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:

TREM2 macrophages drive NK cell paucity and dysfunction in lung cancer

Abstract

Natural killer (NK) cells are commonly reduced in human tumors, enabling many to evade surveillance. Here, we sought to identify cues that alter NK cell activity in tumors. We found that, in human lung cancer, the presence of NK cells inversely correlated with that of monocyte-derived macrophages (mo-macs). In a murine model of lung adenocarcinoma, we show that engulfment of tumor debris by mo-macs triggers a pro-tumorigenic program governed by triggering receptor expressed on myeloid cells 2 (TREM2). Genetic deletion of Trem2 rescued NK cell accumulation and enabled an NK cell-mediated regression of lung tumors. TREM2+ mo-macs reduced NK cell activity by modulating interleukin (IL)-18/IL-18BP decoy interactions and IL-15 production. Notably, TREM2 blockade synergized with an NK cell-activating agent to further inhibit tumor growth. Altogether, our findings identify a new axis, in which TREM2+ mo-macs suppress NK cell accumulation and cytolytic activity. Dual targeting of macrophages and NK cells represents a new strategy to boost antitumor immunity.

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: TREM2+ monocyte-derived macrophages are major constituents of an NK cell-deprived microenvironment.
Fig. 2: Uptake of cellular debris induces the TREM2 gene program in mo-macs.
Fig. 3: TREM2 deficiency restricts tumor growth by remodeling the composition of MNPs.
Fig. 4: Therapeutic effect of TREM2 deficiency is critically dependent on intact IL-18 and IL-15 signaling.
Fig. 5: Therapeutic inhibition of TREM2 synergizes with an NK-cell-stabilizing agent to promote superior NK cell immunity and tumor elimination.

Similar content being viewed by others

Data availability

All murine sequencing data will be made publicly available (GSE184304, GSE184309 and GSE184317). The human dataset is available at the Sequence Read Archive with BioProject accession no. PRJNA609924. Source data are provided with this paper.

Code availability

All murine sequencing code will be made publicly available (GSE184304, GSE184309 and GSE184317). No custom code was generated for this study. Code for generating figures can be provided upon reasonable request.

References

  1. Dunn, G. P., Bruce, A. T., Ikeda, H., Old, L. J. & Schreiber, R. D. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol. 3, 991–998 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Molgora, M., Cortez, V. S. & Colonna, M. Killing the invaders: NK cell impact in tumors and anti-tumor therapy. Cancers 13, 595 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gasser, S., Orsulic, S., Brown, E. J. & Raulet, D. H. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436, 1186–1190 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Raulet, D. H., Gasser, S., Gowen, B. G., Deng, W. & Jung, H. Regulation of ligands for the NKG2D activating receptor. Annu. Rev. Immunol. 31, 413–441 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hayakawa, Y. & Smyth, M. J. NKG2D and cytotoxic effector function in tumor immune surveillance. Semin. Immunol. 18, 176–185 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Smyth, M. J., Hayakawa, Y., Takeda, K. & Yagita, H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat. Rev. Cancer 2, 850–861 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Parham, P. & Guethlein, L. A. Genetics of natural killer cells in human health, disease, and survival. Annu. Rev. Immunol. 36, 519–548 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Garrido, F., Aptsiauri, N., Doorduijn, E. M., Garcia Lora, A. M. & van Hall, T. The urgent need to recover MHC class I in cancers for effective immunotherapy. Curr. Opin. Immunol. 39, 44–51 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171, 1259–1271 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cózar, B. et al. Tumor-infiltrating natural killer cells. Cancer Discov. 11, 34–44 (2021).

    Article  PubMed  Google Scholar 

  11. Shimasaki, N., Jain, A. & Campana, D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 19, 200–218 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. 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 

  13. Leader, A. M. et al. Single-cell analysis of human non-small cell lung cancer lesions refines tumor classification and patient stratification. Cancer Cell 39, 1594–1609 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Mulder, K. et al. Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity 54, 1883–1900 (2021).

    Article  CAS  PubMed  Google Scholar 

  15. DeNardo, D. G. et al. CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell 16, 91–102 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Masetti, M. et al. Lipid-loaded tumor-associated macrophages sustain tumor growth and invasiveness in prostate cancer. J. Exp. Med. 219, e20210564 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. 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 

  18. Park, M. D., Silvin, A., Ginhoux, F. & Merad, M. Macrophages in health and disease. Cell 185, 4259–4279 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Casanova-Acebes, M. et al. Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC cells. Nature 595, 578–584 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Loyher, P.-L. et al. Macrophages of distinct origins contribute to tumor development in the lung. J. Exp. Med. 215, 2536–2553 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Guerreiro, R. et al. TREM2 variants in Alzheimer’s disease. N. Engl. J. Med. 368, 117–127 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Sims, R. et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat. Genet. 49, 1373–1384 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang, S. et al. Anti-human TREM2 induces microglia proliferation and reduces pathology in an Alzheimer’s disease model. J. Exp. Med. 217, e20200785 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Zhou, Y. et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nat. Med. 26, 131–142 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Binnewies, M. et al. Targeting TREM2 on tumor-associated macrophages enhances immunotherapy. Cell Rep. 37, 109844 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Katzenelenbogen, Y. et al. Coupled scRNA-seq and Intracellular protein activity reveal an immunosuppressive role of TREM2 in cancer. Cell 182, 872–885 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. 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 

  28. Esparza-Baquer, A. et al. TREM-2 defends the liver against hepatocellular carcinoma through multifactorial protective mechanisms. Gut 70, 1345–1361 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Perugorria, M. J. et al. Non-parenchymal TREM-2 protects the liver from immune-mediated hepatocellular damage. Gut 68, 533–546 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Tang, W. et al. TREM2 acts as a tumor suppressor in hepatocellular carcinoma by targeting the PI3K/Akt/β-catenin pathway. Oncogenesis 8, 9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Heng, T. S. P. & Painter, M. W. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Cannon, J. P., O’Driscoll, M. & Litman, G. W. Specific lipid recognition is a general feature of CD300 and TREM molecules. Immunogenetics 64, 39–47 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. Daws, M. R. et al. Pattern recognition by TREM-2: binding of anionic ligands. J. Immunol. 171, 594–599 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Filipello, F. et al. The microglial innate immune receptor TREM2 is required for synapse elimination and normal brain connectivity. Immunity 48, 979–991 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Hsieh, C. L. et al. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. J. Neurochem. 109, 1144–1156 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Iizasa E. et al. TREM2 is a receptor for non-glycosylated mycolic acids of mycobacteria that limits anti-mycobacterial macrophage activation. Nat. Commun. 12, 2299 (2021).

  38. Lue, L.-F., Schmitz, C. & Walker, D. G. What happens to microglial TREM2 in Alzheimer’s disease: Immunoregulatory turned into immunopathogenic? Neuroscience 302, 138–150 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Shirotani, K. et al. Aminophospholipids are signal-transducing TREM2 ligands on apoptotic cells. Sci. Rep. 9, 7508 (2019).

  40. Takahashi, K., Rochford, C. D. P. & Neumann, H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J. Exp. Med. 201, 647–657 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, S. et al. Efferocytosis fuels requirements of fatty acid oxidation and the electron transport chain to polarize macrophages for tissue repair. Cell Metab. 29, 443–456 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Hannedouche, S. et al. Oxysterols direct immune cell migration via EBI2. Nature 475, 524–527 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Preuss, I. et al. Transcriptional regulation and functional characterization of the oxysterol/EBI2 system in primary human macrophages. Biochem. Biophys. Res. Commun. 446, 663–668 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Rutkowska, A. et al. The EBI2 signalling pathway plays a role in cellular crosstalk between astrocytes and macrophages. Sci. Rep. 6, 25520 (2016).

  45. Harms, R. Z., Creer, A. J., Lorenzo-Arteaga, K. M., Ostlund, K. R. & Sarvetnick, N. E. Interleukin (IL)-18 binding protein deficiency disrupts natural killer cell maturation and diminishes circulating IL-18. Front. Immunol. 8, 1020 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Zhou, T. et al. IL-18BP is a secreted immune checkpoint and barrier to IL-18 immunotherapy. Nature 583, 609–614 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Chaix, J. et al. Cutting edge: priming of NK cells by IL-18. J. Immunol. 181, 1627–1631 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Nielsen, C. M., Wolf, A.-S., Goodier, M. R. & Riley, E. M. Synergy between common γ chain family cytokines and IL-18 potentiates innate and adaptive pathways of NK cell activation. Front. Immunol. 7, 101 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Rautela, J. & Huntington, N. D. IL-15 signaling in NK cell cancer immunotherapy. Curr. Opin. Immunol. 44, 1–6 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Ferrari de Andrade, L. et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell-driven tumor immunity. Science 359, 1537–1542 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Izawa, S. et al. H2O2 production within tumor microenvironment inversely correlated with infiltration of CD56(dim) NK cells in gastric and esophageal cancer: possible mechanisms of NK cell dysfunction. Cancer Immunol. Immunother. 60, 1801–1810 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Jin, S. et al. NK cell phenotypic modulation in lung cancer environment. PLoS ONE 9, e109976 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Sconocchia, G. et al. NK cells and T cells cooperate during the clinical course of colorectal cancer. Oncoimmunology 3, e952197 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Stankovic, B. et al. Immune cell composition in human non-small cell lung cancer. Front. Immunol. 9, 3101 (2018).

    Article  CAS  PubMed  Google Scholar 

  55. Bonacina, F. et al. Myeloid apolipoprotein E controls dendritic cell antigen presentation and T cell activation. Nat. Commun. 9, 3083 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Gouna, G. et al. TREM2-dependent lipid droplet biogenesis in phagocytes is required for remyelination. J. Exp. Med. 218, e20210227 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. van Eijk, M. & Aerts, J. M. F. G. The unique phenotype of lipid-laden macrophages. Int. J. Mol. Sci. 22, 4039 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Di Conza, G. et al. Tumor-induced reshuffling of lipid composition on the endoplasmic reticulum membrane sustains macrophage survival and pro-tumorigenic activity. Nat. Immunol. 22, 1403–1415 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Nugent, A. A. et al. TREM2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron 105, 837–854 (2020).

    Article  CAS  PubMed  Google Scholar 

  60. Jordan, J. A. et al. Role of IL-18 in acute lung inflammation. J. Immunol. 167, 7060–7068 (2001).

    Article  CAS  PubMed  Google Scholar 

  61. Pechkovsky, D. V., Goldmann, T., Vollmer, E., Müller-Quernheim, J. & Zissel, G. Interleukin-18 expression by alveolar epithelial cells type II in tuberculosis and sarcoidosis. FEMS Immunol. Med. Microbiol. 46, 30–38 (2006).

    Article  CAS  PubMed  Google Scholar 

  62. Barry, K. C. et al. A natural killer–dendritic cell axis defines checkpoint therapy–responsive tumor microenvironments. Nat. Med. 24, 1178–1191 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Böttcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Mattiuz, R. et al. Type 1 conventional dendritic cells and interferons are required for spontaneous CD4+ and CD8+ T-cell protective responses to breast cancer. Clin. Transl. Immunol. 10, e1305 (2021).

    Article  CAS  Google Scholar 

  65. Bonnardel, J. et al. Stellate cells, hepatocytes, and endothelial cells imprint the Kupffer cell identity on monocytes colonizing the liver macrophage niche. Immunity 51, 638–654 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Browaeys, R., Saelens, W. & Saeys, Y. NicheNet: modeling intercellular communication by linking ligands to target genes. Nat. Methods 17, 159–162 (2019).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Merad and Brown laboratories at the Marc and Jennifer Lipschultz Precision Immunology Institute at Mount Sinai and the Tisch Cancer Institute for insightful discussions and feedback; we specifically thank M.J. Lin and J. Brody for lending us flow cytometry antibodies against the NKG2D ligands; the Mount Sinai Flow Cytometry Core, the Human Immune Monitoring Center and the Mount Sinai Biorepository for support. Data in this paper were used in a dissertation as partial fulfillment of the requirements for a PhD at the Graduate School of Biomedical Sciences at Mount Sinai.

Author information

Authors and Affiliations

Authors

Contributions

M.M. conceived the project. M.M., M.D.P. and I.R.T. wrote the manuscript. I.R.T., M.D.P. and M.M. designed the experiments. I.R.T., M.D.P., J.L.B., N.M.L., E.H., S.H., M.B., A.N. and A.R.S. performed experiments. I.R.T., M.D.P., J.L.B. and A.N. maintained mouse colonies and cell cultures for these experiments. M.D.P. and A.M. performed computational analyses, with additional support from T.D., D.D. and S.H. P.H., L.T. and J.G. provided immunohistochemistry stains and analyses. M.M., J.H. and M.C. provided the TREM2 blocking antibody and isotype control. J.N., A.F. and N.B. provided the IL-15 neutralizing antibody; M.J.L. and J.B. provided the antibodies against NKG2D ligands; and L.F.A. provided the MIC-A stabilizing antibody and B16-F10-MICA cell line. A.R.S., B.M., J.C.M., E.K., A.O.K., M.C.A., N.B., A.H., L.F.A., B.D.B., M.C. and T.U.M. provided intellectual input. This work was supported by National Institutes of Health grants R01 AI153363 (to A.O.K.), R01 CA254104 and R01 CA257195 (to M.M. and B.D.B.), 1R37 CA269982-01A1 (to L.F.d.A.), R01CA262684 to M.C. S.H. was supported by the National Cancer Institute predoctoral-to-postdoctoral fellowship K00 CA223043. N.M.L. was supported by the Cancer Research Institute/Bristol Myers Squibb Irvington Postdoctoral Research Fellowship to Promote Racial Diversity (award no. CRI3931). B.D.B. and M.M. were supported by the Applebaum Foundation and the Feldman Foundation. B.D.B. was supported by the Alliance for Cancer Gene Therapy. L.F.d.A. is the recipient of a Cancer Research Institute Clinic and Laboratory Integration Program Grant (award no. CRI3483), Tisch Cancer Institute Developments Funds Award (P30CA196521), Department of Defense Career Development Award (W81XWH2210262 and project number CA210940), Leukemia and Lymphoma Society (award no. 6647-23) and is supported by the Elsa U. Pardee Foundation.

Corresponding author

Correspondence to Miriam Merad.

Ethics declarations

Competing interests

L.F.d.A. is a named inventor of the MIC-A therapeutic antibody in a filed patient (US20200165343A1) and has received royalty payments in relation to this therapeutic. L.F.d.A. is also co-inventor in issued patents about an α-3 domain-specific antibody. M.C. receives research support from NGM Biotechnology and Vigil Neuro, is a scientific advisory board member of NGM Biotechnology and Vigil Neuro and has a patent for TREM2 pending. M.M. serves on the scientific advisory board and holds stock from Compugen, Myeloid Therapeutics, Morphic Therapeutic, Asher Bio, Dren Bio, Nirogy, Oncoresponse, Owkin, Pionyr, OSE and Larkspur. M.M. serves on the scientific advisory board Innate Pharma, DBV and Genenta. M.M. is a named co-inventor on an issued patent for multiplex immunohistochemistry to characterize tumors and treatment responses. The technology is filed through Icahn School of Medicine at Mount Sinai and is currently unlicensed. This technology was used to evaluate tissue in this study and the results could impact the value of this technology. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Florian Greten and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: S. Houston in collaboration with 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 Purification and active signaling pathways in GFPlo and GFPhi mo-macs.

(a) Flow cytometry sort strategy (left) for purifying GFPhi and GFPlo mo-macs from single-cell suspensions of digested KP-GFP tumor-bearing lungs at four weeks post-inoculation of KP-GFP cells. Histograms show the fluorescence spectrum of GFP in mo-macs from tumor-bearing lungs, comprised of GFPhi (green) and GFPlo (blue) mo-macs. Each row represents an individual replicate. Representative confocal immunofluorescence (IF) imaging of FACS-sorted GFPlo and GFPhi mo-macs (right). For each pair of images, a broad-field view with a region of interest (ROI, white outline) (left) and a magnification of the ROI (right) are shown. Data is representative of two independent experiments. (b) Gene set enrichment analysis (GSEA) (Broad Institute) for pathways annotated in the KEGG and Reactome databases performed on the significantly differentially regulated genes in GFPhi mo-macs, relative to GFPlo mo-macs. The top 15 most significantly enriched terms are shown, with notable pathways highlighted in red for up-regulated genes in GFPhi mo-macs (left) and up-regulated genes in GFPlo mo-macs (right).

Extended Data Fig. 2 scRNA-seq profiling of mo-macs in chimeric mice.

(a) Expression of Trem2 and Gpnmb, as hallmark genes of the TREM2 gene program, across cell subtypes (mo-macs, AMs, Ly6Clo and Ly6Chi monocytes, mregDCs, cDC1 and cDC2) identified by unsupervised clustering. (b) Flow cytometry sort strategy to purify CD45.1 WT and CD45.2 KO fractions of MNPs from KP-GFP tumor-bearing chimeric mice. (c) Flow cytometric quantification of GFP+ mo-macs, represented as relative frequency among total mo-macs from the purified CD45.1 WT and CD45.2 KO fractions of MNPs from KP-GFP tumor-bearing mice. Paired values shown for paired CD45.1 WT and CD45.2 KO cells that were purified from the same biological replicate. (mean ± standard error of mean (S.E.M.); paired two-tailed t-test at a 95% confidence interval).

Source data

Extended Data Fig. 3 Contributions of CD8 T cells and NK cells to tumor regression in KO mice.

(a) Flow cytometric quantification of IFN-ɣ/TNF-α-producing CD8 T cells, as a relative frequency of total antigen-specific CD8 T cells, in the tumor-bearing lungs (left) and in the tumor-draining lymph nodes (tdLN) (right) of KP-GFP tumor-bearing WT and KO mice. (n = 4-5 mice per group, mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval). (b) Flow cytometric quantification of NK cells expressing activation/degranulation markers, as a relative frequency of total NK cells. (n = 4-5 mice per group, mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval). (c) Flow cytometric quantification of type 1 conventional dendritic cells (cDC1), as a relative frequency of CD45+ immune cells, in the tumor-bearing lungs of WT mice and KO mice treated with either an isotype control or CD8 (right) (n = 4-5 mice per group) or NK cell (left)-depleting antibody (n = 8-10 mice per group). (mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval. Data is representative of two independent experiments.). (d) Quantification of the tumor area as a percent of the total area of the lung cross-section from tumor-bearing lungs of WT and KO mice treated with either an isotype control of NK cell-depleting antibody. (n = 8-10 mice per group, mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval. Data shown is of at least two independent experiments.). (e) Flow cytometric quantification of NK cells, as a relative frequency of CD45+ immune cells. (n = 8-10 mice per group, mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval. Data is representative of at least three independent experiments.). (f) Flow cytometric quantification of IFN-ɣ-producing (left) and TNF-α-producing (right) CD8 T cells in the tumor-bearing lungs of WT mice with isotype antibody, KO mice with isotype antibody, and KO mice given IL-18 neutralizing antibody. Data shown as a relative frequency among total CD8 T cells. (n = 5-8 mice per group, mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval. Data shown is representative of at least three independent experiments).

Source data

Extended Data Fig. 4 Cooperative IL-15 signaling is needed to confer the therapeutic benefit of a TREM2-deficient setting.

(a) Relative expression of Il15 by mature mregDCs, as determined by scRNA-seq profiling of MNPs in the tumor-bearing lungs of WT and KO mice (Fig. 3C). (b) Representative H&E images of tumor-bearing lungs of WT, isotype antibody (n = 4), WT mice that received the IL-15 neutralizing antibody (n = 4), KO, isotype antibody (n = 4), KO mice that received the IL-15 neutralizing antibody (n = 5) (left). Quantification of the tumor area as a percent of the total area of the lung cross-section (right) is shown. (mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval). (c) Flow cytometric quantification of NK cells, as a relative frequency of CD45+ immune cells, in the KP-GFP tumor-bearing lungs of isotype control WT and KO mice and WT and KO mice treated with the αIL-15 antibody. (n = 4-5 mice per group, mean ± standard error of mean (S.E.M.); unpaired two-tailed t-test at a 95% confidence interval. Data shown is representative of two independent experiments).

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Table 1. Differentially expressed genes between tissue-resident alveolar macrophages and monocyte-derived macrophages in NSCLC. Supplementary Table 2. Gene expression across dendritic cell, monocyte, macrophage and lymphoid cell subsets in NSCLC. Supplementary Table 3. Differentially expressed genes between CD45.1 versus CD45.2 monocyte-derived macrophages identified by scRNA-seq of chimeric mice (day 28 after inoculation of tumor cells).

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 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.

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

Park, M.D., Reyes-Torres, I., LeBerichel, J. et al. TREM2 macrophages drive NK cell paucity and dysfunction in lung cancer. Nat Immunol 24, 792–801 (2023). https://doi.org/10.1038/s41590-023-01475-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01475-4

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing