghai Municipal Health Commission (no. 2022XD012) and the New Cornerstone Science Foundation through the XPLORER PRIZE.

Author information

Authors and Affiliations

Authors

Contributions

G.C. and F.B. conceived and designed the project. A.G. made suggestions to the frame of the data analysis and manuscript. Q.L., L.H., W.L. and W.D. collected the human samples and clinical information. G.C., Q.L., R.W., Y. Xu and S.C. performed the surgical operation. Q.L. and L.H. performed the scRNA-seq experiments. Y. Xiao performed bioinformatic analyses. Q.L. and L.H. performed IHC and IF experiments. W.L. and L.H. performed FISH and 16S rRNA-seq experiments. H.F. performed functional experiments on neutrophils. L.H. performed functional experiments on mesothelial cells. L.H., W.D., R.W. and W.L. constructed the mouse models and analyzed the in vivo data. Q.L., Y. Xiao, L.H., W.L., W.D., H.F., R.W., Y. Xu, S.C., A.G., F.B. and G.C. discussed and interpreted the data. Y. Xiao wrote the paper with input from all authors. G.C., F.B. and A.G. reviewed and finalized the paper. G.C. and F.B. supervised the project.

Corresponding authors

Correspondence to Ajay Goel, Fan Bai or Guoxiang Cai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Iain Tan, Louis Vermeulen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 A single-cell landscape of primary tumors and peritoneal metastases in colorectal cancer.

a, Expression levels of signature genes in major cell clusters and subclusters. b, Comparison of the proportions of major cell clusters in normal peritoneal tissues, peritoneal metastases, normal colonic tissues, and CRC tissues from 12 patient. Boxes indicate medians and interquartile ranges (IQRs), and whiskers indicate the minimum and maximum values; two-tailed Wilcoxon test. c, Expression of representative marker genes in four subsets of macrophages within myeloid cells. d, Selected pathway activities in different subsets of macrophages. The pathway activities were assessed per cell using gene set variation analysis (GSVA).e, UMAP visualization of RNA velocity of seven subsets of macrophages and monocytes in peritoneal metastases. f, mIHC validation of macrophage subset Macrophage-FN1 present in peritoneal metastasis tumor tissues. Scale bars, 20 μm. Images are representative of n = 12 experiments. g, Spatial distribution(representative images of n = 12 experiments) of RGS5+ Pericyte cells, ACTA2+ fibroblast cells, CD66+ neutrophils, CD1C + cDC2 cells and TPASB1+ mast cells in CRC tissue and PM tissue. Scale bars, 20 μm. h, Quantitative proportions of RGS5+ Pericytes, TPASB1+ mast cells, ACTA2+ fibroblasts and CD1C + cDC2 cells in CRC tissue vs. PM tissue by quantification mIHC in 10 patients with PM and without liver metastases. For panels a-d and h: CRC, n = 12 patients; PM, n = 12 patients; normal colon, n = 12 patients; normal peritoneal, n = 9 patients. unpaired two-sides t-test. Data are presented as mean values +/- SD.

Source data

Extended Data Fig. 2 Whole-exon sequencing and CNV subclone analysis of malignant cells.

a, Genomic landscape of 24 samples from 11 patients. b, CNV profiles inferred from patient DNA whole-exome sequencing (top) and scRNA-seq (bottom). The heatmap color shows the log2 CNV ratio: red, amplifications; blue, deletions. CNA inference in malignant cells using the subclone mode; rows correspond to malignant cells ordered by tumor and within a clone cluster tree in the left panel for each patient. The legend color shows the tissue types of malignant cells: blue, CRC; red, PM (peritoneal metastases); orange, LM (liver metastasis). c, Phylogenetic trees were constructed based on the mutations observed in each patient. Scale bar represents the number of mutations.For panels ac: CRC, n = 12 patients; PM, n = 12 patients; normal colon, n = 12 patients; normal peritoneal, n = 9 patients.

Source data

Extended Data Fig. 3 Representative malignant programs and the depletion of two mucosal immune components in CRC.

a, Spatial distribution(representative images of n = 12 experiments) of the expression of COL1A2 and SPP1 in PM and CRC cancer cells. Scale bars, 20 μm. Images are representative of n = 12 patients experiments. b,Violin plots displaying the expression of two cell cycle programs in three cell cycle phases identified by Seurat. c, Expression of 12 patient- or sample-specific programs in UMAPs of malignant cells. d, Ridgeline plots showing the expression of four colon lineage cluster programs in subsets of normal epithelial cells from two scRNA-seq cohort datasets of CRC. e, Density plot showing the distribution of plasma and B cell clusters (left panel) and T/NK clusters (right panel) in different tissue types. f, Proportions of IGHA plasma cells (top) and IEL_GDT cells (bottom) in normal colonic tissue vs CRCs in two external scRNA-seq datasets.Unpaired two-sides Wilcoxon test. Boxes indicate medians and interquartile ranges (IQRs), and whiskers indicate the minimum and maximum values. g, The top 10 genera identified by 16S rRNA sequencing in colorectal cancer and normal colon tissues. h, The paired comparison between cancer and normal tissues of 26 external validation of CRC patients reveals significant differences in three genera among the top 10 identified genera by 16S rRNA sequencing. Boxes indicate medians and interquartile ranges (IQRs), and whiskers indicate the minimum and maximum values. p values were calculated using actual genera counts by paired Wilcoxon rank-sum tests. i, Correlations across neutrophils, IEL_GDT, IgA plasma cell abundance and bacteria counts in CRC samples. Correlations were calculated using Pearson’s r. (CRC, n = 26 patients; normal colon, n = 26 patients). P < 0.0001.

Source data

Extended Data Fig. 4 Dysbiosis of intestinal bacteria and the recruitment of neutrophils in normal peritoneal tissue.

a, Dot plot depicting the selected ligand–receptor interactions between neutrophils and other cells in PM vs normal peritoneal tissue. Communication probability and p values were calculated using CellChat.All p value < 0.0001. The significance of each ligand–receptor interaction score is assessed using a random permutation test. b, Expression of CXCL14 in stromal cells across different tissues. c, Migration of dHL-60 cells in a chemotaxis assay; the y axis represents the ratio of cell counts in the supernatant of the CXCL14 group in comparison with the PBS group; p values were calculated using an unpaired two-side t-test, biological replicate=3. Boxes indicate medians and interquartile ranges (IQRs), and whiskers indicate the minimum and maximum valuesd, UMAP showing the expression levels of selected signature genes in subsets of neutrophils. e, Selected pathway activities among the different neutrophil subsets. Pathway activities were scored per cell using gene set variation analysis (GSVA). Data are presented as mean values +/- SD. f, Functional enrichment (−log10 of FDR-adjusted p value, two-sides hypergeometric test) of differential genes in neutrophils in normal peritoneal tissues and CRC tissues using Metascape, with the selected pathways labeled. g, Top 10 differentially active transcription factors in each tissue.For panels a-g: CRC, n = 12 patients; PM, n = 12 patients; normal colon, n = 12 patients; normal peritoneal, n = 9 patients.

Source data

Extended Data Fig. 5 Heterogeneity of neutrophils and validation using in vitro models.

a, Colony formation assay, in which control dHL-60 cells, NFKB1-overexpressing dHL-60 cells, and STAT4-overexpressing dHL-60 cells were cultured in the upper chamber, and SW480 (left upper) or HC116 cells (left bottom) were cultured in the lower chamber. Images are representative of n = 3 experiments.Bar plot showing the number of SW480 cells (right upper) or HC116 cells (right bottom) in the colony formation assay. p values were calculated using an unpaired two-sides t-test, n=3 biological replicate.b, Migration assay, SW480 cells or HCT116 cells co-cultured with control dHL-60 cells, NFKB1-overexpressing dHL-60 cells, and STAT4-overexpressing dHL-60 cells for 3 days. Their migration capacity was assessed using the Transwell assay. Images are representative of n = 3 experiments. p values were calculated using an unpaired two-sides t-test, biological replicate=3. c, Cytokine assays showing the relative concentration of cytokines in cancer cells co-cultured with NFKB1-overexpressing dHL-60 cells or STAT4-overexpressing dHL-60 cells (biological replicate=3) error bars represent mean +/- SD; P values were determined using an unpaired two-sided Wilcoxon test. d, Their migration capacity was assessed using the migration assay. HCT116 cells co-cultured with control neutrophils, NFKB1-overexpressing neutrophils, and STAT4-overexpressing neutrophils for 3 days. Images are representative of n = 3 experiments. Bar plots showing the migration cells number of HCT116 cells (biological replicate=3). Error bars represent mean +/- SD; P values were determined using an unpaired two-sided t-test. e, Colony formation assay, in which control neutrophils, NFKB1-overexpressing neutrophils, and STAT4-overexpressing neutrophils were cultured in the upper chamber, and HCT116 cells were cultured in the lower chamber. Images are representative of n = 3 experiments.(Empty vector VS copGFP-NFKB1 p = 0.0097; Empty vector VS copGFP-STAT4 p = 0.0353) p values were calculated using an unpaired two-sides t-test, biological replicate=3.f, The gene expression of NLRP3 and TNFSF10 in neutrophils in scRNA-seq analysis. g, qPCR for assessing the activation status of two transcription factors. Relative expression of NFKB1 and STAT4 in two sorted neutrophils and control neutrophils without sorted(biological replicate=4); Error bars represent mean +/- SD; P values were determined using an unpaired two-sided t-test. h, Top panel showing HCT116 cells (left panel) and SW480 cells (right panel) co-cultured with control neutrophils, TNFSF10 + NLRP3- neutrophils, and NLRP3 + TNFSF10- neutrophils for 3 days. Their migration capacity was assessed using the Transwell assay (middle panel). Images are representative of n = 3 experiments. p values were calculated using an unpaired two-sides t-test, biological replicate=3.Bar plot showing the comparison of number of cell migration and colonies among three groups (bottom panel). Plots represent 10 validations, and error bars represent mean +/- SD. p values were calculated using an unpaired two-sides t-test. Data are presented as mean values +/- SD.

Source data

Extended Data Fig. 6 Mesothelial-to-mesenchymal transition.

a, Spatial distribution(representative images of n = 12 experiments) of PM and normal peritoneum stained for MSLN on pathological slides. b, Representative image of fluorescent staining showing the morphology change in MET-5A cells (with GFP) in control and patient-derived CRC and PM tumor organoids co-cultured with RKO cells (with Red CMTPX). c, Spatial distribution(representative images of n = 12 experiments) of fibroblasts (αSMA), mesothelial cells (MSLN), in normal peritoneal tissue by mIHC. Scale bars, 20 μm. d, Spatial distribution(representative images of n = 12 experiments) of fibroblasts (αSMA), mesothelial cells (MSLN), in PMs by mIHC. Scale bars, 20 μm. Images are representative of n = 12 experiments e, Metabolic reaction activities related to glycolysis and fatty acid oxidation were compared in mesothelial cells using the Compass software between normal peritoneal and PM tissues (left). The metabolic reaction activities related to glycolysis and fatty acid oxidation were compared in MET-5A cells between control and after co-cultured with RKO cells using LC/MS metabolomics. p values were calculated using unpaired two-sides Wilcoxon test. f, Representative images of the tumor load as examined by luminescence at 3 weeks in an in vivo model by intraperitoneal injection. g, Flowchart of the mouse drug administration experiment. h, The Western blot results demonstrate the successful knockout of MSLN in MC38 cell line.(n = 3 biological replicate) i, No differences of the weight of colorectal tumors was found between the anti-MSLN group and control group(biological replicate=3). ns, not significantly, unpaired two-sides t-test. Data are presented as mean values +/- SD. j, The peritoneal weight in the anti-MSLN group was significant decreased compared to control group(biological replicate=3); unpaired two-sides t-test. Data are presented as mean values +/- SD. k, The peritoneal metastasis node size in the anti-MSLN group was significant decreased compared to control group (n = 3 biological replicate). ns, not significantly; unpaired two-sides t-test. Data are presented as mean values +/- SD. l, Representative photos of colonic and peritoneal tissue in the anti-MSLN and control groups. The minimum unit of the ruler is millimeter.

Source data

Supplementary information

Source data

Source Data Fig. 6d,e and Extended Data Fig. 6h

Raw blots for Fig.6d,e and Extended Data Fig. 6h.

Source Data Fig. 1

Statistical Source Data for Fig. 1.

Source Data Fig. 2

Statistical Source Data for Fig. 2.

Source Data Fig. 3

Statistical Source Data for Fig. 3.

Source Data Fig. 4

Statistical Source Data for Fig. 4.

Source Data Fig. 5

Statistical Source Data for Fig. 5.

Source Data Fig. 6

Statistical Source Data for Fig. 6.

Source Data Extended Data Fig. 1

Statistical Source Data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical Source Data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical Source Data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical Source Data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical Source Data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical Source Data for Extended Data Fig. 6.

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

Li, Q., Xiao, Y., Han, L. et al. Microbiome dysbiosis, neutrophil recruitment and mesenchymal transition of mesothelial cells promotes peritoneal metastasis of colorectal cancer. Nat Cancer 6, 493–510 (2025). https://doi.org/10.1038/s43018-025-00910-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-025-00910-9

This article is cited by

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