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:

Sirpα on tumor-associated myeloid cells restrains antitumor immunity in colorectal cancer independent of its interaction with CD47

Abstract

Immunosuppressive myeloid cells hinder immunotherapeutic efficacy in tumors, but the precise mechanisms remain undefined. Here, by performing single-cell RNA sequencing in colorectal cancer tissues, we found tumor-associated macrophages and granulocytic myeloid-derived suppressor cells increased most compared to their counterparts in normal tissue and displayed the highest immune-inhibitory signatures among all immunocytes. These cells exhibited significantly increased expression of immunoreceptor tyrosine-based inhibitory motif-bearing receptors, including SIRPA. Notably, Sirpa−/− mice were more resistant to tumor progression than wild-type mice. Moreover, Sirpα deficiency reprogramed the tumor microenvironment through expansion of TAM_Ccl8hi and gMDSC_H2-Q10hi subsets showing strong antitumor activity. Sirpa−/− macrophages presented strong phagocytosis and antigen presentation to enhance T cell activation and proliferation. Furthermore, Sirpa−/− macrophages facilitated T cell recruitment via Syk/Btk-dependent Ccl8 secretion. Therefore, Sirpα deficiency enhances innate and adaptive immune activation independent of expression of CD47 and Sirpα blockade could be a promising strategy to improve cancer immunotherapy efficacy.

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: Tumor-enriched myeloid cells exhibit the highest immune-inhibitory signatures.
Fig. 2: Sirpα deletion inhibits solid tumor development.
Fig. 3: Sirpα deficiency prohibits spontaneous colon cancer development.
Fig. 4: Sirpα deficiency reprograms tumor immune microenvironment to facilitate antitumor immunity.
Fig. 5: Sirpα deficiency enhances macrophage function and T cell activation.
Fig. 6: Sirpα deficient macrophages enhance T cell recruitment via Syk/Btk-dependent Ccl8 secretion.

Similar content being viewed by others

Data availability

The RNA-seq and scRNA-seq data that support the findings of this study have been deposited in the Genome Sequence Archive75 in the National Genomics Data Center76, China National Center for Bioinformation/Beijing Institute of Genomics, Chinese Academy of Sciences (CRA005626 and HRA001948, with processed data available in OMIX921) that are accessible at https://ngdc.cncb.ac.cn. Access of sequence data of human samples is restricted due to patient privacy and China’s human genetic resources regulations. HRA001948 can be accessed by clicking the ‘Request Data’ button and filling in the required information for approval. The public scRNA-seq data are available in GEO (GSE188711 and GSE66343) and Synapse (syn26844071), and the public RNA-seq data are available in GEO (GSE17536 and GSE17537) and TCGA (TCGA-LIHC and TCGA-LUSC). Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding authors on reasonable request.

Code availability

The custom script and exact parameters used for data processing and analysis are available in GitHub at https://github.com/snow55/SIRPAproject.

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. Ostuni, R., Kratochvill, F., Murray, P. J. & Natoli, G. Macrophages and cancer: from mechanisms to therapeutic implications. Trends Immunol. 36, 229–239 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).

    Article  PubMed  Google Scholar 

  5. Sanmamed, M. F. & Chen, L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell 175, 313–326 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sharpe, A. H. Introduction to checkpoint inhibitors and cancer immunotherapy. Immunol. Rev. 276, 5–8 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Drake, C. G., Lipson, E. J. & Brahmer, J. R. Breathing new life into immunotherapy: review of melanoma, lung and kidney cancer. Nat. Rev. Clin. Oncol. 11, 24–37 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Vesely, M. D., Zhang, T. & Chen, L. Resistance mechanisms to anti-PD cancer immunotherapy. Annu. Rev. Immunol. 40, 45–74 (2022).

    Article  CAS  PubMed  Google Scholar 

  9. Kubli, S. P., Berger, T., Araujo, D. V., Siu, L. L. & Mak, T. W. Beyond immune checkpoint blockade: emerging immunological strategies. Nat. Rev. Drug Discov. 20, 899–919 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. Keren, L. et al. A structured tumor-immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging. Cell 174, 1373–1387 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kim, I. S. et al. Immuno-subtyping of breast cancer reveals distinct myeloid cell profiles and immunotherapy resistance mechanisms. Nat. Cell Biol. 21, 1113–1126 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gubin, M. M. et al. High-dimensional analysis delineates myeloid and lymphoid compartment remodeling during successful immune-checkpoint cancer therapy. Cell 175, 1014–1030 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pittet, M. J., Michielin, O. & Migliorini, D. Clinical relevance of tumour-associated macrophages. Nat. Rev. Clin. Oncol. 19, 402–421 (2022).

    Article  PubMed  Google Scholar 

  14. Veillette, A. & Chen, J. SIRPα-CD47 immune checkpoint blockade in anticancer therapy. Trends Immunol. 39, 173–184 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Chao, M. P., Weissman, I. L. & Majeti, R. The CD47-SIRPα pathway in cancer immune evasion and potential therapeutic implications. Curr. Opin. Immunol. 24, 225–232 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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  PubMed Central  Google Scholar 

  18. Majeti, R. et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 138, 286–299 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chao, M. P. et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 142, 699–713 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hutter, G. et al. Microglia are effector cells of CD47-SIRPα antiphagocytic axis disruption against glioblastoma. Proc. Natl Acad. Sci. USA 116, 997–1006 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ingram, J. R. et al. Localized CD47 blockade enhances immunotherapy for murine melanoma. Proc. Natl Acad. Sci. USA 114, 10184–10189 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Advani, R. et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N. Engl. J. Med. 379, 1711–1721 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Veglia, F., Perego, M. & Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 19, 108–119 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ravetch, J. V. & Lanier, L. L. Immune inhibitory receptors. Science 290, 84–89 (2000).

    Article  ADS  CAS  PubMed  Google Scholar 

  26. Rumpret, M. et al. Functional categories of immune inhibitory receptors. Nat. Rev. Immunol. 20, 771–780 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Guo, W. et al. Resolving the difference between left-sided and right-sided colorectal cancer by single-cell sequencing. JCI Insight 7, e152616 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Joanito, I. et al. Single-cell and bulk transcriptome sequencing identifies two epithelial tumor cell states and refines the consensus molecular classification of colorectal cancer. Nat. Genet. 54, 963–975 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Deng, M. et al. LILRB4 signalling in leukaemia cells mediates T cell suppression and tumour infiltration. Nature 562, 605–609 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. 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 

  32. Snider, A. J. et al. Murine model for colitis-associated cancer of the colon. Methods Mol. Biol. 1438, 245–254 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ahn, E. et al. Role of PD-1 during effector CD8 T cell differentiation. Proc. Natl Acad. Sci. USA 115, 4749–4754 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mensali, N. et al. Antigen-delivery through invariant chain (CD74) boosts CD8 and CD4 T cell immunity. Oncoimmunology 8, 1558663 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Okazawa, H. et al. Negative regulation of phagocytosis in macrophages by the CD47-SHPS-1 system. J. Immunol. 174, 2004–2011 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Dong, H. et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat. Med. 8, 793–800 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Waldman, A. D., Fritz, J. M. & Lenardo, M. J. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 20, 651–668 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Khalil, D. N., Smith, E. L., Brentjens, R. J. & Wolchok, J. D. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat. Rev. Clin. Oncol. 13, 273–290 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ren, X. et al. Insights gained from single-cell analysis of immune cells in the tumor microenvironment. Annu. Rev. Immunol. 39, 583–609 (2021).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  44. Molgora, M. et al. TREM2 modulation remodels the tumor myeloid landscape enhancing anti-PD-1 immunotherapy. Cell 182, 886–900 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhao, X. W. et al. CD47-signal regulatory protein-α (SIRPα) interactions form a barrier for antibody-mediated tumor cell destruction. Proc. Natl Acad. Sci. USA 108, 18342–18347 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Alvey, C. M. et al. SIRPA-inhibited, marrow-derived macrophages engorge, accumulate, and differentiate in antibody-targeted regression of solid tumors. Curr. Biol. 27, 2065–2077 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ring, N. G. et al. Anti-SIRPα antibody immunotherapy enhances neutrophil and macrophage antitumor activity. Proc. Natl Acad. Sci. USA 114, E10578–E10585 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gauttier, V. et al. Selective SIRPα blockade reverses tumor T cell exclusion and overcomes cancer immunotherapy resistance. J. Clin. Invest. 130, 6109–6123 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. van Helden, M. J. et al. BYON4228 is a pan-allelic antagonistic SIRPα antibody that potentiates destruction of antibody-opsonized tumor cells and lacks binding to SIRPγ on T cells. J. Immunother. Cancer 11, e006567 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Pan, L. et al. Lack of SIRP-α reduces lung cancer growth in mice by promoting anti-tumour ability of macrophages and neutrophils. Cell Prolif. 56, e13361 (2023).

    Article  CAS  PubMed  Google Scholar 

  51. Bian, Z. et al. Intratumoral SIRPα-deficient macrophages activate tumor antigen-specific cytotoxic T cells under radiotherapy. Nat. Commun. 12, 3229 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  52. Willingham, S. B. et al. The CD47-signal regulatory protein α (SIRPα) interaction is a therapeutic target for human solid tumors. Proc. Natl Acad. Sci. USA 109, 6662–6667 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liu, X. et al. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat. Med. 21, 1209–1215 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Tseng, D. et al. Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc. Natl Acad. Sci. USA 110, 11103–11108 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Liu, M. et al. Metabolic rewiring of macrophages by CpG potentiates clearance of cancer cells and overcomes tumor-expressed CD47-mediated ‘don’t-eat-me’ signal. Nat. Immunol. 20, 265–275 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zhong, C. et al. Poly(I:C) enhances the efficacy of phagocytosis checkpoint blockade immunotherapy by inducing IL-6 production. J. Leukoc. Biol. 110, 1197–1208 (2021).

    Article  CAS  PubMed  Google Scholar 

  58. Sim, J. et al. Discovery of high affinity, pan-allelic, and pan-mammalian reactive antibodies against the myeloid checkpoint receptor SIRPα. MAbs 11, 1036–1052 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Jhunjhunwala, S., Hammer, C. & Delamarre, L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 21, 298–312 (2021).

    Article  CAS  PubMed  Google Scholar 

  60. Kroemer, G., Galassi, C., Zitvogel, L. & Galluzzi, L. Immunogenic cell stress and death. Nat. Immunol. 23, 487–500 (2022).

    Article  CAS  PubMed  Google Scholar 

  61. Grivennikov, S. et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 15, 103–113 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Qin, P. et al. Integrated decoding hematopoiesis and leukemogenesis using single-cell sequencing and its medical implication. Cell Discov. 7, 2 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Wang, X. et al. Reinvestigation of classic T cell subsets and identification of novel cell subpopulations by single-cell RNA sequencing. J. Immunol. 208, 396–406 (2022).

    Article  CAS  PubMed  Google Scholar 

  64. Li, H. et al. Exploring the dynamics and influencing factors of CD4 T cell activation using single-cell RNA-seq. iScience 26, 107588 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  65. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Linderman, G. C., Rachh, M., Hoskins, J. G., Steinerberger, S. & Kluger, Y. Fast interpolation-based t-SNE for improved visualization of single-cell RNA-seq data. Nat. Methods 16, 243–245 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kobak, D. & Berens, P. The art of using t-SNE for single-cell transcriptomics. Nat. Commun. 10, 5416 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  69. Cao, Y. et al. scDC: single cell differential composition analysis. BMC Bioinf. 20, 721 (2019).

    Article  CAS  Google Scholar 

  70. Yaari, G., Bolen, C. R., Thakar, J. & Kleinstein, S. H. Quantitative set analysis for gene expression: a method to quantify gene set differential expression including gene–gene correlations. Nucleic Acids Res. 41, e170 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  72. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  73. Jin, W. et al. Genome-wide detection of DNase I hypersensitive sites in single cells and FFPE tissue samples. Nature 528, 142–146 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhou, J. et al. Anti-PD-1 therapy achieves favorable outcomes in HBV-positive non-liver cancer. Oncogenesis 12, 22 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chen, T. et al. The genome sequence archive family: toward explosive data growth and diverse data types. Genomics Proteomics Bioinformatics 19, 578–583 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  76. CNCB-NGDC Members and Partners. Database resources of the National Genomics Data Center, China National Center for Bioinformation in 2022. Nucleic Acids Res. 50, D27–D38 (2022).

Download references

Acknowledgements

This study was supported by grants from National Key R&D Program of China (2020YFA0509400 to J.C.; 2021YFF1200900 to W.J. and N.H.; 2019YFA0110300 to J.C.; and 2021YFA0909300 to W.J. and N.H.), National Natural Science Foundation of China (82150117 and 82071745 to J.C.; 32170646 and 32370688 to W.J.; 82101329 to W.Y.; and 32300760 to C.H.), the Guangdong project (2019QN01Y212 to J.C. and 2023B1111020006 to W.J.), Guangdong Basic and Applied Basic Research Foundation (2023A1515011908 to N.H.) and Shenzhen Science and Technology Program (JCYJ20220818100401003 and KQTD20180411143432337 to W.J.). We thank the Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine for support in all animal experiments. We thank members of the Chen laboratory and Jin laboratory for assistance and discussion.

Author information

Authors and Affiliations

Authors

Contributions

W.J. and J.C. conceived the project. Y.L. and Q.Z. collected the clinical samples. S.C. and P.Y. conducted the scRNA-seq. Xuefei Wang analyzed the data. C.H. and Y.W. performed the experiments, with contributions from Y.F., Xiumei Wang, J.L., C.M., L.W., Xinyu Wang, W.Y., W.C. and S.C. N.H., W.H., J.C. and W.J. supervised the study. J.C. and W.J. wrote the manuscript, with input from other authors. All authors read and approved the manuscript.

Corresponding authors

Correspondence to Ni Hong, Weiling He, Jun Chen or Wenfei Jin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks the anonymous reviewers 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 Myeloid cells demonstrate pro-tumor signatures in tumor tissues of colorectal cancer patient.

a. t-SNE visualization of cell type specific genes colored by expression level. b. Cell proportion of each cell type in CRCN, CRCP and CRCT tissues calculated by scDC. The p value were calculated by bootstrap resampling through GLM model in scDC. c. Heat map of top 3 differentially expressed genes in each myeloid cell cluster. Color represents scaled gene average expression in each cluster. d, e. Volcano plot of differentially expressed genes in macrophage (d) and neutrophil (e) between tumor tissue and normal/paracancerous tissue (Wilcoxon test, two-sided). Red points and blue points represent tumor tissue highly expressed genes and normal/paracancerous tissue highly expressed genes, respectively. Bonferroni was used for correction of multiple comparisons.

Source data

Extended Data Fig. 2 Tumor enriched myeloid cells exhibits the highest immune-inhibitory signatures in big data.

a–c. UMAP projection of 455,959 immune cells in 131 CRC patients from syn26844071, GSE188711 and GSE178341, colored by cell type (a), tissue (b) and TNM stage (c). d. Immune-inhibitory scores of each cell type in tumor tissues. e. The significantly enriched signatures of TAM specific genes. Color depth represents -Log10 (p value). f-k. The expression of immune-inhibitory genes on macrophages (f) and neutrophils (g) from all the 131 CRC patients, as well as macrophages from stage I (h), stage II (i), stage III (j), and stage IV (k), between tumor and normal tissues (Wilcoxon test, two-sided).

Source data

Extended Data Fig. 3 The basic information of Sirpα knockout mice and its baseline immune profile.

a. Scheme of generation of Sirpa−/− mice. b, c. Sirpα protein in WT BMDMs and Sirpa−/− BMDMs were detected by Western blot (b) and FACS (c). d. Expression level of Sirpa, Sirpb1 and Sirpd in WT BMDMs (n = 3 samples) and Sirpa−/− BMDMs (n = 3 samples) were measured by RT–qPCR. e. Comparison of myeloid cell subsets in intestine between WT mice (n = 7 samples) and Sirpa−/− mice (n = 7 samples). f. Comparison of T cell subsets in intestine between WT mice (n = 7 samples) and Sirpa-/- mice (n = 6 samples). g. Comparison of fraction of NKT cells, NK cells, total myeloid cells, total T cells and B cells in all intestinal immune cells between WT mice (n = 7 samples) and Sirpa-/- mice (n = 7 or 6 samples). h-i. The gating strategies of myeloid cells (h) or lymphocytes (i). All samples were isolated from independent mice. Results (b, c) are from one representative of three independent experiments. Results (d-g) are from one independent experiment. Error bars show mean ± s.e.m. Unpaired Student’s t-test was used for d-g.

Source data

Extended Data Fig. 4 Sirpα deficiency suppresses tumor growth and Sirpα is associated with poor patient prognosis.

a. Survival analysis of WT mice (n = 12 mice) and Sirpa-/- mice (n = 13 mice) that bearing subcutaneous LLC tumor. b. Survival analysis of WT mice (n = 7 mice) and Sirpa-/- mice (n = 7 mice) that bearing subcutaneous Hepa1-6 tumor. c, e, g. The overall survival in patients with SIRPAhigh and SIPRAlow in colon cancer (GSE17536 and GSE17537, n = 232) (c), lung squamous carcinoma (TCGA-LUSC, n = 493) (e) and liver cancer (TCGA-LIHC, n = 365) (g). d, f, h. The disease-free survival in patients with SIRPAhigh and SIPRAlow in colon cancer (GSE17536 and GSE17537, n = 187) (d), lung squamous carcinoma (TCGA-LUSC, n = 476) (f) or liver cancer (TCGA-LIHC, n = 359) (h). i-j. Change of body weight (i) and disease activity index (j) for colitis of WT mice (n = 7 mice) and Sirpa-/- mice (n = 7 mice) during DSS induction. Disease activity index was calculated by summing the scores for body weight loss. k. The expression level of CD47 in MC38-Vector and MC38-CD47-/- cells in basal and inflammatory conditions (n = 3 independent experiments). l. The growths of MC38 tumors in WT mice without treatment (Ctrl; n = 6 mice), WT mice treated with anti-CD47 (n = 6 mice), WT mice treated with Sirpa-ED-FC protein (n = 6 mice) and Sirpa-/- mice without treatment (n = 9 mice). Data are pooled from 2 independent experiments (a), 3 independent experiments (k) or from one representative of five independent experiments(b), and from one independent experiment (i, j, l). P values (a-h) were performed with the log-rank test. P values (i, j, l) were used unpaired Student’s two-tailed t-test, and P values (k) were used ratio paired Student’s two-tailed t-test. Error bars show mean ± s.e.m.

Source data

Extended Data Fig. 5 Sirpα deficiency reshapes cell constitution and immune function of TIICs.

a. t-SNE visualization of all immune cells colored by origins of WT mice and Sirpa-/- mice b. Average expression of top five differentially expressed genes in each cell type of all immune cells. c. Cell proportion of each cell type in AOM/DSS-induced tumor in WT mice and Sirpa-/- mice calculated by scDC. The p value were calculated by bootstrap resampling through GLM model in scDC. d. Percentages of immune cells in AOM/DSS-induced spontaneous colorectal tumors in WT mice (n = 9-12 samples) or Sirpa-/- mice (n = 6-9 samples) analyzed by flow cytometry. e. The number of differentially expressed genes of each cell type between AOM/DSS-induced CRC of WT mice and its Sirpa-/- mice counterpart. f. Comparisons of antigen presentation score, phagocytosis score and inflammatory response score of mMDSC between AOM/DSS-induced CRC of Sirpa-/- mice and WT mice using QuSAGE. X-axis represents log2 (fold change) of the gene set score. g-i. The significantly enriched signatures of ILCs (g), B cells (h) and plasma cells (i) in AOM/DSS-induced CRC of Sirpa-/- mice compared with their counterpart in WT mice (Wilcoxon test, two-sided). All samples were isolated from independent mice. Data (d) are mean ± s.e.m pooled from 4 independent experiments, and performed with unpaired Student’s two-tailed t-test.

Source data

Extended Data Fig. 6 Sirpα deficiency reprogramed macrophage subsets and lineage in tumor.

a. Expression level of the top 3 subset-specific genes of each subset in all myeloid cell subsets. b. The top significantly enriched signatures of cluster mMDSC1-2 and TAM1-4. c. Smoothed gene expression trends of the top 100 genes whose expression values correlate best with lineage 1 (to TAM3) fate probabilities, sorted according to peak in pseudotime. Not all gene names are shown. d. Smoothed expression trends in pseudotime for the lineage 1 (to TAM3) related genes Dnase1l3, Cd81, S100a10 and Tmsb10 (95% CI). e. Expression of upregulated genes (top) and down-regulated genes (bottom) in TAM3 compared to TAM2. All genes show significantly statistic differences between TAM3 and TAM2 subsets (Wilcoxon test, two-sided, p < 0.05).

Source data

Extended Data Fig. 7 Sirpα deficiency reprogramed gMDSCs subsets and lineage in tumor.

a. The significantly enriched signatures of cluster gMDSC1-gMDSC8. b, c. Smoothed gene expression trends of the top 100 genes whose expression values correlate best with the fate probabilities of lineage 1 (to gMDSC7) (b) or lineage 2 (to gMDSC8) (c), sorted according to peak in pseudotime. Not all gene names are shown. d. Smoothed expression trends in pseudotime for the lineage 1 (to gMDSC7) and lineage 2 (to gMDSC8) related genes Ifit3, Rsad2, Ftl1 and Cstb (95% CI). e. Expression of upregulated genes (top) and down-regulated genes (bottom) in gMDSC6/gMDSC7 compared to gMDSC5. All genes show significantly statistic differences between gMDSC6/gMDSC7 and gMDSC5 subsets (Wilcoxon test, two-sided, p < 0.05).

Source data

Extended Data Fig. 8 Sirpα-/- gMDSCs have weak effect in antitumor immunity and TAMs.

a. t-SNE visualization of Sirpa in TIICs from WT mice, colored by expression level. b. Expression of Sirpa in each immune cell type in mouse tumor microenvironment. Middle white dot and line represent median and quartile values. c. The growth of subcutaneous MC38 tumor in WT (n = 9 mice), Sirpa-/- (n = 6 mice), WT + αLy6G (n = 7 mice) and Sirpa-/- + αLy6G group (n = 5 mice). d. The growth of subcutaneous MC38 tumor in WT (n = 5 mice), Sirpa-/- (n = 5 mice), WT + WT-MDSCs (n = 6 mice) and WT + Sirpa-/–MDSCs group (n = 7 mice). e. Enrichment signatures of differentially expressed genes of TAMs between Sirpa-/- mice and WT mice in MC38 model (bulk RNA-seq data) (Wilcoxon test, two-sided). f. Heat map of differentially expressed genes of TAMs between Sirpa-/- and WT mice in MC38 model, in which mainly showed the genes related to chemokine, inflammatory response, phagocytosis and antigen presentation (bulk RNA-seq data). g. Enrichment signatures of Sirpa-/- mice highly expressed genes in TAMs shared between MC38 model (bulk RNA-seq data) and AOM/DSS model (scRNA-seq data) (Wilcoxon test, two-sided). Result (c) is one representative of three independent experiments. Result (d) is from one independent experiment. Error bars show mean ± s.e.m. Unpaired Student’s two-tailed t-test was used for c, d.

Source data

Extended Data Fig. 9 Sirpa-/- myeloid cells enhance the activation of T lymphocytes.

a, b. Expression of differentially expressed genes in CD8+ T cell (a) or CD4+ T cell (b) between WT mice and Sirpa-/- mice in AOM/DSS-induced CRC (Wilcoxon test, two-sided). c-d. Tumor loads (c) and tumor numbers (d) were measured in AOM/DSS-induced WT mice and Sirpa-/- mice treated with CD4+ or CD8+ antibody, WT (n = 9 mice), Sirpa-/- (n = 6 mice), WT + αCD4 (n = 5 mice), Sirpa-/- + αCD4 (n = 3 mice), WT + αCD8 (n = 3 mice), Sirpa-/- + αCD8 (n = 4 mice). e. The L–R pairs in cell–cell contact signaling from TAM, mMDSC, cDC1 and cDC2 (sender) to other cell types (receiver) in AOM/DSS-treated WT mice and Sirpa-/- mice calculated by CellChat. Line width indicates signal strength and line color is the same as signaling sender. f. The dominant sender and receiver cell types of cell–cell contact signaling network calculated by CellChat. Count represents the number of L–R pairs. g. The CD8+ T cells proliferation suppressed by WT (n = 3 samples) or Sirpa-/- (n = 3 samples) gMDSCs at different ratios. All samples were isolated from independent mice Result (c, d, g) are from one independent experiment. Error bars show mean ± s.e.m. Unpaired Student’s two-tailed t-test was used for c, d, g.

Source data

Extended Data Fig. 10 Sirpa-/- myeloid cells enhance the recruitment of T lymphocytes.

a. The L–R pairs in secreting signaling from mMDSC, TAM and gMDSC (sender) to other cell types (receiver) in AOM/DSS-treated WT mice and Sirpa-/- mice calculated by CellChat. Line width indicates signal strength and line color is the same as signaling sender. b. The dominant sender and receiver cell types of chemokine signaling pathways (CCL and CXCL pathways) calculated by CellChat. Count represents the number of L–R pairs. c. The gene expression levels of various chemokines in TAMs of MC38 tumors from WT mice (n = 4 samples) or Sirpa-/- mice (n = 5 samples). d. Boxplot showing the SIRPA expression on macrophages in recurrent GBM patients pre- and post-neoadjuvant PD-1 blockade treatment. All samples were isolated from independent mice.The two-sided unpaired t-test was used to calculate P value. Result (c) is from one independent experiment. Error bars show mean ± s.e.m. Unpaired Student’s two-tailed t-test was used for c.

Source data

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Tables 1–4.

Source data

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. 3

Unprocessed H&E staining of colorectal tissue from WT mice (with tumor) and Sirpa−/− mice (without tumor).

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. 3

Unprocessed western blot for detection of SIRPA in WT mice and Sirpa−/− mice.

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.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10.

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

Huang, C., Wang, X., Wang, Y. et al. Sirpα on tumor-associated myeloid cells restrains antitumor immunity in colorectal cancer independent of its interaction with CD47. Nat Cancer 5, 500–516 (2024). https://doi.org/10.1038/s43018-023-00691-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00691-z

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