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:

Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells

Abstract

Macrophages have a key role in shaping the tumour microenvironment (TME), tumour immunity and response to immunotherapy, which makes them an important target for cancer treatment1,2. However, modulating macrophages has proved extremely difficult, as we still lack a complete understanding of the molecular and functional diversity of the tumour macrophage compartment. Macrophages arise from two distinct lineages. Tissue-resident macrophages self-renew locally, independent of adult haematopoiesis3,4,5, whereas short-lived monocyte-derived macrophages arise from adult haematopoietic stem cells, and accumulate mostly in inflamed lesions1. How these macrophage lineages contribute to the TME and cancer progression remains unclear. To explore the diversity of the macrophage compartment in human non-small cell lung carcinoma (NSCLC) lesions, here we performed single-cell RNA sequencing of tumour-associated leukocytes. We identified distinct populations of macrophages that were enriched in human and mouse lung tumours. Using lineage tracing, we discovered that these macrophage populations differ in origin and have a distinct temporal and spatial distribution in the TME. Tissue-resident macrophages accumulate close to tumour cells early during tumour formation to promote epithelial–mesenchymal transition and invasiveness in tumour cells, and they also induce a potent regulatory T cell response that protects tumour cells from adaptive immunity. Depletion of tissue-resident macrophages reduced the numbers and altered the phenotype of regulatory T cells, promoted the accumulation of CD8+ T cells and reduced tumour invasiveness and growth. During tumour growth, tissue-resident macrophages became redistributed at the periphery of the TME, which becomes dominated by monocyte-derived macrophages in both mouse and human NSCLC. This study identifies the contribution of tissue-resident macrophages to early lung cancer and establishes them as a target for the prevention and treatment of early lung cancer lesions.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: scRNA-seq of lineage-traced blood-derived macrophages reveals two ontogenically distinct populations of macrophages in NSCLC lesions.
Fig. 2: TRMs localize in close proximity to tumour cells after seeding and enhance their antigen presentation and tissue remodelling programs in response to tumour cues.
Fig. 3: TRMs induce NSCLC cells to undergo EMT and promote tumour cell invasiveness.
Fig. 4: Depletion of TRMs before tumour engraftment leads to reduced tumour burden and enhances T cell infiltration.

Similar content being viewed by others

Data availability

All mice sequencing data are publicly available (GEO accession code GSE147671). The human dataset is available at the Sequence Read Archive (SRA) with BioProject accession PRJNA609924Source data are provided with this paper.

Code availability

The clustering analysis used here is described fully in its source manuscript6.

References

  1. Lavin, Y., Mortha, A., Rahman, A. & Merad, M. Regulation of macrophage development and function in peripheral tissues. Nat. Rev. Immunol. 15, 731–744 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mantovani, A., Marchesi, F., Malesci, A., Laghi, L. & Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 14, 399–416 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hashimoto, D. et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 38, 792–804 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86–90 (2012).

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Leader, A. M. et al. CITEseq analysis of non-small-cell lung cancer lesions reveals an axis of immune cell activation associated with tumor antigen load and TP53 mutations. Preprint at https://doi.org/10.1101/2020.07.16.207605 (2020).

  7. Martin, J. C. et al. Single-cell analysis of Crohn’s disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. Cell 178, 1493–1508 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Schneider, C. et al. Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat. Immunol. 15, 1026–1037 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Xue, W. et al. Response and resistance to NF-κB inhibitors in mouse models of lung adenocarcinoma. Cancer Discov. 1, 236–247 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Leach, S. M. et al. Human and mouse transcriptome profiling identifies cross-species homology in pulmonary and lymph node mononuclear phagocytes. Cell Rep. 33, 108337 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sawai, C. M. et al. Hematopoietic stem cells are the major source of multilineage hematopoiesis in adult animals. Immunity 45, 597–609 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Russell, D. G., Cardona, P. J., Kim, M. J., Allain, S. & Altare, F. Foamy macrophages and the progression of the human tuberculosis granuloma. Nat. Immunol. 10, 943–948 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. DuPage, M., Dooley, A. L. & Jacks, T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nat. Protocols 4, 1064–1072 (2009).

    Article  CAS  PubMed  Google Scholar 

  16. Sutherland, K. D. et al. Multiple cells-of-origin of mutant K-Ras-induced mouse lung adenocarcinoma. Proc. Natl Acad. Sci. USA 111, 4952–4957 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. Lim, J. & Thiery, J. P. Epithelial–mesenchymal transitions: insights from development. Development 139, 3471–3486 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Ye, X. et al. Distinct EMT programs control normal mammary stem cells and tumour-initiating cells. Nature 525, 256–260 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Rhim, A. D. et al. EMT and dissemination precede pancreatic tumor formation. Cell 148, 349–361 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Krebs, A. M. et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat. Cell Biol. 19, 518–529 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Thiery, J. P. Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2, 442–454 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Yang, J. et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117, 927–939 (2004).

    Article  CAS  PubMed  Google Scholar 

  24. Dongre, A. & Weinberg, R. A. New insights into the mechanisms of epithelial–mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 20, 69–84 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Lavin, Y. et al. innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169, 750–765 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tada, Y. et al. Targeting VEGFR2 with Ramucirumab strongly impacts effector/ activated regulatory T cells and CD8+ T cells in the tumor microenvironment. J. Immunother. Cancer 6, 106 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Battaglia, A. et al. Metastatic tumour cells favour the generation of a tolerogenic milieu in tumour draining lymph node in patients with early cervical cancer. Cancer Immunol. Immunother. 58, 1363–1373 (2009).

    Article  PubMed  Google Scholar 

  28. He, F. et al. PLAU inferred from a correlation network is critical for suppressor function of regulatory T cells. Mol. Syst. Biol. 8, 624 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Mucida, D. et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 317, 256–260 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  30. Soroosh, P. et al. Lung-resident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J. Exp. Med. 210, 775–788 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mizukami, Y. et al. CCL17 and CCL22 chemokines within tumor microenvironment are related to accumulation of Foxp3+ regulatory T cells in gastric cancer. Int. J. Cancer 122, 2286–2293 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Liu, Z. et al. Fate mapping via Ms4a3-expression history traces monocyte-derived cells. Cell 178, 1509–1525 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Miyake, Y. et al. Critical role of macrophages in the marginal zone in the suppression of immune responses to apoptotic cell-associated antigens. J. Clin. Invest. 117, 2268–2278 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Karasawa, K. et al. Vascular-resident CD169-positive monocytes and macrophages control neutrophil accumulation in the kidney with ischemia-reperfusion injury. J. Am. Soc. Nephrol. 26, 896–906 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Zhang, Y. et al. Model-based analysis of ChIP–seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Sastre-Perona, A. et al. De novo PITX1 expression controls bi-stable transcriptional circuits to govern self-renewal and differentiation in squamous cell carcinoma. Cell Stem Cell 24, 390–404 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Schmidt, U., Weigert, M., Broaddus, C. & Myers, G. Cell detection with star-convex polygons. In International Conference on Medical Image Computing and Computer-Assisted Intervention –MICCAI 2018 (eds Frangi, A. et al.) 265–273 (Springer, 2018).

  46. Remark, R. et al. In-depth tissue profiling using multiplexed immunohistochemical consecutive staining on single slide. Sci. Immunol. 1, aaf6925 (2016).

    Article  PubMed  Google Scholar 

  47. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by an HFSP postdoctoral fellowship (LT000110/2015-L/1) and an AACR–AstraZeneca Immuno-oncology Research Fellowship (20-40-12-CASA) to M.C.-A.; T32 CA078207 to A.L.; F30 CA243210 to S.T.C.; and ANR-10-IDEX-0001-02 PSL and ANR-11-LABX-0043 and Fondation ARC pour la recherche sur le cancer to P.B. This research was supported in part by the Tisch Cancer Institute at Mount Sinai P30 CA196521-Cancer Center Support Grant. We thank the Human Immune Monitoring Center for all the single-cell profiling and epigenetic studies; the Merad laboratory and A. Lujambio for discussions and reagents; the Flow Cytometry and the Imaging Core at Mount Sinai; and the Cancer Biorepository at MSSM for sample acquisition. This work was also supported by National Institutes of Health (NIH)–National Cancer Institute grants CA257195, CA254104, AT011326, AI128949 and R56AI137244  to M.M., who is also a Samuel Waxman Cancer Research Foundation Investigator; CA109182, CA216248 and CA218024 to J.A.A.-G., who is also a Samuel Waxman Cancer Research Foundation Investigator; CA257195, CA254104, AT011326 and AI128949 to B.D.B.; NCI grant T32 CA078207 to E.D.; and NIH grant AG049074 to B.R.

Author information

Authors and Affiliations

Authors

Contributions

M.M. and M.C.-A. conceived the project. M.M., M.C.-A., P.B. and J.A.A.-G. designed the experiments. M.C.-A., E.D., J.A.A.-G. and M.M. wrote the manuscript. A.L., C.C., A.S.-P., E.A.-P., M.D.P. and E.K. performed computational analysis. M.C.-A., E.D., J.N., B.M.M., S.T.C., M.D.P., M.D., A.T., L.T., P.H., B.M. and J.L. performed experiments. B.B. and M.S. provided reagents. M.B. and C.M. performed image analysis of KP tumours. T.M. provided intellectual input and facilitated access to human samples. C.M.S. and B.R. provided Pdzk1ip1-creER R26Tom/Tom mice. All authors contributed to manuscript editing.

Corresponding authors

Correspondence to María Casanova-Acebes or Miriam Merad.

Ethics declarations

Competing interests

 J.A.A.-G. is a scientific co-founder of, scientific advisory board member and equity owner in HiberCell and receives financial compensation as a consultant for HiberCell, a Mount Sinai spin-off company focused on therapeutics that prevent or delay cancer recurrence. M.M. serves on the scientific advisory boards of Compugen Inc., Innate Pharma Inc., Morphic Therapeutic, Myeloid Therapeutics, Celsius Therapeutics and Genenta. M.M. receives funding from Genentech Inc., Regeneron Inc., Boerhinger Ingelheim Inc. and Takeda Inc. M.M. has ownership interest of less than 5% in Compugen Inc., Celsius Therapeutics Inc., Dren Bio Inc., and Asher Bio Inc. B.M.M., M.B. and C.M. declare that they are Genentech/Roche employees.

Additional information

Peer review information Nature thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Macrophage distribution and profile in NSCLC lesions.

a, Gating strategy for sorting of myeloid cells from naive and KP tumour-bearing lungs of Map17creER/+R26tdTom mice for scRNA-seq analysis. Monocytes and macrophages in the lung were gated as singlets, DAPICD45+LinCD11b−/+Ly6GCD11clo-int-+SIGLECF+ and Tom or Tom+. b, Confocal imaging of tdTomato+ bone-marrow-derived leukocytes and TRMs (CD206+, yellow). Images are representative of a single experiment and three tumours imaged. c, Gating strategy for sorting of myeloid cells from naive and KP tumour-bearing lungs of Cx3cr1creER/+R26YFP mice. Monocytes and macrophages were gated as in a and further separated based on YFP expression. d, Confocal imaging of YFP+ bone-marrow-derived leukocytes (red) and TRMs (CD206+, yellow). Scale bar, 50 μm. Images are representative of a single experiment and three tumours imaged. e, Spearman correlation of variable gene expression between the human monocyte and macrophage clusters detected in ref. 6 and those in ref. 10. f, The log2-transformed fold change (FC) between human TRM expression and the maximum cluster expression of non-TRM monocytes and macrophages, determined from data in ref. 6 (x axis) or ref. 10 (y axis). The human alveolar macrophage genes published previously11 are highlighted in red. g, Spearman correlation of variable gene expression between the mouse monocyte and macrophage clusters detected in the present study and in ref. 10. h, Average expression of selected mouse genes from scRNA-seq data in cluster groups I to IV (see Supplementary Table 2). i, Average expression of selected human genes from scRNA-seq data (see Supplementary Table 1). j, Confocal imaging of CD169cre/+R26tdTom KP lesions (day 30, KP-GFP, green) with CD206 (yellow). Scale bar, 100 μm. Images are representative of a single experiment and three tumours imaged. k, Gating strategy for the identification of TRMs and MDMs in naive lungs. TRMs were gated as live/deadCD45+Ly6GCD64+MERTK+CD2+CD169+CD206+SIGLECF+; MDMs were gated as singlets, live/deadCD45+Ly6GCD64+Mertk+CD2CD11bhiCD169CD206SIGLECF.

Source data

Extended Data Fig. 2 Fate-mapping of macrophages in KP tumours.

a, Lineage tracing experiment in Map17creER/+R26tdTom mice. b, Fraction of labelled (red, tdTom+) cells in the peripheral blood and lung of non-tumour-bearing Map17creER/+R26tdTom mice 6 months after tamoxifen injection (n = 6 blood, n = 5 lung; two independent experiments). Blood Ly6Chi monocytes were identified as singlets, DAPICD45+CD11b+CD115+Ly6Chi or Ly6Clo monocytes as CD45+CD11b+CD115+Ly6Clo. Lung monocytes were gated as CD45+CD11b+CX3CR1+Ly6Chi or Ly6Clo. Neutrophils in blood and lungs were identified as singlets, DAPICD45+CD11b+Ly6G+. TRMs were identified as singlets, DAPICD45+Ly6GCD11blo/−SIGLECFhiCD11chiCD206hiCD169hi. Data are mean ± s.e.m. c, Frequencies of labelled (red, tdTom+) or not labelled (grey, tdTom) cells within each cluster groups as defined in b in tumour-bearing mice. d, Lineage tracing experiment in Cx3cr1creER/+R26YFP mice. e, Fraction of labelled (green, YFP+) cells in the peripheral blood and lung of non-tumour-bearing mice (n= 8, pool of two independent experiments). Data are mean ± s.e.m. f, Frequency of labelled (green, YFP+) and non-labelled (grey, YFP) cells within each cluster groups as defined in b in KP tumour-bearing mice.

Source data

Extended Data Fig. 3 Longitudinal analysis of TRMs in NSCLC.

a, TRMs (red, CD206+) distribution in KP-GEMMs (green) 14 weeks (wks) after injection of adenovirus-SPC5Cre in KP mice. White dotted lines delimit the tumour area. Unpaired two-tailed Student’s t-test; *P = 0.020. CD206+ macrophage distribution was analysed in n = 4 tumour-bearing mice from one experiment. Data are mean ± s.e.m. b, Gene Ontology categories for ATAC–seq significant (P < 0.05) open (day 15) and closed (day 30) chromatin regions identified in KP-associated TRMs. c, Longitudinal analysis of CD45+ leukocytes, TRMs and Ly6Chi and Ly6Clo monocytes in naive (n = 5) and in day-15 (n = 5) and day-30 (n = 4) KP-bearing mice. One-way ANOVA with Tukey’s test. Data are mean ± s.e.m. TRMs were gated as singlets, live/deadGFPCD45+Ly6GCD64+MERTK+CD2+CD169+CD206+Siglecf+; MDMs were gated as singlets, live/deadGFPCD45+Ly6GCD64+MERTK+CD2CD11bhiCD169SIGLECF. Lung monocytes were gated as CD45+CD11b+CX3CR1+Ly6Chi or Ly6Clo. d, Longitudinal imaging analysis of TRMs identified by the co-expression of CD206 (red) and SIGLECF (yellow) in the KP-GFP orthotopic model. KP tumour cells, green. White asterisks indicate CD206+SIGLECF macrophages. Yellow asterisks depict SIGLECF+CD206 leukocytes, which are also found in overt tumours and are most probably SIGLECF+ eosinophils. Scale bars, 50 μm (D5, D10 and D15); 100 μm (D25–30). Images are representative of one experiment; n = 3–5 mice; 2–3 tumours analysed per time point. e, Longitudinal imaging analysis of TRMs in the KP-GEMM model. Tumour cells are identified by positive staining with pan-cytokeratin in green. Images are representative of one experiment; 2–3 tumours analysed per mouse; 3 mice per time point. Scale bars, 50 μm. f, Immunohistochemistry converted to pseudofluorescence image of CD206 (red), CD10 (yellow) and cytokeratin (CK, green) staining in non-involved lung and NSCLC tissue. White asterisks indicate CD206+CD10+ TRMs. Scale bars, 250 μm (bottom images); 250 μm (top images). Images are representative of one experiment. g, Immunohistochemistry converted to pseudofluorescence image of CD206 (red), FABP4 (yellow) and cytokeratin (CK, green) staining in non-involved lung and NSCLC tissue. White asterisks indicate CD206+FABP4+ TRMs. Scale bars, 500 μm (left); 400 μm (right). Dotted lines delineate tumour border. Representative images from two non-involved lung and two NSCLC tumours. Images are representative of one experiment.

Source data

Extended Data Fig. 4 Bulk RNA-seq and ATAC–seq of KP TRMs.

a, Heat map of DEGs of TRMs in naive lungs, day-15 and day-30 KP tumours. Red indicates the most significant upregulated and blue the most significant downregulated gene transcripts (P < 0.05, log2-transformed fold change (log2FC) > 1 and log2FC < 1, respectively). TRMs were sorted as singlets, DAPICD45+Ly6GCD11blo/−CD64+MERTK+CD2+CD169+SIGLECFhiCD206+. b, Gene Ontology analysis of upregulated DEGs between naive and early KP-TRMs (day 15) (P < 0.05 and log2FC > 1). c, Number of peaks and heat map representing average ATAC–seq peaks (pks) unchanged (cluster 1), differentially closed (cluster 2) or opened (cluster 3) in TRMs at different times after KP injection. d, e, Representative tracks of significant TRM DEGs (P < 0.05) showing increased chromatin accessibility (dotted red lines) (d) or lower chromatin accessibility (e) in TRMs. Tracks are representative of three pooled mice examined over one single experiment. Data are mean ± s.e.m.

Source data

Extended Data Fig. 5 TRMs promote EMT and a pro-invasive signature in KP spheroids, whereas MDMs favour growth.

a, Venn diagrams for DEGs upregulated and downregulated in spheroids co-cultured with TRMs or bone marrow monocytes (BMMs). The number of DEGs uniquely controlled by TRMs or BMMs is shown in blue (TRMs) and red (BMMs). b, Gene Ontology (GO) analysis of significant DEGs (P < 0.05) with upregulated signature controlled by TRMs (blue) and by BMMs (red), respectively. c, Gating strategy for BALF (bronchoalveolar fluid) TRMs gated as CD45+CD11bloCD11c+SIGLECF+CD206+CD169+ and purity quantification in n = 4 mice. BALF routinely showed around 85% pure TRMs among CD45+ leukocytes. d, Confocal representative images and quantification of E-cadherin (red), TWIST1 (white) and β-catenin (red) in KP spheroids cultured alone or with TRMs or BMMs. Scale bars, 5 μm (inset) and 25 μm. One-way ANOVA with Tukey’s test. Data are mean ± s.e.m. Two independent experiments. e, Bar graphs showing the expression (in transcripts per million, TPM) of EMT-signature selected genes in KP-spheroids alone, with TRMs or BMMs. Data are mean ± s.e.m. Results are representative of one experiment. f, Size quantification for KP oncospheres co-cultured with TRMs or BMMs. Data are mean ± s.e.m. Data are representative of two independent experiments. g, Quantification of the number of KP oncospheres in co-cultures with TRMs (blue) or BMMs (red) compared to KP alone in the presence of GM-CSF (light blue) or M-CSF (light red), respectively. Data are mean ± s.e.m. Data are representative of two independent experiments. h, Quantification of KP 3D-matrigel spheroids with invasive protrusions co-cultured with TRMs or BMMs and their respective controls. Data are mean ± s.e.m. Data are representative of two independent experiments. One-way ANOVA with Tukey’s test (fh). i, Bright-field microscopy images of the spheroids quantified in h. Scale bars, 100 μm. j, k, Number (j) and size (k) of KP oncospheres cultured alone or co-cultured with tumour-associated tTRMs or tMDMs. Results are representative of two independent experiments analysed using one-way ANOVA. Data are mean ± s.e.m. *P < 0.05, **P < 0.01, ****P < 0.0001.

Source data

Extended Data Fig. 6 Immune suppression is governed by TRMs in early KP tumours.

a, Frequency, CTLA-4 and CD73 MFI levels of Treg cells induced by tTRMs and tMDMs with naive CD62L+CD44CD4+ T cells (one-way ANOVA with Tukey’s test and two-tailed unpaired t-test). Two independent experiments. b, Frequency and phenotype of Treg cells in TRM-depleted mice at day 15 after KP injection. n = 5 mice per group. Two-tailed unpaired t-test; **P = 0.003, *P = 0.025; ***P = 0.0003 and ****P < 0.0001, respectively. c, Expression of Ccl17 and Tgfb1 in TRMs from naive mice (grey) and tumour-bearing mice (day 15 light green, day 30 dark green). Data from DEGs (P < 0.05 likelihood ratio test) list in Fig. 2d (see Supplementary Table 3). One experiment, n = 3 naive, n = 2 KP-TRM day 15 and n = 3 KP-TRM day 30 mice. d, Imaging of TRM-sufficient and deficient mice after instillation of diphtheria toxin. SIGLECF, green; CD206, red. Quantification of Ly6Chi/lo monocytes, neutrophils, MDMs and TRMs in wild-type or CD169-DTR lungs one week after the last dose of diphtheria toxin. n = 5 mice per group. Two-tailed unpaired t-test. TRMs were gated as singlets, live/deadCD45+Ly6GCD64+MERTK+CD2+CD169+SIGLECF+CD206+; MDMs as singlets, live/deadCD45+Ly6GCD64+MERTK+CD2CD11bhiCD169SIGLECF; monocytes as CD45+CD11b+CX3CR1+Ly6Chi or Ly6Clo (Ly6Chi and Ly6Clo, respectively); and neutrophils as live/deadCD45+CD11b+Ly6G+. e, Levels of CD169 in Tomato+ monocytes (CD45+CD11b+CX3CR1+) and MDMs (live/deadCD45+Ly6GCD64+MERTK+CD2CD11bhiSIGLECF) in naive and two-week KP lesions from Ms4a3-tdTom reporter mice. n = 3 mice per group. Two-way ANOVA with Tukey’s multiple comparisons test; ns, not significant. f, Frequencies of MDMs in wild-type and CD169-DTR lungs after DT treatment with diphtheria toxin in naive mice and KP lesions (two weeks). n = 5 per genotype for naive mice and n = 3 per genotype for the KP tumour group. Two-way ANOVA with Tukey’s multiple comparisons test. g, Quantification of Treg cells in spleen and lymph nodes of tumour-bearing mice (day 15) in WT + DT (black) and CD169-DTR + DT mice (red). n = 6 mice per group. Unpaired two-tailed t-test. h, Percentage of KP cells from lungs of wild-type or CD169-DTR mice treated with diphtheria toxin, 24 h after KP injection. n = 6 mice per group. Two-tailed unpaired t-test. i, Image analysis of Ki67+ and CC3+ KP cells in day-15 lesions, and p27+ KP cells in day-5 lesions of WT + DT and CD169-DTR + DT mice. Asterisks show positive KP cells. One-tailed unpaired t-test. n = 3 WT + DT and n = 4 CD169-DTR + DT for Ki67 and CC3; n = 3 WT + DT and n = 4 CD169-DTR + DT for p27. Two to three independent experiments. Scale bars, 25 μm (main images); 10 μm (inset). Data are mean ± s.e.m. (ai). j, Diphtheria toxin treatment and KP injections in wild-type or CD169-DTR mice. k, Tumour burden in wild-type or CD169-DTR mice that were TRM-depleted after tumour implantation. l, Quantification of Treg cells, IFNγ+TNF+CD8+ effector cells and ratio of CD8/Treg cells in mice from k. Effector T cells were gated as singlets, DAPICD45+TCR+CD8+; Treg cells as singlets, DAPICD45+TCR+CD4+FOXP3+. n = 5 WT + DT and n = 7 CD169-DTR + DT mice. Three independent experiments. Data are mean ± s.e.m; two-tailed unpaired t-test. (k, l). m, Imaging and quantification of infiltrating FOXP3+ Treg cells (top) and CD3+ T cells (bottom) in WT + DT or CD169-DTR + DT mice at days 12 and 15 after KP injection. Two-tailed unpaired t-test. Data are mean ± s.e.m. Two independent experiments. Scale bar, 100 μm.

Extended Data Fig. 7 TRMs modulate T cell effector function in an antigen-independent manner.

a, Scheme of OT-I and OT-II adoptive transfer experiments in B16-F10/OVA wild-type and CD169-DTR mice. b, Relative quantification of OT-I T cells in the lungs and tumour-draining lymph nodes (tdLN). OT-I T cells were gated as viable, CD45.1+TCR+CD8+. n = 4—5 mice. Data are representative of two independent experiments. c, Quantification of OT-I cells in the tumours of mice in b. CD45.1+ OT-I T cells were quantified in 7–8 tumours from mice in b. n= 7 WT + DT and N = 8 CD169-DTR + DT from one experiment. Scale bar, 50 μm. d, Quantification of OT-II cells in the tumours of mice in b. OT-II T cells were gated as viable, CD45.1+TCR+CD4+. n = 5–8 mice per group. Data are representative of one experiment. Data are mean ± s.e.m; unpaired two-tailed t-test (bd). e, Scheme of the contribution of the TRM compartment and MDMs to tumour progression. This scheme was created with BioRender.com.

Supplementary information

Reporting Summary

Supplementary Table 1

scRNAseq differential average expression of cells per group over transcript expression in all groups of monocytes and macrophages in human.

Supplementary Table 2

scRNAseq differential average expression of cells per group over transcript expression in all groups of monocytes and macrophages in mouse.

Supplementary Table 3

Differential expression gene list in bulk RNA-seq of TRMs (alveolar macrophages, AM) in naïve, KP D15 and KP D30 tumours. Significant genes (KP D15 over naïve) are included (p-value<0.05). All data is also included in this table with no further statistical test.

Supplementary Table 4

Differential expression ATAC-seq peaks detected in TRMs in naïve, KP D15 and KP D30 tumours. Significant genes (KP D15 over naïve and KP D30 over naïve) are included (p-value<0.05).

Supplementary Table 5

Differential expression gene list in bulk RNA-seq of KP-spheroids co-cultured with TRMs and BMMs. Significant genes uniquely found in either KP+TRM over KP alone, and KP+BMM over KP alone are included (p-value<0.05), as well as significant genes found in both conditions.

Supplementary Tables 6-8

These tables include the list of antibodies used for flow cytometry (Supplementary Table 6), antibodies used for Multiplexed immunohistochemical consecutive staining on a single slide (Supplementary Table 7) and antibodies used for Clearing imaging (Supplementary Table 8).

Supplementary Data

This zipped file includes the Supplementary Imaging Macros and a guide.

Video 1

KP-spheroids co-cultured with TRMs. KP-GFP expressing spheroids were co-cultured with alveolar macrophages and imaged for 4 hours. Invasive protrusions are visualized. Scale bar corresponds to 200 μm.

Video 2

KP-Spheroids co-cultured with BMMs. KP-GFP expressing spheroids were co-cultured with bone-marrow monocytes and imaged for 4 hours. Note the size of KP-GFP spheroids co-cultured under these conditions. Scale bar corresponds to 200 μm.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Casanova-Acebes, M., Dalla, E., Leader, A.M. et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature 595, 578–584 (2021). https://doi.org/10.1038/s41586-021-03651-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03651-8

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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