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:

IRF4 expression by NK precursors predetermines exhaustion of NK cells during tumor metastasis

Abstract

Natural killer (NK) cells are essential for controlling tumor metastasis, but their protective capacity diminishes when entering an exhaustion state. The mechanisms underlying NK cell exhaustion are incompletely understood. Here, we show that the susceptibility of NK cells to exhaustion is predetermined early during their development and is governed by the transcription factor IRF4. Notably, IRF4 is highly expressed in CD27CD24+ NK precursors but is nearly absent in immature and mature NK cells. Deletion of IRF4 redirects NK cell development, enabling NK precursors to generate more mature NK cells that resist exhaustion, thereby decreasing melanoma lung metastasis. This resistance to exhaustion is evident by increased effector molecule production and decreased expression of inhibitory receptors such as TIGIT and Pik3ip1. Deleting Pik3ip1 also enhances NK cell ability to counteract melanoma lung metastasis. These findings enhance our understanding of NK cell exhaustion and have implications for preventing cancer metastasis using NK cells.

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

Access options

Buy this article

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

Fig. 1: IRF4 deficiency reduces melanoma metastasis via NK cells.
Fig. 2: IRF4 expression in NKP cells and its effect on mature NK cell frequency.
Fig. 3: Single-cell transcriptomics identifies CD24+CD27 as markers of IRF4-expressing NKP cells.
Fig. 4: IRF4 expression in CD24+CD27 NKP cells restrains NK cell development.
Fig. 5: Irf4 deficiency during NK cell development renders mature NK cells less prone to exhaustion.
Fig. 6: Irf4 deficiency alters chromatin accessibility in naive NK cells, shaping terminal differentiation and exhaustion.
Fig. 7: Irf4 deficiency prevents expression of inhibitory receptor Pik3ip1 by NK cells.

Similar content being viewed by others

Data availability

RNA-seq, scRNA-seq and ATAC-seq data have been deposited in the Gene Expression Omnibus under accession codes GSE249188, GSE249230 and GSE283690. Raw Illumina sequencing reads of RNA-seq, scRNA-seq and ATAC-seq datasets were aligned to reference mouse genome mm10 (Ensembl 93), http://jul2018.archive.ensembl.org/Mus_musculus/Info/Index. All other data supporting the findings of this study are available within the article and Supplementary Information. Source data are provided with this paper.

Code availability

ScRNA-seq and ATAC-seq data analyses were conducted using publicly available codes and open-source software packages. No new algorithms were developed for this study.

References

  1. Lopez-Soto, A., Gonzalez, S., Smyth, M. J. & Galluzzi, L. Control of metastasis by NK cells. Cancer Cell 32, 135–154 (2017).

    CAS  PubMed  Google Scholar 

  2. Liu, S. et al. NK cell-based cancer immunotherapy: from basic biology to clinical development. J. Hematol. Oncol. 14, 7 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Wu, S. Y., Fu, T., Jiang, Y. Z. & Shao, Z. M. Natural killer cells in cancer biology and therapy. Mol. Cancer 19, 120 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Menard, C. et al. Natural killer cell IFN-gamma levels predict long-term survival with imatinib mesylate therapy in gastrointestinal stromal tumor-bearing patients. Cancer Res. 69, 3563–3569 (2009).

    CAS  PubMed  Google Scholar 

  5. Smyth, M. J. et al. Perforin is a major contributor to NK cell control of tumor metastasis. J. Immunol. 162, 6658–6662 (1999).

    CAS  PubMed  Google Scholar 

  6. Street, S. E., Cretney, E. & Smyth, M. J. Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis. Blood 97, 192–197 (2001).

    CAS  PubMed  Google Scholar 

  7. Seaman, W. E., Sleisenger, M., Eriksson, E. & Koo, G. C. Depletion of natural killer cells in mice by monoclonal antibody to NK-1.1. Reduction in host defense against malignancy without loss of cellular or humoral immunity. J. Immunol. 138, 4539–4544 (1987).

    CAS  PubMed  Google Scholar 

  8. Liu, Y. et al. Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive cancers. Oncogene 36, 6143–6153 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhang, Q. et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 19, 723–732 (2018).

    CAS  PubMed  Google Scholar 

  10. Judge, S. J., Murphy, W. J. & Canter, R. J. Characterizing the dysfunctional NK cell: assessing the clinical relevance of exhaustion, anergy, and senescence. Front. Cell Infect. Microbiol. 10, 49 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Bi, J. & Tian, Z. NK cell exhaustion. Front. Immunol. 8, 760 (2017).

    PubMed  PubMed Central  Google Scholar 

  12. Ardolino, M. et al. Cytokine therapy reverses NK cell anergy in MHC-deficient tumors. J. Clin. Invest. 124, 4781–4794 (2014).

    PubMed  PubMed Central  Google Scholar 

  13. da Silva, I. P. et al. Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol. Res 2, 410–422 (2014).

    PubMed  PubMed Central  Google Scholar 

  14. Komita, H. et al. Expression of immune checkpoint molecules of T cell immunoglobulin and mucin protein 3/galectin-9 for NK cell suppression in human gastrointestinal stromal tumors. Oncol. Rep. 34, 2099–2105 (2015).

    CAS  PubMed  Google Scholar 

  15. Benson, D. M. Jr. et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood 116, 2286–2294 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Kondo, M., Weissman, I. L. & Akashi, K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91, 661–672 (1997).

    CAS  PubMed  Google Scholar 

  17. Abel, A. M., Yang, C., Thakar, M. S. & Malarkannan, S. Natural killer cells: development, maturation, and clinical utilization. Front Immunol. 9, 1869 (2018).

    PubMed  PubMed Central  Google Scholar 

  18. Chiossone, L. et al. Maturation of mouse NK cells is a 4-stage developmental program. Blood 113, 5488–5496 (2009).

    CAS  PubMed  Google Scholar 

  19. Fu, B. et al. CD11b and CD27 reflect distinct population and functional specialization in human natural killer cells. Immunology 133, 350–359 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Tamura, T., Yanai, H., Savitsky, D. & Taniguchi, T. The IRF family transcription factors in immunity and oncogenesis. Annu. Rev. Immunol. 26, 535–584 (2008).

    CAS  PubMed  Google Scholar 

  21. Ohteki, T. et al. The transcription factor interferon regulatory factor 1 (IRF-1) is important during the maturation of natural killer 1.1+ T cell receptor-alpha/beta+ (NK1+ T) cells, natural killer cells, and intestinal intraepithelial T cells. J. Exp. Med. 187, 967–972 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lohoff, M. et al. Deficiency in the transcription factor interferon regulatory factor (IRF)-2 leads to severely compromised development of natural killer and T helper type 1 cells. J. Exp. Med. 192, 325–336 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Taki, S., Nakajima, S., Ichikawa, E., Saito, T. & Hida, S. IFN regulatory factor-2 deficiency revealed a novel checkpoint critical for the generation of peripheral NK cells. J. Immunol. 174, 6005–6012 (2005).

    CAS  PubMed  Google Scholar 

  24. Adams, N. M. et al. Transcription factor IRF8 orchestrates the adaptive natural killer cell response. Immunity 48, 1172–1182.e1176 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Santosa, E. K. et al. Control of nutrient uptake by IRF4 orchestrates innate immune memory. Nat. Immunol. 24, 1685–1697 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Geary, C. D. et al. Non-redundant ISGF3 components promote NK cell survival in an auto-regulatory manner during viral infection. Cell Rep. 24, 1949–1957.e1946 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Yan, H. et al. The transcription factor IRF4 determines the anti-tumor immunity of CD8+ T cells. iScience 26, 108087 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Yu, A. et al. Continuous expression of interferon regulatory factor 4 sustains CD8+ T cell immunity against tumor. Research (Wash. DC) 6, 0271 (2023).

    CAS  Google Scholar 

  29. Wang, G. et al. IRF4 ablation in B cells abrogates allogeneic B cell responses and prevents chronic transplant rejection. J. Heart Lung Transpl. 40, 1122–1132 (2021).

    Google Scholar 

  30. Rosmaraki, E. E. et al. Identification of committed NK cell progenitors in adult murine bone marrow. Eur. J. Immunol. 31, 1900–1909 (2001).

    CAS  PubMed  Google Scholar 

  31. Fathman, J. W. et al. Identification of the earliest natural killer cell-committed progenitor in murine bone marrow. Blood 118, 5439–5447 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Mittrucker, H. W. et al. Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science 275, 540–543 (1997).

    CAS  PubMed  Google Scholar 

  33. Huntington, N. D. et al. NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation. J. Immunol. 178, 4764–4770 (2007).

    CAS  PubMed  Google Scholar 

  34. Malaise, M. et al. KLRG1+ NK cells protect T-bet-deficient mice from pulmonary metastatic colorectal carcinoma. J. Immunol. 192, 1954–1961 (2014).

    CAS  PubMed  Google Scholar 

  35. Alvarez, M. et al. Regulation of murine NK cell exhaustion through the activation of the DNA damage repair pathway. JCI Insight 5, e127729 (2019).

    PubMed  Google Scholar 

  36. Uche, U. U. et al. PIK3IP1/TrIP restricts activation of T cells through inhibition of PI3K/Akt. J. Exp. Med. 215, 3165–3179 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hsu, J. et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Invest. 128, 4654–4668 (2018).

    PubMed  PubMed Central  Google Scholar 

  38. Chauvin, J. M. et al. IL15 Stimulation with TIGIT blockade reverses CD155-mediated NK-cell dysfunction in melanoma. Clin. Cancer Res. 26, 5520–5533 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. He, Y. et al. Contribution of inhibitory receptor TIGIT to NK cell education. J. Autoimmun. 81, 1–12 (2017).

    CAS  PubMed  Google Scholar 

  40. Townsend, M. J. et al. T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells. Immunity 20, 477–494 (2004).

    CAS  PubMed  Google Scholar 

  41. Gordon, S. M. et al. The transcription factors T-bet and Eomes control key checkpoints of natural killer cell maturation. Immunity 36, 55–67 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Bauer, S. et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285, 727–729 (1999).

    CAS  PubMed  Google Scholar 

  43. Sciume, G. et al. CX3CR1 expression defines 2 KLRG1+ mouse NK-cell subsets with distinct functional properties and positioning in the bone marrow. Blood 117, 4467–4475 (2011).

    CAS  PubMed  Google Scholar 

  44. Wijaya, R. S. et al. KLRG1+ natural killer cells exert a novel antifibrotic function in chronic hepatitis B. J. Hepatol. 71, 252–264 (2019).

    CAS  PubMed  Google Scholar 

  45. Chen, Y. et al. Pik3ip1 is a negative immune regulator that inhibits antitumor T-cell immunity. Clin. Cancer Res. 25, 6180–6194 (2019).

    CAS  PubMed  Google Scholar 

  46. Narni-Mancinelli, E. et al. Fate mapping analysis of lymphoid cells expressing the NKp46 cell surface receptor. Proc. Natl Acad. Sci. USA 108, 18324–18329 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Wu, J. et al. Ablation of transcription factor IRF4 promotes transplant acceptance by driving allogenic CD4+ T cell dysfunction. Immunity 47, 1114–1128 e1116 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Wang, G. et al. The RNA helicase DHX15 is a critical regulator of natural killer-cell homeostasis and functions. Cell Mol. Immunol. 19, 687–701 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Zou, D. et al. T cell exhaustion is associated with antigen abundance and promotes transplant acceptance. Am. J. Transpl. 20, 2540–2550 (2020).

    CAS  Google Scholar 

  50. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 e1821 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Vivier (Aix Marseille University) for the Ncr1Cre mouse strain and L. L. Lanier (University of California San Francisco) for the RMA-S cell line. We would like to thank the Single Cell Genomics Core at Baylor College of Medicine, the Biostatistics and Bioinformatics (BBI) shared resources at Houston Methodist Neal Cancer Center and the Houston Methodist Flow Cytometry Core Facility for their excellent services. This work was supported by internal fund from Houston Methodist Research Institute (to W.C.).

Author information

Authors and Affiliations

Authors

Contributions

X.Z., J.W., X.X. and W.C. designed the study. X.Z., X.C.L. and W.C. wrote the manuscript. X.Z. performed core experimental work and data analysis. Z.Y. performed computational analyses. J.W., X.X., D.Z., G.W., J.F., P.L., L.J.M., X.C.L. and W.C. performed supporting experimental work and data analysis.

Corresponding author

Correspondence to Wenhao Chen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Dagmar Gotthardt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Nick Bernard, in collaboration with the Nature Immunology team. Peer reviewer reports are available.

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 IRF4 deletion in T cells, dendritic cells, or macrophages does not reduce melanoma metastasis.

(a) Representative image of lung metastasis in Irf4fl/fl and Irf4fl/flCd4-Cre mice 20 days post B16-F10 cell intravenous injection, and a graph showing percentage survival rates for the injected mice. n = 6 mice per group; ns, not significant. (b) Representative lung metastasis image (day 20 post B16-F10 cell injection) in Irf4fl/fl and Irf4fl/flLysM-Cre mice, and percentage survival rates. n = 7 mice per group; ns, not significant. (c) Percentage survival rates of Irf4fl/flCd11c-Cre and Irf4fl/fl mice post B16-F10 cell intravenous injection. n = 5 mice per group; ns, not significant. (d) Percentages of remaining CD3NK1.1+ cells within splenocytes of WT and Irf4–/– mice three days post anti-NK1.1 mAb (clone PK136) treatment. Results are representative of two or three independent experiments. Data were analyzed by a log-rank test.

Source data

Extended Data Fig. 2 Irf4‒/‒ NK cells exhibit a more mature phenotype.

(a) Representative flow cytometry plots of CD3NK1.1+ cells and bar graphs showing NK cell numbers in the liver and blood (per ml) of naïve WT and Irf4−/− mice. For the liver, n = 6 mice per group, **P = 0.0022; for the blood, n = 5 mice per group, ***P = 0.0001. (b) Representative flow cytometry plots show CD27 and CD11b expression in NK cells. The bar graph shows the percentage of CD27CD11b+ cells among total NK cells in the indicated tissues of naïve WT and Irf4−/− mice. n = 6 mice per group; from left to right: ****P < 0.0001, ***P = 0.0009, ***P = 0.0010, * P = 0.0128. (c) Sorted NK cells from naïve WT and Irf4−/− mice were labeled with CTV and stimulated in vitro with IL-12 (10 ng/ml), IL-18 (50 ng/ml), and IL-15 (50 ng/ml) for 3 days to assess proliferation. (d) Sorted NK cells from naïve WT and Irf4−/− mice were stimulated in vitro with IL-12, IL-18, and IL-15 for 3 days, followed by apoptosis assessment. n = 5 biologically independent samples per group; ****P < 0.0001. Data are presented as mean ± SD (a,b,d). Results are representative of two or three independent experiments. Data were analyzed by a two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 3 Irf4–/– BM cell reconstitution does not enhance the generation of other immune cells.

(a) Flow cytometry analysis confirming that WT and Irf4–/– BM cells were mixed at a 1:1 ratio prior to transfer into lethally irradiated Rag1–/– mice. (b) Flow cytometry analysis of the percentage of NKP cells in the bone marrow of WT and Irf4–/– mice. n = 3 mice per group; ns, not significant. (c and d) Lethally irradiated Rag1−/− mice were reconstituted with a 1:1 mixture of CD45.1+ WT and CD45.2+ Irf4–/– BM cells. Flow cytometry was performed 6 weeks post transfer to assess the percentage of live cells (c) and CD3+NK1.1 T cells (d) derived from transferred WT or Irf4–/– BM cells. n = 3 mice; for c: ***P = 0.0003, ns, not significant; for d: **P = 0.0052, ns, not significant. Results are representative of two or three independent experiments. Data are presented as mean ± SD (b,c,d). Data were analyzed by a two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 4 IRF4 expression in NKP and NK cells during melanoma metastasis.

(a and b) Irf4GFP reporter mice were intravenously injected with B16-F10 cells. Shown are the percentages of IRF4.GFP+ cells within NK cells in spleen and lung (a) and the percentages of IRF4.GFP+CD27 cells within BM NKP cells (b) at different time points post B16-F10 injection. n = 3 mice per time point. (c and d) Rag2−/−γc−/− mice were transferred with a 1:1 mixture of WT and Irf4−/− NKP cells. (c) Gating strategy used to sort BM NKP cells for transfer. (d) Percentages of NK cells derived from the transferred WT or Irf4−/− NKP cells at 6 weeks post-transfer. n = 4 mice; **P = 0.0021. Data are presented as mean ± SD (a,b,d). Data were analyzed by a two-tailed unpaired Student’s t-test (d).

Source data

Extended Data Fig. 5 Identification of IRF4-expressing CD24+CD27 NKP cells.

(a and b) scRNA-seq analysis of a total of 20,836 cells, including 12,740 WT NKP and 8,096 Irf4−/− NKP cells. UMAP plots (a) illustrating the normalized expression of Irf4 and Cd24a in NKP cells. Bubble plots (b) illustrating the normalized expression of Irf4 and Cd24a across identified NKP cell clusters (matching cluster IDs in Fig. 3a). The color intensity corresponds to the average expression level within the cluster. The size of each bubble indicates the percentage of cells expressing the gene within the cluster. (ce) CD24CD27+ NKP cells from WT CD45.1+ BM and CD24+CD27 NKP cells from WT CD45.2+ BM were adoptively co-transferred into Rag2−/−γc−/− mice in a 1:1 ratio, followed by flow cytometry analysis six weeks later. Shown are percentages of CD45.1+ and CD45.2+ cells among CD3NK1.1+ splenocytes (c, n = 3 mice, **P = 0.0036), expression of CD27 versus CD11b among CD3NK1.1+ splenocytes derived from the transferred NKP cells (d), and expression of CD27 in CD3NK1.1 splenocytes derived from the transferred NKP cells (e). (f) CD27CD24med NKP cells from WT CD45.1+ BM and CD27CD24high NKP cells from WT CD45.2+ BM were adoptively co-transferred into Rag2−/−γc−/− mice in a 1:1 ratio. The percentages of splenic CD3NK1.1+ NK cells derived from these transferred NKP cells were assessed two weeks post-transfer (n = 3 mice; ****P < 0.0001). Data are presented as mean ± SD (c,f). Data were analyzed by a two-tailed unpaired Student’s t-test (c,f).

Source data

Extended Data Fig. 6 Evaluation of NK cells from WT and Irf4‒/‒ mice.

(a,b) Splenic NK cells from naïve WT and Irf4‒/‒ mice were stimulated with IL-12, IL-15, and IL-18. Shown are the percentages of IFN-γ+ and perforin+ cells (a), as well as granzyme B+ cells (b), within WT and Irf4‒/‒ NK cells. For IFN-γ, n = 5 mice per group; for perforin, n = 4 mice per group; for granzyme B, n = 3 mice per group; ns, not significant. (c) Splenic NK cells from naïve WT and Irf4‒/‒ mice were cocultured with RMA/S-GFP target cells at the indicated ratios. Percentage killing of target cells were illustrated. n = 3 biologically independent samples per group. (d) Percentages of granzyme B+ cells within lung NK cells, comparing WT versus Irf4–/– mice 20 days post B16-F10 cell intravenous injection. n = 3 mice per group; **P = 0.0038. (e) Expression of TIGIT in NK cells from the spleens of naïve WT and Irf4−/− mice. n = 4 mice per group; ns, not significant. (f) NK cells sorted from the spleens of naïve WT and Irf4−/− mice were stimulated in vitro with plate-bound anti-NK1.1 (PK136) antibody (20 µg/ml), IL-2 (10 ng/ml), and IL-15 (5 ng/ml) for 3 days, followed by assessment of TIGIT expression. n = 6 biologically independent samples per group; ****P < 0.0001. Data are presented as mean ± SD. Data were analyzed by a two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 7 Deleting IRF4 in Ncr1-expressing NK cells does not prolong survival in the melanoma metastasis model.

(a) Irf4fl/fl mice from Jackson Laboratory contain an eGFP gene positioned in the opposite orientation upstream of the Irf4 promoter. Upon Cre-mediated Irf4 deletion, eGFP is expressed, although at low levels in some cells. Shown is GFP expression in NK cells from the spleens of naïve Irf4fl/fl and Irf4fl/flNcr1Cre+/‒ mice. (b) Expression of Nkp46 in NK cells from the spleens of naïve Irf4fl/fl and Irf4fl/flNcr1Cre+/‒ mice. (c) Percentages of Nkp46+ cells among CD3NK1.1+ splenocytes in naïve Irf4fl/fl and Irf4fl/flNcr1Cre+/‒ mice. n = 3 mice per group; ns, not significant. (d) Percentage survival rates of Irf4fl/fl, Irf4w/wNcr1Cre+/‒, and Irf4fl/flNcr1Cre+/‒ mice following intravenous injection of B16-F10 cells. n = 5 mice per group; ns, not significant. Data are presented as mean ± SD (c). Data were analyzed by a two-tailed unpaired Student’s t-test (c) and a log-rank test (d).

Source data

Extended Data Fig. 8 Irf4 deficiency in NKP cells renders mature NK cells less prone to exhaustion.

(a and b) Rag2−/−γc−/− mice were transferred with a 1:1 mixture of CD45.1+ WT NKP and CD45.2+ Irf4−/− NKP cells. Two weeks later, the mice were intravenously injected with B16-F10 cells. Splenic NK cells derived from the transferred NKP cells were analyzed 14 days after B16-F10 injection. Shown are the percentages of WT and Irf4–/– NK cells among total splenic NK cells (a) and the percentages of TIGIT+ cells within WT and Irf4–/– NK cells (b). For a: n = 3 mice, ***P = 0.0002; for b: n = 4 mice, ***P = 0.0008. (c) Lethally irradiated Rag1−/− mice were reconstituted with a 1:1 mixture of CD45.1+ WT BM and CD45.2+ Irf4–/– BM cells. Six weeks later, the mice were injected with B16-F10 cells. NK cells derived from the transferred BM NKP cells were analyzed 25 days after B16-F10 injection. The top flow plots show the percentages of WT and Irf4–/– NK cells, and the bottom flow plots show TIGIT expression in WT and Irf4–/– NK cells in the indicated tissues. The bar graph shows the percentages of TIGIT+ cells within WT and Irf4–/– NK cells. n = 6 mice; from left to right: ****P < 0.0001 ****P < 0.0001, *P = 0.0162, ****P < 0.0001, ****P < 0.0001. Data are presented as mean ± SD. Data were analyzed by a two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 9 Chromatin accessibility at indicated loci in NK cells.

ATAC-seq was performed on NK cells sorted from naïve WT, Irf4–/–, Irf4fl/fl, and Irf4fl/flNcr1Cre+/‒ mice (related to Fig. 6). Genome tracks show chromatin accessibility at the Prf1, Ifng, Tigit and Gzmb loci across the indicated NK cell groups.

Extended Data Fig. 10 Pik3ip1 deficiency reduces NK cell exhaustion during melanoma metastasis.

(a) Percentages of NK cells in the spleen of naïve WT and Pik3ip1‒/‒ mice. n = 5 mice per group; ns, not significant. (b) Percentages of TIGIT+ cells within spleen and lung NK cells, comparing WT vs. Pik3ip1‒/‒ mice 20 days post B16-F10 cell injection. n = 3 mice per group; *P = 0.0169, **P = 0.0050. (c) Percentages of IFN-γ+ and perforin+ cells within lung NK cells, comparing WT vs. Pik3ip1‒/‒ mice 20 days post B16-F10 injection. n = 3 mice per group; **P = 0.0023, *P = 0.0155. Data are presented as mean ± SD. Results are representative of two or three independent experiments. Data were analyzed by a two-tailed unpaired Student’s t-test.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Cluster proportion source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, X., Yin, Z., Wu, J. et al. IRF4 expression by NK precursors predetermines exhaustion of NK cells during tumor metastasis. Nat Immunol 26, 1062–1073 (2025). https://doi.org/10.1038/s41590-025-02176-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-025-02176-w

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