Abstract
Diverse cellular insults converge on activation of the heat shock factor 1 (HSF1), which regulates the proteotoxic stress response to maintain protein homoeostasis. HSF1 regulates numerous gene programmes beyond the proteotoxic stress response in a cell-type- and context-specific manner to promote malignancy. However, the role(s) of HSF1 in immune populations of the tumour microenvironment remain elusive. Here, we leverage an in vivo model of HSF1 activation and single-cell transcriptomic tumour profiling to show that augmented HSF1 activity in natural killer (NK) cells impairs cytotoxicity, cytokine production and subsequent anti-tumour immunity. Mechanistically, HSF1 directly binds and regulates the expression of key mediators of NK cell effector function. This work demonstrates that HSF1 regulates the immune response under the stress conditions of the tumour microenvironment. These findings have important implications for enhancing the efficacy of adoptive NK cell therapies and for designing combinatorial strategies including modulators of NK cell-mediated tumour killing.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
The scRNA-seq, ATAC-seq, ChIP-seq and RNA-seq data that support the findings of this study have been deposited in the GEO under the accession code GSE167552. Previously published data that were re-analysed here are available under the following accession codes: GSE72056 (ref. 17), GSE120575 (ref. 18) and GSE134814 (ref. 19). The mm10/GRCm38 reference genome is available at the NCBI under the RefSeq assembly GCF_000001635.20. All other data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.
Code availability
No custom code was used for analysis. Analysis of ChIP-seq, ChIPmentation, ATAC-seq and RNA-seq was performed using the SNS pipeline (https://igordot.github.io/sns/) as described in the Methods. scRNA-seq analysis was performed using standard packages as described in the Methods.
References
Luo, J., Solimini, N. L. & Elledge, S. J. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136, 823–837 (2009).
Akerfelt, M., Morimoto, R. I. & Sistonen, L. Heat shock factors: integrators of cell stress, development and lifespan. Nat. Rev. Mol. Cell Biol. 11, 545–555 (2010).
Dai, C. The heat-shock, or HSF1-mediated proteotoxic stress, response in cancer: from proteomic stability to oncogenesis. Philos. Trans. R. Soc. Lond. B Biol. Sci. 373, 20160525 (2018).
Li, J., Labbadia, J. & Morimoto, R. I. Rethinking HSF1 in stress, development, and organismal health. Trends Cell Biol. 27, 895–905 (2017).
Gomez-Pastor, R., Burchfiel, E. T. & Thiele, D. J. Regulation of heat shock transcription factors and their roles in physiology and disease. Nat. Rev. Mol. Cell Biol. 19, 4–19 (2018).
Dai, C., Whitesell, L., Rogers, A. B. & Lindquist, S. Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis. Cell 130, 1005–1018 (2007).
Mendillo, M. L. et al. HSF1 drives a transcriptional program distinct from heat shock to support highly malignant human cancers. Cell 150, 549–562 (2012).
Santagata, S. et al. Tight coordination of protein translation and HSF1 activation supports the anabolic malignant state. Science 341, 1238303 (2013).
Dong, B., Jaeger, A. M. & Thiele, D. J. Inhibiting heat shock factor 1 in cancer: a unique therapeutic opportunity. Trends Pharmacol. Sci. 40, 986–1005 (2019).
Dai, C. & Sampson, S. B. HSF1: guardian of proteostasis in cancer. Trends Cell Biol. 26, 17–28 (2016).
Kourtis, N. et al. FBXW7 modulates cellular stress response and metastatic potential through HSF1 post-translational modification. Nat. Cell Biol. 17, 322–332 (2015).
Kourtis, N. et al. Oncogenic hijacking of the stress response machinery in T cell acute lymphoblastic leukemia. Nat. Med. 24, 1157–1166 (2018).
Binder, R. J. Functions of heat shock proteins in pathways of the innate and adaptive immune system. J. Immunol. 193, 5765–5771 (2014).
Srivastava, P. Roles of heat-shock proteins in innate and adaptive immunity. Nat. Rev. Immunol. 2, 185–194 (2002).
Scherz-Shouval, R. et al. The reprogramming of tumor stroma by HSF1 is a potent enabler of malignancy. Cell 158, 564–578 (2014).
Vihervaara, A. et al. Transcriptional response to stress in the dynamic chromatin environment of cycling and mitotic cells. Proc. Natl Acad. Sci. USA 110, E3388–E3397 (2013).
Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).
Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).
Scortegagna, M. et al. Siah2 control of T-regulatory cells limits anti-tumor immunity. Nat. Commun. 11, 99 (2020).
Smithies, O., Gregg, R. G., Boggs, S. S., Koralewski, M. A. & Kucherlapati, R. S. Insertion of DNA sequences into the human chromosomal beta-globin locus by homologous recombination. Nature 317, 230–234 (1985).
Mansour, S. L., Thomas, K. R. & Capecchi, M. R. Disruption of the proto-oncogene int-2 in mouse embryo-derived stem cells: a general strategy for targeting mutations to non-selectable genes. Nature 336, 348–352 (1988).
Wang, J. et al. UV-induced somatic mutations elicit a functional T cell response in the YUMMER1.7 mouse melanoma model. Pigment Cell Melanoma Res. 30, 428–435 (2017).
Karlhofer, F. M. & Yokoyama, W. M. Stimulation of murine natural killer (NK) cells by a monoclonal antibody specific for the NK1.1 antigen. IL-2-activated NK cells possess additional specific stimulation pathways. J. Immunol. 146, 3662–3673 (1991).
Lanier, L. L. Up on the tightrope: natural killer cell activation and inhibition. Nat. Immunol. 9, 495–502 (2008).
Martinet, L. & Smyth, M. J. Balancing natural killer cell activation through paired receptors. Nat. Rev. Immunol. 15, 243–254 (2015).
Sun, J. C. & Lanier, L. L. NK cell development, homeostasis and function: parallels with CD8(+) T cells. Nat. Rev. Immunol. 11, 645–657 (2011).
Cursons, J. et al. A gene signature predicting natural killer cell infiltration and improved survival in melanoma patients. Cancer Immunol. Res 7, 1162–1174 (2019).
Chiossone, L. et al. Maturation of mouse NK cells is a 4-stage developmental program. Blood 113, 5488–5496 (2009).
Huntington, N. D., Cursons, J. & Rautela, J. The cancer-natural killer cell immunity cycle. Nat. Rev. Cancer 20, 437–454 (2020).
Guillerey, C., Huntington, N. D. & Smyth, M. J. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 17, 1025–1036 (2016).
Bezman, N. A. et al. Molecular definition of the identity and activation of natural killer cells. Nat. Immunol. 13, 1000–1009 (2012).
Fogel, L. A., Sun, M. M., Geurs, T. L., Carayannopoulos, L. N. & French, A. R. Markers of nonselective and specific NK cell activation. J. Immunol. 190, 6269–6276 (2013).
Okamura, H., Kashiwamura, S., Tsutsui, H., Yoshimoto, T. & Nakanishi, K. Regulation of interferon-γ production by IL-12 and IL-18. Curr. Opin. Immunol. 10, 259–264 (1998).
Lopez-Soto, A., Gonzalez, S., Smyth, M. J. & Galluzzi, L. Control of metastasis by NK cells. Cancer Cell 32, 135–154 (2017).
Souza-Fonseca-Guimaraes, F., Cursons, J. & Huntington, N. D. The emergence of natural killer cells as a major target in cancer immunotherapy. Trends Immunol. 40, 142–158 (2019).
Heng, T. S. et al. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).
Adams, N. M. et al. Transcription factor IRF8 orchestrates the adaptive natural killer cell response. Immunity 48, 1172–1182.e6 (2018).
Carson, W. E. et al. Interleukin (IL) 15 is a novel cytokine that activates human natural killer cells via components of the IL-2 receptor. J. Exp. Med. 180, 1395–1403 (1994).
Tang, F. et al. A pan-cancer single-cell panorama of human natural killer cells. Cell 186, 4235–4251.e20 (2023).
Abel, A. M., Yang, C., Thakar, M. S. & Malarkannan, S. Natural killer cells: development, maturation, and clinical utilization. Front. Immunol. 9, 1869 (2018).
Mace, E. M. et al. Biallelic mutations in IRF8 impair human NK cell maturation and function. J. Clin. Invest. 127, 306–320 (2017).
Becknell, B. & Caligiuri, M. A. Interleukin-2, interleukin-15, and their roles in human natural killer cells. Adv. Immunol. 86, 209–239 (2005).
Jounaidi, Y., Cotten, J. F., Miller, K. W. & Forman, S. A. Tethering IL2 to its receptor IL2Rβ enhances antitumor activity and expansion of natural killer NK92 cells. Cancer Res. 77, 5938–5951 (2017).
Hietanen, T., Kapanen, M. & Kellokumpu-Lehtinen, P. L. Natural killer cell viability after hyperthermia alone or combined with radiotherapy with or without cytokines. Anticancer Res. 38, 655–663 (2018).
Inoue, S., Ikehara, S., Nakamura, T., Good, R. A. & Hamashima, Y. Two natural killer-cell subpopulations distinguished by heat sensitivity. J. Clin. Immunol. 5, 421–426 (1985).
Won, S. J. & Lin, M. T. Thermal stresses reduce natural killer cell cytotoxicity. J. Appl Physiol. 79, 732–737 (1995).
Kokolus, K. M. et al. Baseline tumor growth and immune control in laboratory mice are significantly influenced by subthermoneutral housing temperature. Proc. Natl Acad. Sci. USA 110, 20176–20181 (2013).
Krämer, B. et al. Early IFN-alpha signatures and persistent dysfunction are distinguishing features of NK cells in severe COVID-19. Immunity 54, 2650–2669.e14 (2021).
Lee, N. et al. Strategy of using intratreatment hypoxia imaging to selectively and safely guide radiation dose de-escalation concurrent with chemotherapy for locoregionally advanced human papillomavirus-related oropharyngeal carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 96, 9–17 (2016).
Shimasaki, N., Jain, A. & Campana, D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 19, 200–218 (2020).
Laskowski, T. J., Biederstadt, A. & Rezvani, K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat. Rev. Cancer 22, 557–575 (2022).
Le Masson, F. et al. Identification of heat shock factor 1 molecular and cellular targets during embryonic and adult female meiosis. Mol. Cell. Biol. 31, 3410–3423 (2011).
Chiriboga, L., Callis, G. M., Wang, Y. & Chlipala, E. Guide for collecting and reporting metadata on protocol variables and parameters from slide-based histotechnology assays to enhance reproducibility. J. Histotechnol. 45, 132–147 (2022).
Day, W. A. et al. Covalently deposited dyes: a new chromogen paradigm that facilitates analysis of multiple biomarkers in situ. Lab Invest. 97, 104–113 (2017).
Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.21–21.29.29 (2015).
Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).
Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).
Nam, A. S. et al. Somatic mutations and cell identity linked by genotyping of transcriptomes. Nature 571, 355–360 (2019).
Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).
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).
Yu, G., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).
Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).
Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).
Acknowledgements
We thank members of the Aifantis laboratory for helpful discussions; J. Weber for the B16-F10 cell line and M. Bosenberg for the YUMM1.7 and YUMMER1.7 cell lines; S. Koralov for expertise in Southern blotting technology; the members of the Interdisciplinary Melanoma Cooperative Group at the NYU Perlmutter Cancer Center for scientific input and discussions; A. Heguy and the NYU Genome Technology Center, supported in part by the grant P30CA016087 from the National Institutes of Health (NIH) and the National Cancer Institute (NCI), for expertise with sequencing experiments; the NYU Histology Core (supported in part by the grant P30CA016087 from the NIH, NCI) for assistance; and the NYU Langone Cytometry and Cell Sorting Laboratory (supported in part by grant P30CA016087 from the NIH/NCI) for cell sorting and flow cytometry technologies. This work was supported in part by the Laura and Isaac Perlmutter Cancer Center Support Grant; NIH/NCI P30CA016087 and the NYULH Center for Biospecimen Research and Development, Histology and Immunohistochemistry Laboratory (RRID:SCR_018304). This work used computing resources at the High-Performance Computing Facility at the NYU Medical Center. Figure 2a,c,g and Extended Data Fig. 4d were created with BioRender.com released under an Academic Licence. I.A. is supported by the NCI at the NIH (grant nos. RO1CA202025, RO1CA202027, RO1CA228135 and 1P50CA225450). K.H. is supported by the NIH (training grants MSTP T32 GM136573, T32 CA009161 (Levy) and the Ruth L. Kirschstein Predoctoral NRSA F30 CA232704). N.K. is supported a Human Frontiers Science Program Long Term Fellowship (LT000150/2013-L) and previously by a Charles H. Revson Senior Fellowship in Biomedical Science (15–31) and a European Molecular Biology Organization (EMBO) Long Term Fellowship (ALTF 850-2012). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.
Author information
Authors and Affiliations
Contributions
I.A., N.K. and K.H. conceptualized the project. K.H., N.K., G.G. and L.C. designed and performed biological experiments with the assistance of E.B., O.I., G.G., M.G., L.C., K.A., X.C., K.C., K.B. and Z.S. G.J. performed analysis of immunohistochemistry experiments. T.S., H.Z. and I.D. developed software and data curation for computational analysis. K.H., T.S., E.B., G.J., I.D., H.Z. and M.S. performed computational analyses. A.T. and I.A. provided access to instrumentation, computing resources, analysis tools and supervision. K.H., T.S., N.K. and M.S. prepared data and schematics for visualization. L.C. provided resources for multiplex immunohistochemistry experiments. I.O. provided patient samples from the IMCG database. K.H., N.K. and I.A. were responsible for funding acquisition. K.H., N.K. and I.A. wrote the original draft. K.H., N.K., T.S., E.B., M.G., I.O., A.T. and I.A. reviewed the manuscript. K.H., N.K. and I.A. edited the manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Variability of HSF1 activity and regulation between populations of the tumour microenvironment.
a, Heatmap of the HSF1 signature in distinct populations in tumours of patients with melanoma16. b, Violin plots and boxplots of the stress score across tumour-infiltrating immune populations in patients with melanoma17. One-way ANOVA. The n values for each cell type are as follows: MF 1267, pDC 282, Treg 3947, CD8T 6744, gdT 948, B 1729, NK 724. c, Heatmap of the HSF1 signature in distinct populations in murine melanoma tumours in wild-type mice18. d, Violin plots and boxplots of the stress score across tumour-infiltrating immune populations in murine melanoma18. One-way ANOVA. The n values for each cell type are as follows: CD8T 474, Treg 182, NK 398, DC 202, MF 2437. e, Boxplots of HSF1 (left) and FBXW7 (right) expression in individual cells from tumour-infiltrating immune populations assessed by scRNA-seq16. The n values for each cell type are as follows: CD8T 1044, MF 125, Treg 57, NK 52, B 515, CD4T 35, T 504, Th 428. b, d, e, Boxplots represent the bottom quartile to top quartile with median indicated. Whiskers represent −1.5 x IQR to 1.5 x IQR. Dots represent outliers from the minima to the maxima of the data.
Extended Data Fig. 2 An in vivo model of augmented HSF1 exhibiting normal hematopoiesis.
a, Schematic of knock-in targeting strategy. AG to GC mutations are indicated by the asterisk. Length in bp: long homology arm (LA) – 6714, point mutation – 14, middle arm (MA) – 404, short homology arm (SA) – 2014, targeted region – 418. b, Identification of embryonic stem cell clones with stable incorporation of the targeting vector. The wild-type (WT, 8591 bp) and knock-in (MUT or S303/7 A, 7567 bp) bands as indicated; the clone (#8) selected for microinjection as indicated by the asterisk. c, Sanger sequencing of the nucleotides corresponding to amino acids 302-308 of the HSF1 protein in mice wild-type or homozygous for the knock-in allele. Positions 303 and 307 are underlined. d, Kaplan-Meier survival analysis of the lifespan of WT or S303/7 A mice (n = 12). Log-rank (Mantel-Cox) test. e, Immunohistochemistry staining with anti-HSF1 antibody of the indicated tissue types from WT (left) or S303/7 A (right) mice. Representative of 3 biological replicates. f, Gating strategy for myeloid populations. g, Gating strategy for lymphoid populations. h, Frequency of myeloid populations in S303/7 A or WT spleen. i, Total count of myeloid populations in S303/7 A or WT spleen. j, Frequency of lymphoid populations in S303/7 A or WT spleen. k, Total count of lymphoid populations in S303/7 A or WT spleen. l, Gating strategy for multipotent progenitors (MPPs) and lineage-biased progenitors. Lineage includes B220, CD11b, CD3e, CD4, CD8a, Ly-6C/Ly-6G, NK1.1, Ter-119. m, Frequency of MPPs in S303/7 A or WT bone marrow. n, Total count of MPPs in S303/7 A or WT bone marrow. o, Frequency of lineage-biased progenitors in S303/7 A or WT bone marrow. p, Total count of lineage-biased progenitors in S303/7 A or WT bone marrow. h-k, m-p, n = 4 biological replicates. p calculated by two-tailed t-test, bars represent the mean and error bars represent the SEM.
Extended Data Fig. 3 Expression of NK cell ligands and receptors.
a, Tumour volumes over time of 1 × 105 YUMMER1.7 cells injected into WT (n = 4) or S303/7 A (n = 5) mice. b, Tumour volumes over time of 2.5 × 105 YUMMER1.7 cells injected into WT (n = 4) or S303/7 A (n = 5) mice. c, Tumour volumes over time of 5 × 105 YUMMER1.7 cells injected into WT (n = 4) or S303/7 A (n = 5) mice. e, Representative histograms of NK cell receptor ligands and isotype controls in B16-F10 and YUMMER1.7 cells. f, quantification of expression of NK cell receptor ligands normalized to isotype controls in B16-F10 and YUMMER1.7. n = 3 technical replicates. bars represent the mean for visualization. g, Immunoblot analysis of HSF1 in total lysate of thymocytes from Hsf1+/+ or Hsf1−/− mice. n = 3 biological replicates. h, Frequency of Hsf1+/+ or Hsf1−/− NK cells expressing surface activating receptors. i, Frequency of Hsf1+/+ or Hsf1−/− NK cells expressing surface inhibitory receptors. j, Frequency of Hsf1+/+ or Hsf1−/− NK cells across the maturation stages. h – j, n = 3 biological replicates, bars represent the mean and error bars represent the SEM, p calculated by two-tailed t-test.
Extended Data Fig. 4 HSF1-stabilized NK cells exhibit decreased IFN-γ production and proliferation.
a, Quantification of CD62L on naïve splenic NK cells in WT or S303/7 A mice. Representative of 2 independent experiments, n = 4 biological replicates. b, Histograms displaying the expression level of GZMB in NK cells cultured with B16-F10 (1:1) or PMA/Ionomycin for 4 h. c, Quantification of GZMB in WT (n = 5) or S303/7 A (n = 4) NK cells following stimulation (4 h) with the conditions indicated. d, Schematic of adoptive transfer of WT or S303/7 A NK cells into Rag2−/−Il2rg−/− mice followed by metastatic challenge with B16-F10 through tail vein injection. e, Representative H&E staining of lung from S303/7 A mice (n = 5) or WT mice (n = 4). Scale bars as indicated. f, Quantification of metastatic lung nodules from H&E images (left) and quantification of tumour as a proportion of total lung tissue (right). p calculated by two-tailed t-test. Boxplots represent the bottom quartile to top quartile with median indicated. Whiskers represent the minima to the maxima of the data. n = 5 for S303/7 A mice and n = 4 for WT mice g, Immunoblot analysis of HSF1 in nuclear lysate from WT or S303/7 A NK cells with or without heat shock as indicated. h, Cell number over time of NK cells from WT (n = 5) or S303/7 A (n = 4) mice expanded with IL-15. p calculated by two-way ANOVA, data presented as mean values, error bars represent the SEM. a, c, bars represent the mean, error bars represent the SEM. p calculated using two-tailed t-test.
Extended Data Fig. 5 Single-cell transcriptomic characterization of the HSF1-stabilized microenvironment.
a, Schematic of experiment approach. b, Gating strategy for enrichment of CD45+ cells. Live, viable, singlet cells are shown. c, Uniform Manifold Approximation and Projection (UMAP) representation depicting the integration of 6654 HSF1WT (WT) and 4802 HSF1S303/7A (SS03/7 A) cells. d, UMAP representation of colour-coded clustering of the melanoma tumour microenvironment (11,456 cells). e, Normalized expression level of Ptprc (CD45) overlaid on UMAP representation. f, Copy-number variant (CNV) analysis to identify melanoma cells. Heatmap representation of CNVs by chromosome. g, CNV clusters from analysis in F overlaid on UMAP representation. h, A heatmap representation of population-specific markers of immune populations. i, Cell type identification (left) and population proportions (right) based on transcriptional signatures from the Immunological Genome Project. Immune cell types were called based on similarity from bulk transcriptomic signatures of FACS-sorted immune populations. j, Bar graphs of cell cycle status within each cluster by HSF1 status. k, Log2 UMI score of the NK effector signature across CD56brightCD16lo and CD56dimCD16hi NK cell from blood, normal tissue, and tumour, separated by high (>1 standard deviation above the mean) and low (<1 standard deviation below the mean) ‘stress’ score. The n values for each category/cell type are as follows: CD56brightCD16lo: tumour (high) 2081, tumour (low) 1643, normal (high) 532, normal (low) 499, blood (high) 876, blood (low) 932; CD56dimCD16hi: tumour (high) 3351, tumour (low) 2673, normal (high) 2497, normal (low) 2224, blood (high) 6273, blood (low) 7083. p calculated by Kruskal-Wallis t-test. Boxplots represent the bottom quartile to top quartile with median indicated. Whiskers represent −1.5 x IQR to 1.5 x IQR. Dots represent outliers from the minima to the maxima of the data.
Extended Data Fig. 6 Enrichment analysis of genes directly bound by HSF1 in NK cells.
a, Gene set enrichment analysis (GSEA) of HSF1 peaks ranked by adjusted p-value. Top 10 enriching gene sets from the REACTOME database are shown, with those comprised of molecular chaperones indicated in red. b, GSEA of HSF1 peaks ranked by adjusted p-value. Top 10 enriching gene sets from the KEGG database are shown, with natural killer cell-mediated cytotoxicity indicated in red. c, Venn diagram of overlapping peaks in HSF1S303/7A (S303/7 A) and wild-type (WT) NK cells. d, Pie chart of genomic distribution of regions of HSF1 occupancy (3′ UTR, 5′ UTR, distal intergenic, downstream, exon, intron, promoter) in primary NK cells. a, b, p estimation using an adaptive multi-level split Monte-Carlo scheme using Benjamini–Hochberg adjustment for multiple comparisons.
Supplementary information
Supplementary Table 1
Table of sequences for primers and oligonucleotides used in the manuscript.
Supplementary Table 2
Table of fluorescence cytometry antibody information.
Supplementary Table 3
Table of buffer recipes for ChIP-seq and ChIPmentation protocols.
Source data
Source Data Fig. 1
Statistical source data for Fig. 1.
Source Data Fig. 2
Statistical source data for Fig. 2.
Source Data Fig. 2
Unprocessed source data for Fig. 2b.
Source Data Fig. 3
Statistical source data for Fig. 3.
Source Data Fig. 4
Statistical source data for Fig. 4.
Source Data Fig. 4
Unprocessed source data for Fig. 4k.
Source Data Fig. 5
Statistical source data for Fig. 5.
Source Data Extended Data Fig. 2
Statistical source data for Extended Data Fig. 2.
Source Data Extended Data Fig. 2
Unprocessed source data for Extended Data Fig. 2b.
Source Data Extended Data Fig. 3
Statistical source data for Extended Data Fig. 3.
Source Data Extended Data Fig. 3
Unprocessed source data for Extended Data Fig. 3g.
Source Data Extended Data Fig. 4
Statistical source data for Extended Data Fig. 4.
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.
About this article
Cite this article
Hockemeyer, K., Sakellaropoulos, T., Chen, X. et al. The stress response regulator HSF1 modulates natural killer cell anti-tumour immunity. Nat Cell Biol (2024). https://doi.org/10.1038/s41556-024-01490-z
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41556-024-01490-z