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:

The ligation between ERMAP, galectin-9 and dectin-2 promotes Kupffer cell phagocytosis and antitumor immunity

Abstract

Kupffer cells, the liver tissue resident macrophages, are critical in the detection and clearance of cancer cells. However, the molecular mechanisms underlying their detection and phagocytosis of cancer cells are still unclear. Using in vivo genome-wide CRISPR-Cas9 knockout screening, we found that the cell-surface transmembrane protein ERMAP expressed on various cancer cells signaled to activate phagocytosis in Kupffer cells and to control of liver metastasis. ERMAP interacted with β-galactoside binding lectin galectin-9 expressed on the surface of Kupffer cells in a manner dependent on glycosylation. Galectin-9 formed a bridging complex with ERMAP and the transmembrane receptor dectin-2, expressed on Kupffer cells, to induce the detection and phagocytosis of cancer cells by Kupffer cells. Patients with low expression of ERMAP on tumors had more liver metastases. Thus, our study identified the ERMAP–galectin–9-dectin-2 axis as an ‘eat me’ signal for Kupffer cells.

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: CRISPR-Cas9 screen identifies ERMAP as a suppressor of tumor liver metastasis.
Fig. 2: ERMAP binds to Kupffer cells and promotes the phagocytosis of cancer cells by Kupffer cells.
Fig. 3: ERMAP ligates Gal-9 in a glycosylation-dependent manner.
Fig. 4: ERMAP–Gal-9 interaction promotes the phagocytosis of cancer cells by Kupffer cells and represses liver metastasis.
Fig. 5: Gal-9 mediates the interaction between ERMAP and dectin-2.
Fig. 6: Dectin-2 is essential for ERMAP-induced Kupffer cells phagocytosis and control of liver metastasis.
Fig. 7: Expression of ERMAP in human tumor tissues correlates with liver metastasis.

Similar content being viewed by others

Data availability

CRISPR screen sequencing datasets have been deposited in the Gene Expression Omnibus database under accession code GSE232126. Source data are provided with this paper.

References

  1. Darvin, P., Toor, S. M., Sasidharan Nair, V. & Elkord, E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp. Mol. Med. 50, 1–11 (2018).

    Article  PubMed  Google Scholar 

  2. Zhou, G., Boor, P. P. C., Bruno, M. J., Sprengers, D. & Kwekkeboom, J. Immune suppressive checkpoint interactions in the tumour microenvironment of primary liver cancers. Br. J. Cancer 126, 10–23 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. Bae, J. et al. IL-2 delivery by engineered mesenchymal stem cells re-invigorates CD8(+) T cells to overcome immunotherapy resistance in cancer. Nat. Cell Biol. 24, 1754–1765 (2022).

    Article  CAS  PubMed  Google Scholar 

  4. & Saadey, A. A. et al. Rebalancing TGFbeta1/BMP signals in exhausted T cells unlocks responsiveness to immune checkpoint blockade therapy.Nat. Immunol. 24, 280–294 (2023).

    Article  CAS  PubMed  Google Scholar 

  5. Li, M. O. et al. Innate immune cells in the tumor microenvironment. Cancer cell 39, 725–729 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Jacquelot, N., Seillet, C., Vivier, E. & Belz, G. T. Innate lymphoid cells and cancer. Nat. Immunol. 23, 371–379 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Pinho, S. et al. VCAM1 confers innate immune tolerance on haematopoietic and leukaemic stem cells. Nat. Cell Biol. 24, 290–298 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Feng, M. et al. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat. Rev. Cancer 19, 568–586 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Gul, N. et al. Macrophages eliminate circulating tumor cells after monoclonal antibody therapy. J. Clin. Invest. 124, 812–823 (2014).

    Article  PubMed Central  PubMed  Google Scholar 

  10. Feng, M. et al. Programmed cell removal by calreticulin in tissue homeostasis and cancer. Nat. Commun. 9, 3194 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  11. Chen, J. et al. SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin. Nature 544, 493–497 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  12. Gordon, S. R. et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 545, 495–499 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Jaiswal, S. et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 138, 271–285 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Barkal, A. A. et al. Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat. Immunol. 19, 76–84 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Barkal, A. A. et al. CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature 572, 392–396 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Crispe, I. N. The liver as a lymphoid organ. Annu. Rev. Immunol. 27, 147–163 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Li, P., He, K., Li, J., Liu, Z. & Gong, J. The role of Kupffer cells in hepatic diseases. Mol. Immunol. 85, 222–229 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Mielgo, A. & Schmid, M. C. Liver tropism in cancer: The hepatic metastatic niche. Cold Spring Harb. Perspect. Med. 10, a037259 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  20. Poon, R. T., Fan, S. T., Lo, C. M., Liu, C. L. & Wong, J. Intrahepatic recurrence after curative resection of hepatocellular carcinoma: long-term results of treatment and prognostic factors. Ann. Surg. 229, 216–222 (1999).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Bayon, L. G. et al. Role of Kupffer cells in arresting circulating tumor cells and controlling metastatic growth in the liver. Hepatology 23, 1224–1231 (1996).

    Article  CAS  PubMed  Google Scholar 

  22. Meterissian, S. H., Toth, C. A., Steele, G. Jr. & Thomas, P. Kupffer cell/tumor cell interactions and hepatic metastasis in colorectal cancer. Cancer Lett. 81, 5–12 (1994).

    Article  CAS  PubMed  Google Scholar 

  23. Matsumura, H. et al. Kupffer cells decrease metastasis of colon cancer cells to the liver in the early stage. Int. J. Oncol. 45, 2303–2310 (2014).

    Article  PubMed  Google Scholar 

  24. Joung, J. et al. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening. Nat. Protoc. 12, 828–863 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Yang, D. et al. CRISPR screening uncovers a central requirement for HHEX in pancreatic lineage commitment and plasticity restriction. Nat. Cell Biol. 24, 1064–1076 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Belk, J. A. et al. Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Cancer cell 40, 768–786 e767 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Hallin, J. et al. Anti-tumor efficacy of a potent and selective non-covalent KRASG12D inhibitor. Nat. Med. 28, 2171–2182 (2022).

    Article  CAS  PubMed  Google Scholar 

  28. Thiemann, S. & Baum, L. G. Galectins and immune responses: Just how do they do those things they do? Annu. Rev. Immunol. 34, 243–264 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Daley, D. et al. Dectin 1 activation on macrophages by galectin 9 promotes pancreatic carcinoma and peritumoral immune tolerance. Nat. Med. 23, 556–567 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Kimura, Y. et al. The innate immune receptor Dectin-2 mediates the phagocytosis of cancer cells by Kupffer cells for the suppression of liver metastasis. PNAS 113, 14097–14102 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. Lu, M., Zessin, A. S., Glover, W. & Hsu, D. S. Activation of the mTOR Pathway by Oxaliplatin in the Treatment of Colorectal Cancer Liver Metastasis. PLoS One 12, e0169439 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  32. McMullin, R. P. et al. A BRCA1 deficient-like signature is enriched in breast cancer brain metastases and predicts DNA damage-induced poly (ADP-ribose) polymerase inhibitor sensitivity. Breast cancer Res.: BCR 16, R25 (2014).

    Article  PubMed Central  PubMed  Google Scholar 

  33. Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Gordon, S. Phagocytosis: An immunobiologic process. Immunity 44, 463–475 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Christofides, A. et al. The complex role of tumor-infiltrating macrophages. Nat. Immunol. 23, 1148–1156 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Lu, Y. et al. Immunological conversion of solid tumours using a bispecific nanobioconjugate for cancer immunotherapy. Nat. Nanotechnol. 17, 1332–1341 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  37. Robinson, M. J. et al. Dectin-2 is a Syk-coupled pattern recognition receptor crucial for Th17 responses to fungal infection. J. Exp. Med. 206, 2037–2051 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  38. Duan, J. L. et al. E3 ligase c-Cbl regulates intestinal inflammation through suppressing fungi-induced noncanonical NF-kappaB activation. Sci. Adv. 7, eabe5171 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Sato, K. et al. Dectin-2 is a pattern recognition receptor for fungi that couples with the Fc receptor gamma chain to induce innate immune responses. J. Biol. Chem. 281, 38854–38866 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Johswich, A. et al. N-glycan remodeling on glucagon receptor is an effector of nutrient sensing by the hexosamine biosynthesis pathway. J. Biol. Chem. 289, 15927–15941 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Pinho, S. S. & Reis, C. A. Glycosylation in cancer: mechanisms and clinical implications. Nat. Rev. Cancer 15, 540–555 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Su, M., Lin, Y., Cui, C., Tian, X. & Lai, L. ERMAP is a B7 family-related molecule that negatively regulates T cell and macrophage responses. Cell. Mol. Immunol. 18, 1920–1933 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Matsushita, N. et al. Requirement of divalent galactoside-binding activity of ecalectin/galectin-9 for eosinophil chemoattraction. J. Biol. Chem. 275, 8355–8360 (2000).

    Article  CAS  PubMed  Google Scholar 

  44. Ray, A. & Dittel, B. N. Isolation of mouse peritoneal cavity cells.J. Vis. Exp. (35), 1488 (2010).

    Google Scholar 

  45. Charni-Natan, M. & Goldstein, I. Protocol for primary mouse hepatocyte isolation. STAR Protoc. 1, 100086 (2020).

    Article  PubMed Central  PubMed  Google Scholar 

  46. Yuan, J. H. et al. A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer cell 25, 666–681 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Li, J. et al. SLC38A4 functions as a tumour suppressor in hepatocellular carcinoma through modulating Wnt/beta-catenin/MYC/HMGCS2 axis. Br. J. Cancer 125, 865–876 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  48. Yuan, J. H. et al. The MBNL3 splicing factor promotes hepatocellular carcinoma by increasing PXN expression through the alternative splicing of lncRNA-PXN-AS1. Nat. Cell Biol. 19, 820–832 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Dai, Y. Z. et al. METTL16 promotes hepatocellular carcinoma progression through downregulating RAB11B-AS1 in an m(6)A-dependent manner. Cell. Mol. Biol. Lett. 27, 41 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  50. Theruvath, J. et al. Anti-GD2 synergizes with CD47 blockade to mediate tumor eradication. Nat. Med. 28, 333–344 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We thank X. Liu from the Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine and J. Ding from Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University for technical assistance. This work was supported by grants from the National Natural Science Foundation of China (Grant No. 81830085 to S.S.; 92059111 and 81972738 to J.Y.), the Science and Technology Innovation Action Plan of Shanghai (22140902100 to J.Y.; 23ZR1477300 to J.L.), the Shanghai Key Laboratory of Cell Engineering (14DZ2272300 to J.Y.), and the Shanghai Municipal Health Commission Clinical Research Program (20214Y0014 to J.L.).

Author information

Authors and Affiliations

Authors

Contributions

J.Y., S.S. J.L. and G.C. designed the experiments. J.L., J.Y., X.L., R.G., Y.Y., Y.L., W.L., M.H. and X.H. performed the experiments. J.Y., J.L., X.L., R.G., Y.Y., J.W. and G.C. analyzed the data. J.Y. and J.L. wrote the manuscript. J.Y. and S.S. supervised the project. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Guoxiang Cai, Shu-han Sun or Ji-hang Yuan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks André Veillette, Bing Su and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the Nature Immunology team.

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 ERMAP is a suppressor of liver metastasis.

a, Representative liver images of nude mice at four weeks post-intrasplenic injection of Hepa1-6 cells transduced with the GeCKO library. b, Flow cytometry showing the expression of ERMAP in wild-type (WT) and ERMAPKO Hepa1-6 cells. c, Flow cytometry showing the expression of ERMAP in ERMAPEV and ERMAPOE Hepa1-6 cells. d, Representative HE-stained images of liver tissues isolated from C57BL/6 mice at day 19 post-intrasplenic injection of 5 × 106 wild-type or ERMAPKO Hepa1-6 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 2 mm. e, Representative HE-stained images of liver tissues isolated from C57BL/6 mice at day 49 post-intrasplenic injection of 5 × 106 ERMAPEV or ERMAPOE Hepa1-6 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. f, Flow cytometry showing the expression of ERMAP in ERMAPshCtrl and ERMAPKD SNU-398 cells. g, Flow cytometry showing the expression of ERMAP in ERMAPEV and ERMAPOE SNU-398 cells. h, Flow cytometry showing the expression of ERMAP in ERMAPshCtrl and ERMAPKD B16F10 cells. i, Representative HE-stained images of liver tissues isolated from nude mice at day 10 post-intrasplenic injection of 2×106 ERMAPshCtrl or ERMAPKD B16F10 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. j, Flow cytometry showing the expression of ERMAP in ERMAPEV and ERMAPOE B16F10 cells. k, Representative HE-stained images of liver tissues isolated from nude mice at day 14 post-intrasplenic injection of 2×106 ERMAPEV or ERMAPOE B16F10 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. l, Representative HE-stained images of liver tissues isolated from C57BL/6 mice at day 14 post-intrasplenic injection of 2 × 106 ERMAPshCtrl or ERMAPKD B16F10 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. m, Representative HE-stained images of liver tissues isolated from C57BL/6 mice at day 14 post-intrasplenic injection of 2 × 106 ERMAPEV or ERMAPOE B16F10 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. Results are shown as mean ± s.e.m. or representative images of n = 3 independent experiments (b, c, f-h, j) or n = 6 (d, e, i, k-m) mice in each group and were analyzed by Kruskal–Wallis test followed by Dunn’s multiple comparisons test (d, i, l) or two-sided Mann–Whitney test (e, k, m).

Source data

Extended Data Fig. 2 ERMAP had no effect on lung metastasis, but repressed subcutaneous tumor growth.

a, Representative HE-stained images of lung tissues isolated from nude mice at day 28 post-tail vein injection of 5 × 106 wild-type (WT) or ERMAPKO Hepa1-6 cells and numbers of lung metastases per mouse. Scale bars, 1 mm. b, Representative HE-stained images of lung tissues isolated from nude mice at day 28 post-tail vein injection of 5 × 106 ERMAPOE Hepa1-6 cells and numbers of lung metastases per mouse. Scale bars, 1 mm. c, Representative HE-stained images of lung tissues isolated from nude mice at day 14 post-tail vein injection of 2 × 106 ERMAPKD B16F10 cells and percentages of tumor-replaced areas in lung tissues. Scale bars, 1 mm. d, Representative HE-stained images of lung tissues isolated from nude mice at day 14 post-tail vein injection of 2 × 106 ERMAPOE B16F10 cells and percentages of tumor-replaced areas in lung tissues. Scale bars, 1 mm. e, Subcutaneous tumor volume at days 5, 8, 11, 14, and tumor weight and tumor image at day 14 post-subcutaneous injection of 5 × 106 wild-type or ERMAPKO1 Hepa1-6 cells into nude mice. f, Subcutaneous tumor volume at days 5, 8, 11, 14, and tumor weight and tumor image at day 14 post-subcutaneous injection of 5 × 106 wild-type or ERMAPKO2 Hepa1-6 cells into nude mice. g, Subcutaneous tumor volume at days 5, 8, 11, 14, 21, and tumor weight and tumor image at day 21 post-subcutaneous injection of 5 × 106 ERMAPOE Hepa1-6 cells into nude mice. h, Subcutaneous tumor volume at days 3, 7, 14, 21, tumor weight and tumor image at day 21 post-subcutaneous injection of 5 × 106 ERMAPOE SNU-398 cells into nude mice. i, Subcutaneous tumor weight and tumor image at day 13 post-subcutaneous injection of 5 × 106 ERMAPOE B16F10 cells into nude mice. Results are shown as mean ± s.e.m. of n = 6 (a-d, h) mice, n = 8 (e, g, i) mice, or n = 7 (f) mice in each group and were analyzed by Kruskal–Wallis test followed by Dunn’s multiple comparisons test (a, c) or two-sided Mann–Whitney test (b, d-i).

Source data

Extended Data Fig. 3 ERMAP promotes in vitro phagocytosis of cancer cells by Kupffer cells.

a,b, Live-cell microscopy showing in vitro phagocytosis of pHrodo Red+ GFP+ ERMAPKO (a) or ERMAPOE (b) Hepa1-6 cells by Kupffer cells at 0, 6, 12, 24 hours post co-culture. Scale bars, 100 µm. c,d, Flow cytometry (c) or fluorescence microscopy (d) showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPKD SNU-398 cells after co-culture for 2 h. e,f, Flow cytometry (e) or fluorescence microscopy (f) showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPKD B16F10 cells after co-culture for 2 h. g,h, Flow cytometry (g) or fluorescence microscopy (h) showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE SNU-398 cells after co-culture for 2 h. i,j, Flow cytometry (i) or fluorescence microscopy (j) showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE B16F10 cells after co-culture for 2 h. Results are shown as mean ± s.e.m. of n = 3 independent experiments and were analyzed by one-way ANOVA followed by Dunnett’s multiple comparisons test (a, c-f) or two-sided Student’s t-test (b, g-j).

Source data

Extended Data Fig. 4 ERMAP-induced phagocytosis is not dependent on CD47, PD-L1 and CD24, and is tumor cell specific.

a-c, Flow cytometry showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPKO Hepa1-6 cells (a), ERMAPOE Hepa1-6 cells (b), or ERMAPOE B16f10 cells (c) after co-culture for 2 h in the presence of CD47 or PD-L1 blocking Ab or IgG control. d, Flow cytometry showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE SNU-398 cells after co-culture for 2 h in the presence of CD24 blocking Ab or IgG control. e, Flow cytometry showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE hepatocytes, T cells, B cells, NK cells, MC38, and CT26 cells after co-culture for 2 h. Results are shown as mean ± s.e.m. of n = 3 independent experiments and were analyzed by one-way ANOVA followed by Dunnett’s multiple comparisons test (a) or two-sided Student’s t-test (b-e).

Source data

Extended Data Fig. 5 ERMAP promotes in vivo phagocytosis of cancer cells by Kupffer cells.

a, Flow cytometry representative plots and percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPKD SNU-398 cells 12 h post-intrasplenic inoculation. b, Confocal microscopy showing percentages of CFSE-labeled ERMAPKD SNU-398 cells phagocytosed by F4/80+ Kupffer cells 12 h post-intrasplenic inoculation. c, Flow cytometry representative plots and percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE SNU-398 cells 12 h post-intrasplenic inoculation. d, Confocal microscopy showing percentages of CFSE-labeled ERMAPOE SNU-398 cells phagocytosed by F4/80+ Kupffer cells 12 h post-intrasplenic inoculation. e, Flow cytometry representative plots and percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPKD B16F10 cells 12 h post-intrasplenic inoculation. f, Confocal microscopy showing percentages of CFSE-labeled ERMAPKD B16F10 cells phagocytosed by F4/80+ Kupffer cells 12 h post-intrasplenic inoculation. g, Flow cytometry representative plots and percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE B16F10 cells 12 h post-intrasplenic inoculation. h, Confocal microscopy showing percentages of CFSE-labeled ERMAPOE B16F10 cells phagocytosed by F4/80+ Kupffer cells 12 h post-intrasplenic inoculation. Results are shown as mean ± s.e.m. of n = 5 mice in each group and were analyzed by Kruskal–Wallis test followed by Dunn’s multiple comparisons test (a, b, e, f) or two-sided Mann–Whitney test (c, d, g, h).

Source data

Extended Data Fig. 6 Kupffer cells phagocytosis of cancer cells controls liver metastasis.

a,b, Luciferase signal intensities at days 0, 1, 3 after intrasplenic injection of ERMAPOE Hepa1-6 (a) or SNU-398 (b) cells in nude mice that received intraperitoneal injection with clodronate liposomes on day -1. c,d, Flow cytometry showing the apoptosis of non-phagocytosed Hepa1-6 cells using Annexin V (c) or cleaved caspase-3 (d) staining after co-culture of CFSE-labeled wild-type (WT) or ERMAPKO Hepa1-6 cells with Kupffer cells for 12 h. e,f, Flow cytometry showing the apoptosis of non-phagocytosed Hepa1-6 cells using Annexin V (e) or cleaved caspase-3 (f) staining after co-culture of CFSE-labeled ERMAPOE Hepa1-6 cells with Kupffer cells for 12 h. g-i, Confocal microscopy showing percentages of cleaved caspase-3 (g), Ki67 (h), or PCNA (i) positive non-phagocytosed CFSE-labeled ERMAPKO or ERMAPOE Hepa1-6 cells 12 h post-intrasplenic inoculation. Scale bars, 5 µm. j,k Representative HE-stained images of liver tissues isolated from C57BL/6 mice that received intraperitoneal injection with clodronate liposomes on day -1 before the intrasplenic injection with ERMAPOE Hepa1-6 (j) or B16F10 (k) cells and analyzed at day 28 (j) or 10 (k) post-intrasplenic injection and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. Results are shown as mean ± s.e.m. of n = 3 independent experiments (c-f), n = 6 (a, b, j, k) or n = 5 (g-i) mice in each group and were analyzed by two-sided Mann–Whitney test (a, b, comparisons between ERMAPEV and ERMAPOE of g-i, j, k), one-way ANOVA followed by Dunnett’s multiple comparisons test (c, d), two-sided Student’s t-test (e, f), or Kruskal–Wallis test followed by Dunn’s multiple comparisons test (comparisons between ERMAPKO1, ERMAPKO2 and WT of g-i).

Source data

Extended Data Fig. 7 ERMAP–Gal-9 interaction promotes the in vitro phagocytosis of cancer cells by Kupffer cells.

a, Flow cytometry showing the expression of ERMAP in ERMAPOE or ERMAPN135AOE Hepa1-6 cells. b, Flow cytometry showing the expression of ERMAP in ERMAPOE or ERMAPN132AOE SNU-398 cells. c,d, Flow cytometry (c) or fluorescence microscopy (d) showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE or ERMAPN132AOE SNU-398 cells after co-culture for 2 h in the presence or absence of Gal-9 neutralizing antibody. e,f, Flow cytometry (e) or fluorescence microscopy (f) showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPKD SNU-398 cells after co-culture for 2 h in the presence or absence of Gal-9 neutralizing antibody. g, Flow cytometry showing the expression of ERMAP in ERMAPOE or ERMAPN135AOE B16F10 cells. h,i, Flow cytometry (h) or fluorescence microscopy (i) showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE or ERMAPN135AOE B16F10 cells after co-culture for 2 h in the presence or absence of Gal-9 neutralizing antibody. Results are shown as mean ± s.e.m. or representative images of n = 3 independent experiments in each group and were analyzed by two-way ANOVA followed by Tukey’s multiple comparisons test.

Source data

Extended Data Fig. 8 ERMAP–Gal-9 interaction promotes the in vivo phagocytosis of cancer cells by Kupffer cells and represses liver metastasis.

a, Flow cytometry representative plots and percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE or ERMAPN132AOE SNU-398 cells 12 h post-intrasplenic inoculation. b, Confocal microscopy showing percentages of CFSE-labeled ERMAPOE or ERMAPN132AOE SNU-398 cells phagocytosed by F4/80+ Kupffer cells 12 h post-intrasplenic inoculation. c, Flow cytometry representative plots and percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE or ERMAPN135AOE B16F10 cells 12 h post-intrasplenic inoculation. d, Confocal microscopy showing percentages of CFSE-labeled ERMAPOE or ERMAPN135AOE B16F10 cells phagocytosed by F4/80+ Kupffer cells 12 h post-intrasplenic inoculation. e, Representative HE-stained images of liver tissues isolated from C57BL/6 mice at day 49 post-intrasplenic injection with ERMAPOE or ERMAPN135AOE Hepa1-6 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. f, Representative HE-stained images of liver tissues isolated from nude mice at day 14 post-intrasplenic injection with ERMAPOE or ERMAPN135AOE B16F10 cells and number of hepatic metastases per mouse. Scale bars, 1 mm. Results are shown as mean ± s.e.m. or representative images of n = 5 (a-d) or n = 6 (e, f) mice in each group and were analyzed by Kruskal–Wallis test followed by Dunn’s multiple comparisons test.

Source data

Extended Data Fig. 9 ERMAP–Gal-9–dectin-2 ligation promotes the phagocytosis of cancer cells by Kupffer cells and represses liver metastasis.

a,b, Flow cytometry (a) or fluorescence microscopy (b) showing percentages of wild-type (Clec6a+/+) or Clec6a-/- Kupffer cells phagocytizing CFSE-labeled ERMAPOE SNU-398 cells after co-culture for 2 h. c,d, Flow cytometry (c) or fluorescence microscopy (d) showing percentages of wild-type or Clec6a-/- Kupffer cells phagocytizing CFSE-labeled ERMAPKD SNU-398 cells after co-culture for 2 h. e,f, Flow cytometry (e) or fluorescence microscopy (f) showing percentages of wild-type or Clec6a-/- Kupffer cells phagocytizing CFSE-labeled ERMAPOE B16F10 cells after co-culture for 2 h. g, Flow cytometry representative plots and percentages of wild-type or Clec6a-/- F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE B16F10 cells 12 h post-intrasplenic inoculation. h, Confocal microscopy showing percentages of CFSE-labeled ERMAPOE B16F10 cells phagocytosed by wild-type or Clec6a-/- F4/80+ Kupffer cells12h post-intrasplenic inoculation. i, Representative HE-stained images of liver tissues isolated from wild-type or Clec6a-/- mice at day 14 post-intrasplenic injection with ERMAPOE B16F10 cells and percentages of tumor-replaced areas in liver tissues. Scale bars, 1 mm. j, Flow cytometry showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPOE B16F10 cells after co-culture for 2 h in the presence of the indicated inhibitors, control [dimethyl sulfoxide (DMSO)], Syk kinase inhibitor (piceatannol), mTOR inhibitor (Torin1), Src family kinase inhibitor (PP2), PI3K inhibitor (LY294002), NF-κB inhibitor (JSH-23), P38 inhibitor (SB203580), BTK inhibitor (LFM-A13), or MEK inhibitor (PD98059). k, Flow cytometry showing percentages of F4/80+ Kupffer cells phagocytizing CFSE-labeled ERMAPEV or ERMAPOE B16F10 cells in the presence of Syk kinase inhibitor (piceatannol). Results are shown as mean ± s.e.m. of n = 3 independent experiments (a-f, j, k), n = 5 (g, h) or n = 6 (i) mice in each group and were analyzed by two-way ANOVA followed by Tukey’s multiple comparisons test (a-f), two-sided Mann–Whitney test (g-i) or two-sided Student’s t-test (j, k).

Source data

Extended Data Fig. 10 ERMAP–Gal-9–dectin-2 ligation doesn’t mediate the phagocytosis of cancer cells by BMDMs.

a, b, Flow cytometry (a) or immunofluorescence using FITC-conjugated antibody against human Fc (b) showing the binding of ERMAP-Fc to wild-type (Clec6a+/+) or Clec6a-/- BMDMs. Scale bars, 100 µm. c,d, Flow cytometry (c) or fluorescence microscopy (d) showing percentages of wild-type or Clec6a-/- BMDMs phagocytizing CFSE-labeled ERMAPOE Hepa1-6 cells after co-culture for 2 h. e,f, Flow cytometry (e) or fluorescence microscopy (f) showing percentages of wild-type or Clec6a-/- BMDMs phagocytizing CFSE-labeled ERMAPOE SNU-398 cells after co-culture for 2 h. Results are shown as mean ± s.e.m. or representative images of n = 3 independent experiments and were analyzed by two-way ANOVA followed by Tukey’s multiple comparisons test.

Source data

Supplementary information

Supplementary Information

Supplementary Figures 1-6, Supplementary Tables 3 and 4, and Supplementary Video legends.

Reporting Summary

Supplementary Video

Kupffer cells preferentially phagocytizes wild-type Hepa1-6 cells compared to ERMAPKO Hepa1-6 cells. Kupffer cells were labeled with Dil dye (red), wild-type Hepa1-6 cells were labeled with CFSE (green), and ERMAPKO Hepa1-6 cells were labeled with Violet (blue). The phagocytoses of Hepa1-6 cells by Kupffer cells were detected by time-lapse imaging using the Nikon CSU-W1 spinning disc microscope. Scale bars, 10 µm.

Supplementary Table

Table S1_sgRNAs enriched, Table S2_proteins_MS, Supplem. Figure 1_Source, Supplem. Figure 2_Source, Supplem. Figure 3_Source, Supplem. Figure 4_Source, Supplem. Figure 5_Source.

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 Fig. 7

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

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.

Source Data Extended Data Fig. 10

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

Li, J., Liu, Xg., Ge, Rl. et al. The ligation between ERMAP, galectin-9 and dectin-2 promotes Kupffer cell phagocytosis and antitumor immunity. Nat Immunol 24, 1813–1824 (2023). https://doi.org/10.1038/s41590-023-01634-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01634-7

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