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:

ILC1s control leukemia stem cell fate and limit development of AML

A Publisher Correction to this article was published on 15 June 2022

This article has been updated

Abstract

Type I innate lymphoid cells (ILC1s) are critical regulators of inflammation and immunity in mammalian tissues. However, their function in cancer is mostly undefined. Here, we show that a high density of ILC1s induces leukemia stem cell (LSC) apoptosis. At a lower density, ILC1s prevent LSCs from differentiating into leukemia progenitors and promote their differentiation into non-leukemic cells, thus blocking the production of terminal myeloid blasts. All of these effects, which require ILC1s to produce interferon-γ after cell–cell contact with LSCs, converge to suppress leukemogenesis in vivo. Conversely, the antileukemia potential of ILC1s wanes when JAK–STAT or PI3K–AKT signaling is inhibited. The relevant antileukemic properties of ILC1s are also functional in healthy individuals and impaired in individuals with acute myeloid leukemia (AML). Collectively, these findings identify ILC1s as anticancer immune cells that might be suitable for AML immunotherapy and provide a potential strategy to treat AML and prevent relapse of the disease.

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: ILC1s induce apoptosis in LSCs.
Fig. 2: ILC1-secreted IFNγ controls the direction of LSC differentiation.
Fig. 3: ILC1s inhibit the differentiation of LSCs into myeloid blasts.
Fig. 4: ILC1s and IFNγ improve survival of leukemic mice.
Fig. 5: ILC1s produce more IFNγ than NK cells when interacting with LSCs.
Fig. 6: ILC1-derived IFNγ inhibits LSC differentiation via JAK–STAT and PI3K–AKT signaling.
Fig. 7: ILC1s are functionally impaired in AML.

Similar content being viewed by others

Data availability

Sequencing datasets are accessible from the Gene Expression Omnibus (GEO) under accession number GSE198783. Source data are provided with this paper.

Change history

References

  1. Nair, R., Salinas-Illarena, A. & Baldauf, H. M. New strategies to treat AML: novel insights into AML survival pathways and combination therapies. Leukemia 35, 299–311 (2021).

  2. Ballester, G., Tirona, M. T. & Ballester, O. Hematopoietic stem cell transplantation in the elderly. Oncology 21, 1576–1583 (2007).

    PubMed  Google Scholar 

  3. Klose, C. S. N. & Artis, D. Innate lymphoid cells control signaling circuits to regulate tissue-specific immunity. Cell Res. 30, 475–491 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Goc, J. et al. Dysregulation of ILC3s unleashes progression and immunotherapy resistance in colon cancer. Cell 184, 5015–5030 (2021).

  5. Moral, J. A. et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature 579, 130–135 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Nabekura, T., Riggan, L., Hildreth, A. D., O’Sullivan, T. E. & Shibuya, A. Type 1 innate lymphoid cells protect mice from acute liver injury via interferon-γ secretion for upregulating Bcl-xL expression in hepatocytes. Immunity 52, 96–108 (2020).

    Article  CAS  PubMed  Google Scholar 

  7. Klose, C. S. N. et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 157, 340–356 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Klose, C. S. et al. A T-bet gradient controls the fate and function of CCR6-RORγt+ innate lymphoid cells. Nature 494, 261–265 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Weizman, O. E. et al. ILC1 confer early host protection at initial sites of viral infection. Cell 171, 795–808 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Shannon, J. P. et al. Group 1 innate lymphoid-cell-derived interferon-γ maintains anti-viral vigilance in the mucosal epithelium. Immunity 54, 276–290 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang, B. et al. Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia. Nat. Med. 24, 450–462 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zorko, N. A. et al. Mll partial tandem duplication and Flt3 internal tandem duplication in a double knock-in mouse recapitulates features of counterpart human acute myeloid leukemias. Blood 120, 1130–1136 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Shipounova Nifontova, I. N., Bigil’diev, A. E., Svinareva, D. A. & Drize, N. I. Characteristics of leukemia stem cells of murine myeloproliferative disease involving the liver. Bull. Exp. Biol. Med. 149, 293–297 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. Almishri, W. et al. TNFα augments cytokine-induced NK cell IFNγ production through TNFR2. J. Innate Immun. 8, 617–629 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Artis, D. & Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Appelbaum, F. R., Rowe, J. M., Radich, J. & Dick, J. E. Acute myeloid leukemia. Hematology Am. Soc. Hematol. Educ. Program 2001, 62–86 (2001).

  17. Peng, C. et al. LSK derived LSK cells have a high apoptotic rate related to survival regulation of hematopoietic and leukemic stem cells. PLoS ONE 7, e38614 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kumar, R., Fossati, V., Israel, M. & Snoeck, H. W. Lin-Sca1+Kit bone marrow cells contain early lymphoid-committed precursors that are distinct from common lymphoid progenitors. J. Immunol. 181, 7507–7513 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Joshi, K., Zhang, L., Breslin, S. J. P. & Zhang, J. Leukemia stem cells in the pathogenesis, progression, and treatment of acute myeloid leukemia. Adv. Exp. Med. Biol. 1143, 95–128 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Adane, B. et al. The hematopoietic oxidase NOX2 regulates self-renewal of leukemic stem cells. Cell Rep. 27, 238–254 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Park, S. M. et al. IKZF2 drives leukemia stem cell self-renewal and inhibits myeloid differentiation. Cell Stem Cell 24, 153–165 (2019).

    Article  CAS  PubMed  Google Scholar 

  22. Laouedj, M. et al. S100A9 induces differentiation of acute myeloid leukemia cells through TLR4. Blood 129, 1980–1990 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Filén, J. J. et al. Quantitative proteomics reveals GIMAP family proteins 1 and 4 to be differentially regulated during human T helper cell differentiation. Mol. Cell. Proteom. 8, 32–44 (2009).

    Article  Google Scholar 

  24. Sharma, A. et al. Constitutive IRF8 expression inhibits AML by activation of repressed immune response signaling. Leukemia 29, 157–168 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Bank, U. et al. c-FLIP is crucial for IL-7/IL-15-dependent NKp46+ ILC development and protection from intestinal inflammation in mice. Nat. Commun. 11, 1056 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ma, S. et al. The RNA m6A reader YTHDF2 controls NK cell antitumor and antiviral immunity. J. Exp. Med. 218, e2020279 (2021).

    Article  Google Scholar 

  27. Smith, M. et al. Adult acute myeloid leukaemia. Crit. Rev. Oncol. Hematol. 50, 197–222 (2004).

    Article  PubMed  Google Scholar 

  28. Gao, Y. et al. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat. Immunol. 18, 1004–1015 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Robinette, M. L. et al. Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat. Immunol. 16, 306–317 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Villarino, A. V., Kanno, Y. & O’Shea, J. J. Mechanisms and consequences of JAK–STAT signaling in the immune system. Nat. Immunol. 18, 374–384 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Yamashita, M., Dellorusso, P. V., Olson, O. C. & Passegué, E. Dysregulated haematopoietic stem cell behaviour in myeloid leukaemogenesis. Nat. Rev. Cancer 20, 365–382 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Levis, M. & Perl, A. E. Gilteritinib: potent targeting of FLT3 mutations in AML. Blood Adv. 4, 1178–1191 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Castro, F., Cardoso, A. P., Gonçalves, R. M., Serre, K. & Oliveira, M. J. Interferon-γ at the crossroads of tumor immune surveillance or evasion. Front. Immunol. 9, 847 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Razaghi, A., Owens, L. & Heimann, K. Review of the recombinant human interferon γ as an immunotherapeutic: impacts of production platforms and glycosylation. J. Biotechnol. 240, 48–60 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Srivastava, S. et al. Immunogenic chemotherapy enhances recruitment of CAR-T cells to lung tumors and improves antitumor efficacy when combined with checkpoint blockade. Cancer Cell 39, 193–208 (2020).

  36. Song, M. et al. Low-dose IFNγ induces tumor cell stemness in tumor microenvironment of non-small cell lung cancer. Cancer Res. 79, 3737–3748 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Maloy, K. J. & Uhlig, H. H. ILC1 populations join the border patrol. Immunity 38, 630–632 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Houshmand, M. et al. Bone marrow microenvironment: the guardian of leukemia stem cells. World J. Stem Cells 11, 476–490 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Dunn, G. P., Koebel, C. M. & Schreiber, R. D. Interferons, immunity and cancer immunoediting. Nat. Rev. Immunol. 6, 836–848 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Munneke, J. M. et al. Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood 124, 812–821 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Berrien-Elliott, M. M. et al. Multidimensional analyses of donor memory-like NK cells reveal new associations with response after adoptive immunotherapy for leukemia. Cancer Discov. 10, 1854–1871 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Fehniger, T. A. et al. Fatal leukemia in interleukin 15 transgenic mice follows early expansions in natural killer and memory phenotype CD8+ T cells. J. Exp. Med. 193, 219–231 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen, S., Zhou, Y., Chen, Y. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Institutes of Health (CA210087, CA265095 and CA163205 to M.A.C.; NS106170, AI129582, CA247550, CA264512 and CA223400 to J.Y.) and the Leukemia and Lymphoma Society (1364-19 to J.Y.). Research reported in this publication also included work performed in the Hematopoietic Tissue Biorepository Core supported by the National Cancer Institute of the National Institutes of Health under grant number P30CA033572. We appreciate S.T. Wilkinson and L. Sage for editing the manuscript. Images were created with BioRender.com and Adobe Photoshop.

Author information

Authors and Affiliations

Authors

Contributions

J.Y. and M.A.C. conceived and designed the study. Z.L., R.M., S.M., L.T. and T.L developed the methodology. Z.L., R.M., S.M., L.T., T.L., B.L.M.-B., B.Z. and G.M. acquired the data (for example, provided animals, acquired and managed donors, provided facilities and so on). Z.L., J.Y. and J.Z. analyzed and interpreted data (for example, statistical analysis, biostatistics and computational analysis). Z.L., R.M., S.M., J.Y. and M.A.C. wrote, reviewed and/or revised the manuscript. J.Z. provided administrative, technical or material support (for example, reporting or organizing data and constructing databases). J.Y. and M.A.C. supervised the study and acquired funding. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Michael A. Caligiuri or Jianhua Yu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Eric Vivier and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: N. Bernard and Z. Fehervari, 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 ILC1s are functionally impaired in mice with AML.

a, 0.2×106 LSKs isolated from the liver of healthy mice or MllPTD/WT: Flt3ITD/ITD mice with AML were i.v. injected into immunodeficient Rag2/γc/ mice. The survival of those mice was analyzed by the Kaplan-Meier method and log-rank test (n = 3 individual mice). b, LSCs were isolated from the spleen of MllPTD/WT: Flt3ITD/ITD mice with AML (CD45.2) and then transplanted into lethally irradiated mice (CD45.1). The percentages of LSCs in the liver of the CD45.1 mice were analyzed 9 weeks after LSC transplantation using flow cytometry. c, Gating strategy for flow cytometry analysis of the mouse ILC1s isolated from livers. The mouse ILC1s were defined as CD3CD19NK1.1+NKp46+CD49bCD49a+. d-f, 2×106 C1498 cells were i.v. injected into C57BL/6 J mice. Twenty-one days later, the production of IFN-γ and TNF by ILC1s from the liver (d), bone marrow (e), and spleen (f) of those healthy mice or mice with AML are shown (n = 5 individual mice). g, GSEA plot shows the relative abundance of genes involved in the TNF–NF-κB signaling pathways in liver ILC1s isolated from mice with AML or healthy mice (n = 3 individual mice). Data are representative of two (a, b, d, e, and f) independent experiments. Data (d-f) are shown as mean ± s.d. and are assessed by unpaired two-tailed Student’s t test. NS, not significant.

Source data

Extended Data Fig. 2 Cell purity and gating strategy for flow cytometry analysis.

a,b, Purity of LSCs (a) and ILC1s (b) after cell sorting. c, Gating strategy for flow cytometric analysis of apoptosis of LSCs co-cultured with or without ILC1s, using 7-AAD. CTV: CellTrace™ Violet. d, Gating strategy for flow cytometry analysis of apoptosis of LSCs cocultured with ILC1s using the Violet Live Cell Caspase Probe. e, Gating strategy for flow cytometry analysis of human ILC1s isolated from peripheral blood. Lineage markers: CD3, CD4, CD8, CD14, CD15, CD16, CD19, CD20, CD33, CD34, CD203c, and FceRI. Human ILC1s were defined as LinCD56CD127+c-KitCRTH2. f, Gating strategy for flow cytometry analysis of human LSCs. Lineage markers: CD2, CD3, CD4, CD8, CD14, CD16, CD19, Mac-1, CD56, and CD235a. Human LSCs were defined as LinCD45dimCD34+CD38.

Extended Data Fig. 3 IFN-γ—but not TNF—induces apoptosis of LSCs.

a, 5,000–10,000 mouse liver ILC1s were sorted and transferred into the top wells of a 96-well Transwell plate. The bottom chambers of the plate were loaded with 10,000–20,000 LSCs from the spleens of MllPTD/WT: Flt3ITD/ITD mice with AML. The cells were then cocultured for 3 days. The percentages of LSCs that were apoptotic were measured by flow cytometry (n = 3 individual mice). b,c, LSCs from the spleen of MllPTD/WT: Flt3ITD/ITD mice with AML were treated with or without the indicated doses of IFN-γ or TNF for 3 days. Representative images (top, 5× magnification, scale bar 200 µm) and flow cytometry plots (bottom) of the percentages of apoptotic cells in LSCs are shown. Data in a are shown as mean ± s.d. and are assessed by one-way ANOVA models. Data in a, b, and c are representative of two independent experiments. NS, not significant.

Source data

Extended Data Fig. 4 ILC1s and IFN-γ transform the differentiation of LSCs.

a, LSCs from the spleen of MllPTD/WT: Flt3ITD/ITD mice with AML were cocultured with or without 0.1 ng/ml, 1 ng/ml, or 10 ng/ml recombinant mouse IFN-γ. The percentages of LinSca-1+c-Kit+, LinSca-1c-Kit+, and LinSca-1+c-Kit cells were measured by flow cytometry (n = 4 individual mice). b, ILC1s from healthy mouse liver were sorted and transferred into the top well of a 96-well Transwell plate. The bottom chamber of the plate was loaded with LSCs from the spleen of MllPTD/WT: Flt3ITD/ITD mice with AML, and coincubated for 3 days (n = 3 individual mice). Then the percentages of LinSca-1+c-Kit+, LinSca-1c-Kit+, and LinSca-1+c-Kit cells were measured by flow cytometry. All data are representative of three independent experiments, shown as mean ± s.d., and assessed by one-way ANOVA.

Source data

Extended Data Fig. 5 ILC1s and IFN-γ do not affect leukemia progenitor cell differentiation into myeloid blasts.

a,b, Mouse LSCs labeled with CTV were cocultured with or without mouse ILC1s in the presence or absence of anti-IFN-γ or anti-TNF antibody. Statistics of absolute cell numbers of Mac-1+ (a) and Gr-1+ (b) cells are shown (n = 3 individual mice). c, Leukemia progenitor cells were sorted from the spleen of MllPTD/WT: Flt3ITD/ITD mice with AML and cocultured with or without WT ILC1s, IFN-γ/ ILC1s, or IFN-γ. Representative flow cytometry plots (top) and statistics of the percentages (bottom) of Mac-1+ and Gr-1+ cells are shown (n = 4 individual mice). d, A working model of how ILC1s and their secreted IFN-γ regulate differentiation of LSCs. Data in a, b, and c are representative of three independent experiments, shown as mean ± s.d., and assessed by one-way ANOVA models. NS, not significant.

Source data

Extended Data Fig. 6 ILC1s do not induce HSC apoptosis nor impair their differentiation.

a, Wild-type mouse HSCs from bone marrow of mice were cocultured with or without ILC1s. Representative images and statistics of the percentages of apoptotic cells (5× magnification, scale bar 200 µm, n = 5 individual mice). b, HSCs from blood of healthy donors were cocultured with or without ILC1s. Representative images and statistics of the percentages of apoptotic cells (n = 4 individual donors). c-e, Mouse HSCs were cocultured with or without ILC1s, and representative flow cytometry plots (c), statistics of cell numbers (d), and percentages (e) of LinSca-1+c-Kit+ and LinSca-1c-Kit+ cells (n = 4 individual mice). f, Representative flow cytometry plots and statistics of cell numbers of Mac-1+Gr-1+ cells (n = 4 individual mice). g, Experimental scheme for (h-j). Mouse HSCs from CD45.2 mice were injected into lethally irradiated CD45.1 mice. One day later, ILC1s were injected into those CD45.1 mice. Three weeks later, donor hematopoietic and progenitor cells, myeloid cell subsets, and WBCs were analyzed. h, The cell numbers of donor LSKs, myeloid progenitor cells (LSK+, LinSca-1c-Kit+ cells), early lymphoid-committed precursors (LS+K, LinSca-1+c-Kit cells), short-term hematopoietic stem cells (STHSC, LinSca-1+c-Kit+Flt3CD150CD48 cells), long-term hematopoietic stem cells (LTHSC, LinSca-1+c-Kit+Flt3CD150+CD48 cells), multipotent progenitors 1 and 2 (MPP1, LinSca-1+c-Kit+Flt3CD150+CD48+ cells; MPP2, LinSca-1+c-Kit+Flt3CD150CD48+ cells), Mac-1+Gr-1+ cell subsets, and WBCs derived from CD45.2 mice were analyzed (n = 4 individual mice in no ILC1 group; n = 3 individual mice in ILC1 group). i, Representative flow cytometry plots and statistics of cell numbers of Mac-1+Gr-1+ cells derived from CD45.2 mice (n = 4 individual mice in no ILC1 group; n = 3 individual mice in ILC1 group). j, Statistics of cell numbers of WBCs (n = 4 individual mice in no ILC1 group; n = 3 individual mice in ILC1 group). Data in a, b, d, e, f, h, i, and j are representative of two independent experiments and shown as mean ± s.d.. Statistics are assessed by one-way ANOVA (a and b) or unpaired two-tailed Student’s t test (d, e, h, i, and j). NS, not significant.

Source data

Extended Data Fig. 7 ILC1s control the leukemia burden in mice transplanted with LSCs.

a, LSCs were i.v. co-injected into lethally irradiated (900 cGy) CD45.2 recipient mice on day 0 along with bone marrow cells isolated from IL-15 transgenic mice (CD45.2) as support cells. On day 1, the mice were i.v. injected with WT ILC1s from the liver of C57BL/6 J (CD45.2) mice or i.p. injected daily with recombinant mouse IFN-γ (0.5 μg/mouse/day). Statistics of the numbers of total WBCs at week 5 (n = 5 individual mice in no ILC1 group; n = 4 individual mice in WT ILC1group; n = 6 individual mice in recombinant IFN-γ group). All absolute cell numbers of WBCs were determined by cell counting with the Element HT5 Hematology Analyzer. b, Representative flow cytometry plots of the percentages of CD45.1+ and CD45.2+ cells. c, LSCs were co-injected into lethally irradiated (900 cGy) CD45.1 recipient mice on day 0 along with bone marrow cells isolated from CD45.1 mice (as support cells). Mice were injected with WT ILC1s or IFN-γ−/− ILC1s from the liver of C57BL/6 J (CD45.2) mice on day 1 or injected daily with recombinant mouse IFN-γ (0.5 μg/mouse/day). Statistics of the number of total WBCs at week 3 (n = 7 individual mice). All absolute cell numbers of WBCs were determined by cell counting with the Element HT5 Hematology Analyzer followed by flow cytometry. For box plots, boxplots (a and c) display the median and interquartile range (25th percentile –75th percentile) with whiskers representing the upper- and lower-quartile (1.5× the 75th and 25th percentile values). Data in a and c are representative of two independent experiments and shown as mean ± s.d. and assessed by one-way ANOVA.

Source data

Extended Data Fig. 8 The role of ILC1s and ILC1-derived IFN-γ in controlling LSCs.

a, Healthy mouse liver ILC1s or NK cells were cocultured with or without LSCs in the presence or absence of anti-IL-7Rα neutralizing antibody or isotype IgG control for 12 h along with IL-12 plus IL-15. Representative flow cytometry plots of IFN-γ production by ILC1s (n = 6 individual mice). b, Representative flow cytometry plots of IFN-γ production in healthy liver ILC1s or healthy liver NK cells after treatment with or without IL-7 (100 ng/ml) in the presence of IL-12 plus IL-15 (n = 5 individual mice). c,d, Mouse LSCs were cocultured with or without mouse ILC1s or NK cells for 3 days in the presence or absence of mouse anti-IFN-γ antibody. Representative images (c; 5× magnification, scale bar 200 µm) and statistics of absolute cell numbers (d) are shown (n = 3 individual mice). e,f, Representative flow cytometry plots (e) and statistics of the percentages of apoptotic LSCs (f; n = 3 individual mice). g, To deplete ILC1s or NK cells, WT mice were i.p. injected with IgG control (CTRL), anti-NK1.1, or anti-asialo-GM1 antibody. Three days later, the percentages of NK cells (LinNK1.1+NKp46+CD49b+) and ILC1s (LinNK1.1+NKp46+CD49a+) in the liver of WT mice were measured by flow cytometry. Data (d and f) are representative of two independent experiments and shown as mean ± s.d. and are assessed by one-way ANOVA models. NS, not significant.

Source data

Extended Data Fig. 9 RNA-seq identifies gene transcriptional changes and signaling pathways in LSCs treated with ILC1s or IFN-γ.

a, Experimental design for RNA sequencing (RNA-Seq). Mouse LSCs were sorted and treated with or without sorted ILC1s or IFNγ for 3 days. LSCs were resorted from cocultured ILC1s or IFN-γ using FACS before RNA-Seq. b, Purity of LSCs (left) and ILC1s (right) after cell sorting. c, A heat map showing differential expression of RNA of 627 genes (n = 3 individual mice) is shown. d-f, Volcano plots showing significantly differentially expressed genes in RNA pools from AML LSCs treated with ILC1s vs. Ctrl (untreated) (d), IFN-γ vs. Ctrl (e), and IFN-γ vs. ILC1s (f) (n = 3 individual mice). g, Hallmark pathway analysis in LSC RNA pools (IFN-γ vs. Ctrl). The left panel shows signaling pathways downregulated in LSCs. The right panel shows signaling pathways upregulated in LSCs (n = 3 individual mice). Genes with an FDR-adjusted P-value < 0.05 and a fold change (FC) > 1.5 or < 0.7 were considered as significantly upregulated and downregulated genes, respectively.

Extended Data Fig. 10 ILC1s or IFN-γ inhibit LSC differentiation via the JAK-STAT and AKT signaling pathways.

a, GSEA plots show enrichment of the indicated target genes in LSCs co-cultured with ILC1s. The x axis shows the rank orders (ILC1s vs. Ctrl) of all the genes. b, GSEA plots show enrichment of the indicated target genes in LSCs treated with IFN-γ. The x axis shows the rank orders (IFN-γ vs. Ctrl) of all the genes. c,d, Heat maps showing differential expression of RNAs of genes downstream of IFN-γ. e-h, Mouse LSCs labeled with CTV were treated with or without the indicated JAK and AKT inhibitors for 30 min and then cocultured with or without WT or IFN-γ−/− ILC1s in the presence of IL-12 and IL-15 for 3 days. Statistics of the percentages of LinSca-1+c-Kit+, LinSca-1c-Kit+, LinSca-1+c-Kit, and LinSca-1c-Kit cells (n = 3 individual mice). Genes with an FDR-adjusted P-value < 0.05 and a fold change (FC) > 1.5 or < 0.7 were considered to be significantly upregulated or downregulated. Data in e-h are representative of three independent experiments, shown as mean ± s.d., and assessed by one-way ANOVA models. NS, not significant.

Source data

Supplementary information

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 and unprocessed gels.

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. 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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Z., Ma, R., Ma, S. et al. ILC1s control leukemia stem cell fate and limit development of AML. Nat Immunol 23, 718–730 (2022). https://doi.org/10.1038/s41590-022-01198-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01198-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer