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.

  • Letter
  • Published:

Activation of the transcription factor NFAT5 in the tumor microenvironment enforces CD8+ T cell exhaustion

Abstract

Persistent exposure to antigen during chronic infection or cancer renders T cells dysfunctional. The molecular mechanisms regulating this state of exhaustion are thought to be common in infection and cancer, despite obvious differences in their microenvironments. Here we found that NFAT5, an NFAT family transcription factor that lacks an AP-1 docking site, was highly expressed in exhausted CD8+ T cells in the context of chronic infections and tumors but was selectively required in tumor-induced CD8+ T cell exhaustion. Overexpression of NFAT5 in CD8+ T cells reduced tumor control, while deletion of NFAT5 improved tumor control by promoting the accumulation of tumor-specific CD8+ T cells that had reduced expression of the exhaustion-associated proteins TOX and PD-1 and produced more cytokines, such as IFNɣ and TNF, than cells with wild-type levels of NFAT5, specifically in the precursor exhausted PD–1+TCF1+TIM–3CD8+ T cell population. NFAT5 did not promote T cell exhaustion during chronic infection with clone 13 of lymphocytic choriomeningitis virus. Expression of NFAT5 was induced by TCR triggering, but its transcriptional activity was specific to the tumor microenvironment and required hyperosmolarity. Thus, NFAT5 promoted the exhaustion of CD8+ T cells in a tumor-selective fashion.

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: NFAT5 is upregulated in TI CD8+ T cells.
Fig. 2: NFAT5 deletion in tumor-specific T cells improves tumor control.
Fig. 3: NFAT5 inactivation has limited effect on CD8+ T cell activity during chronic LCMV infection.
Fig. 4: NFAT5 inactivation has a stronger impact on TI CD8+ Tpex cells.
Fig. 5: NFAT5 expression in CD8+ T cells is driven by TCR signaling and NFAT5 activity is induced in a tumor-specific manner.

Similar content being viewed by others

Data availability

Omics data are deposited and accessible under accession numbers GSE224913, GSE237313 and GSE209683. Source data are available for each figure or upon request to the corresponding author. Source data are provided with this paper.

References

  1. Blank, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

    CAS  PubMed  Google Scholar 

  3. Speiser, D. E., Ho, P. C. & Verdeil, G. Regulatory circuits of T cell function in cancer. Nat. Rev. Immunol. 16, 599–611 (2016).

    CAS  PubMed  Google Scholar 

  4. Thommen, D. S. & Schumacher, T. N. T cell dysfunction in cancer. Cancer Cell 33, 547–562 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Cheng, H., Ma, K., Zhang, L. & Li, G. The tumor microenvironment shapes the molecular characteristics of exhausted CD8+ T cells. Cancer Lett. 506, 55–66 (2021).

    CAS  PubMed  Google Scholar 

  6. Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211 (2019).

    CAS  PubMed  Google Scholar 

  7. Baitsch, L. et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Invest. 121, 2350–2360 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Lopez-Rodriguez, C., Aramburu, J., Rakeman, A. S. & Rao, A. NFAT5, a constitutively nuclear NFAT protein that does not cooperate with Fos and Jun. Proc. Natl Acad. Sci. USA 96, 7214–7219 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Cheung, C. Y. & Ko, B. C. NFAT5 in cellular adaptation to hypertonic stress–regulations and functional significance. J. Mol. Signal 8, 5 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Kim, N. H. et al. Reactive oxygen species regulate context-dependent inhibition of NFAT5 target genes. Exp. Mol. Med. 45, e32 (2013).

    PubMed  PubMed Central  Google Scholar 

  11. Alberdi, M. et al. Context-dependent regulation of Th17-associated genes and IFNɣ expression by the transcription factor NFAT5. Immunol. Cell Biol. 95, 56–67 (2016).

    PubMed  PubMed Central  Google Scholar 

  12. Aramburu, J. & Lopez-Rodriguez, C. Regulation of inflammatory functions of macrophages and T lymphocytes by NFAT5. Front. Immunol. 10, 535 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Carmona, S. J., Siddiqui, I., Bilous, M., Held, W. & Gfeller, D. Deciphering the transcriptomic landscape of tumor-infiltrating CD8 lymphocytes in B16 melanoma tumors with single-cell RNA-seq. Oncoimmunology 9, 1737369 (2020).

    PubMed  PubMed Central  Google Scholar 

  14. Xiong, H. et al. Coexpression of inhibitory receptors enriches for activated and functional CD8+ T cells in murine syngeneic tumor models. Cancer Immunol. Res 7, 963–976 (2019).

    CAS  PubMed  Google Scholar 

  15. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 176, 404 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–13086 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Andreatta, M. et al. Interpretation of T cell states from single-cell transcriptomics data using reference atlases. Nat. Commun. 12, 2965 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Prevost-Blondel, A. et al. Tumor-infiltrating lymphocytes exhibiting high ex vivo cytolytic activity fail to prevent murine melanoma tumor growth in vivo. J. Immunol. 161, 2187–2194 (1998).

    CAS  PubMed  Google Scholar 

  20. Martinez-Usatorre, A. et al. Enhanced phenotype definition for precision isolation of precursor exhausted tumor-infiltrating CD8 T cells. Front .Immunol. 11, 340 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Van den Eynde, B., Mazarguil, H., Lethe, B., Laval, F. & Gairin, J. E. Localization of two cytotoxic T lymphocyte epitopes and three anchoring residues on a single nonameric peptide that binds to H-2Ld and is recognized by cytotoxic T lymphocytes against mouse tumor P815. Eur. J. Immunol. 24, 2740–2745 (1994).

    PubMed  Google Scholar 

  22. Shanker, A. et al. CD8 T cell help for innate antitumor immunity. J. Immunol. 179, 6651–6662 (2007).

    CAS  PubMed  Google Scholar 

  23. Giordano, M. et al. Molecular profiling of CD8 T cells in autochthonous melanoma identifies Maf as driver of exhaustion. EMBO J. 34, 2042–2058 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Giordano, M. et al. The tumor necrosis factor α-induced protein 3 (TNFAIP3, A20) imposes a brake on antitumor activity of CD8 T cells. Proc. Natl Acad. Sci. USA 111, 11115–11120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Tong, E. H. et al. Regulation of nucleocytoplasmic trafficking of transcription factor OREBP/TonEBP/NFAT5. J. Biol. Chem. 281, 23870–23879 (2006).

    CAS  PubMed  Google Scholar 

  26. Martinez, G. J. et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Berga-Bolanos, R., Alberdi, M., Buxade, M., Aramburu, J. & Lopez-Rodriguez, C. NFAT5 induction by the pre-T-cell receptor serves as a selective survival signal in T-lymphocyte development. Proc. Natl Acad. Sci. USA 110, 16091–16096 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Chamoto, K., Yaguchi, T., Tajima, M. & Honjo, T. Insights from a 30-year journey: function, regulation and therapeutic modulation of PD1. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-023-00867-9, (2023).

  32. Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).

    CAS  PubMed  Google Scholar 

  33. Charmoy, M., Wyss, T., Delorenzi, M. & Held, W. PD-1+ Tcf1+ CD8+ T cells from established chronic infection can form memory while retaining a stableimprint of persistent antigen exposure. Cell Rep. 36, 109672 (2021).

    CAS  PubMed  Google Scholar 

  34. Kumar, R. et al. NFAT5, which protects against hypertonicity, is activated by that stress via structuring of its intrinsically disordered domain. Proc. Natl Acad. Sci. USA 117, 20292–20297 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Drews-Elger, K., Ortells, M. C., Rao, A., Lopez-Rodriguez, C. & Aramburu, J. The transcription factor NFAT5 is required for cyclin expression and cell cycle progression in cells exposed to hypertonic stress. PLoS ONE 4, e5245 (2009).

    PubMed  PubMed Central  Google Scholar 

  36. Conzelmann, A., Corthesy, P., Cianfriglia, M., Silva, A. & Nabholz, M. Hybrids between rat lymphoma and mouse T cells with inducible cytolytic activity. Nature 298, 170–172 (1982).

    CAS  PubMed  Google Scholar 

  37. Verdeil, G., Chaix, J., Schmitt-Verhulst, A. M. & Auphan-Anezin, N. Temporal cross-talk between TCR and STAT signals for CD8 T cell effector differentiation. Eur. J. Immunol. 36, 3090–3100 (2006).

    CAS  PubMed  Google Scholar 

  38. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Hunt, S. E. et al. Ensembl variation resources. Database 2018, bay119 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  41. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 4, 1521 (2015).

    PubMed  Google Scholar 

  42. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Huynh-Thu, V. A., Irrthum, A., Wehenkel, L. & Geurts, P. Inferring regulatory networks from expression data using tree-based methods. PLoS ONE 5, e12776 (2010).

    PubMed  PubMed Central  Google Scholar 

  44. Herrmann, C., Van de Sande, B., Potier, D. & Aerts, S. i-cisTarget: an integrative genomics method for the prediction of regulatory features and cis-regulatory modules. Nucleic Acids Res. 40, e114 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Imrichova, H., Hulselmans, G., Atak, Z. K., Potier, D. & Aerts, S. i-cisTarget 2015 update: generalized cis-regulatory enrichment analysis in human, mouse and fly. Nucleic Acids Res. 43, W57–W64 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innov. 2, 100141 (2021).

    CAS  Google Scholar 

  48. Andreatta, M. & Carmona, S. J. UCell: robust and scalable single-cell gene signature scoring. Comput Struct. Biotechnol. J. 19, 3796–3798 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank H. Zdimerova for editing the paper, J. Guillaume for contribution to the plasmid production, and P.-C.Ho and T. Walzer for suggestions about the project. This work was funded by the University of Lausanne, and grants from the Max Cloëtta Foundation (G.V.), the Swiss National Science Foundation (G.V.: 310030_182680, 310030_215153, W.H.: 310030_200898), ISREC Institute (D.C.) and the Swiss Cancer Research foundation (G.V.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the paper.

Author information

Authors and Affiliations

Authors

Contributions

L.T, D.C, W.H, M.I, G.C, and G.V conceived and designed the experiments. L.T., D.C., P.R., M.C., G.B., M.M.L., and G.V. performed the experiments. T.W. and J.L. analyzed the scRNA-seq data and M.A., S.C., S.N and I.C. performed bioinformatic analysis. C.L.R. provided Cd4-Cre; Nfat5fl/fl mice. L.T., D.C. and G.V. prepared the figures and wrote the paper with input from all authors. D.E.S., W.H., C.L.R. and S.C. reviewed the paper.

Corresponding author

Correspondence to Grégory Verdeil.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Hai-Hui Xue and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are avaliable. Primary Handling Editor: Ioana Visan 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 Characterization of NFAT5mCherry reporter mice.

a) Fold change of Nfat5 expression level in Tpex or Tex over indicated populations (left). Mean Nfat5 expression level of various CD8+ T cell populations in indicated studies assessed by TILatlas (right). b) NFAT5 reporter mouse strain: The TAG stop codon in exon 14 of the mouse Nfat5 gene was replaced by CRISPR/Cas-mediated genome engineering with the P2A-mCherry cassette to create a knock-in NFAT5-P2A-mCherry reporter model in C57BL/6 mice. c) Example of a gating strategy used throughout the study. d–f) Spleen (d), lymph node (e), and thymus (f) from WT, NFAT5m/wt and NFAT5m/m mice were collected and analyzed for their immune compartment, and thymic development. (e) by flow cytometry. Each dot represents one mouse. g, Relative expression (2-deltaCq) of mCherry and Nfat5 assessed by RT-PCR and normalized to β-2-microglobulin of NFAT5m/wt or NFAT5m/m CD8+ T cells cultured for 72 h under different hyperosmolar conditions ranging from 300 mOsm/kg to 500 mOsm/kg. Each dot represents the mean from a technical replicate from cells from one mouse. h) Histogram showing mCherry expression of NFAT5m/m CD8+ T cells from the lymph node in comparison to a littermate control mouse (WT). i) Western blot showing the total protein level of NFAT5flox/flox CD4-Cre−/− (WT), CD4-Cre+/− NFAT5flox/flox (NFAT5KO), NFAT5m/wt and NFAT5m/m P14 CD8+ T cells cultured in complete RPMI with 1 uM GP33 peptide and 20 U/ml rhIL-2 at 420 mOsm/kg- for 72 hours. β-actin serves as a housekeeping gene. Ratio of NFAT5 level on β-actin has been calculated after quantification of the bands with ImageJ. Biological duplicates shown for each condition. This experieent is representative of 3 independent experiments.

Source data

Extended Data Fig. 2 Overexpression of NFAT5 in tumor specific P1A cell led to reduced tumor control.

a) The four murine isoforms of NFAT5 were aligned on the software Geneious using the entries from GenBank and Ensemble. Domains described by Cheung et al. were aligned against mouse isoforms 203, whose length corresponds to human isoform C. NES: Nuclear export signal, AD1: Activation domain 1, AES: Auxiliary export signal, DRL: DNA recognition loop, DD: Dimerization domain, RHD: Rel homology region, AD2: Activation domain 2, AD3: Activation domain 3. b) Retroviral vector where the various isoforms were cloned. c) Nfat5 mRNA levels in CD8+ T cells transduced with a vector encoding for NFAT5 isoform A (NFAT5 A) (left) or control eGFP (right) and sorted by flow cytometry according to GFP expression. d) Timeline of the experiment. Activated TCRP1A CD8+ T cells were transduced, sorted on the base of GFP expression and transferred into Rag1−/−B10D2 mice seven days post P511 tumor engraftment. CD8+ T cells were sorted for RNA-sequencing on day 14. e) Tumor growth in mice transferred with TCRP1A CD8+ T cells transduced and sorted according to GFP expression with the indicated constructs (control eGFP, NFAT5 A, NFAT5 A lacking the DNA binding domain (DBD), NFAT5 D or NFAT1 CA-RIT). n = 5 mice per group (left) n = 5 mice eGFP, n = 6 mice NFAT5 IsoA, n = 7 mice NFAT5 noDBD, and n = 5 NFAT5 IsoD (middle, right). Two-ways ANOVA was performed. Error bars represent SEM. f) Bioluminescence of the luciferase-expressing TCRP1A CD8+ T cells after injection of the mice with luciferin. Each dot is representing one mouse. g) PC analysis of sorted GFP+ CD8+ TILs transduced with control eGFP, NFAT5 A, or NFAT1 CA-RIT. Each dot is representing one mouse. h, Venn diagram showing the number of genes upregulated in NFAT5 A transduced CD8+ TILs, NFAT1 CA-RIT transduced TILs, or both.

Source data

Extended Data Fig. 3 Tumor infiltrated NFAT5KO P14 cells had superior anti-tumor potential compared to TI NFAT5WT P14 cells in a co-transfer setting.

a) Timeline of the experiment: activated NFAT5WT (CD45.1) and NFAT5KO (CD45.2) P14 CD8+ T cells were co-transferred into B16-GP33bearing CD45.1.2 mice. CD8+ TILs were analyzed 7 days post-ACT. b) Overlay of the GMFI of indicated proteins of NFAT5WT and NFAT5KO on the day of 1:1 transfer. c,d) Representative overlaid histograms of NFAT5WT, NFAT5KO P14, and endogenous CD8+ TILs of indicated proteins (up) with two-sided paired t-test comparison (below). Each pair of dots representing TILs analyzed from one mouse.

Source data

Extended Data Fig. 4 NFAT5 controled Tox expression in hyperosmolar conditions.

a) Analysis of scRNA-seq data of human melanoma tumors: Pearson correlation coefficients were calculated between NFAT5 mRNA levels and mRNA levels of indicated genes. b) Schematic representation of the protocol used to generate in vitro ‘exhausted’ CD8+ T cells (up). Comparison of NFAT5WT (black) and NFAT5KO (red) level of the indicated proteins below. n = 2 mice NFAT5WT and n = 3 mice NFAT5KO. Mann-Whitney test, 2 sided. c), ATACseq and CUT&RUN-seq analysis of the Tox locus. Enhancers 1 and 2 are indicated. d) Frequency of mCherry+ CD8+ cells among GFP+ transduced with reporter constructs for enhancer 1 and enhancer 2 (left). Representative contour plots for the TCR + NaCl conditions is shown (right). n = 2 culture wells (enhancer 1) and n = 3 culture wells (enhancer 2). Data from 2 pooled experiments (2 biological replicates per enhancer). Mann-Whitney test, 2 sided. e) Vectors used for the reporter constructs using enhancers 1 or 2. USCS’s representations of the 2 regions cloned into the promoter region with potential binding sites for NFAT15 (using JASPAR prediction tool).

Source data

Extended Data Fig. 5 scRNAseq analysis of NFAT5KO/WT P14 cells during LCMV cl13 infection showed no significant difference.

a) UMAP where cells are clustered using functions of the Seurat package. b) Cells are colored according to cluster ID, to see if one or the other cluster is enriched in cells from different origins. c) Marker genes used to annotate each cluster at resolution 0.1. d,e) We calculated a module score per cell for the genes that were found to be differentially expressed in the bulk RNA sequencing experiment of NFAT5KO vs NFAT5WT in tumors. The DEG genes with adj. p-value < 0.05 and FC > 1.5 were selected to calculate a score for each single cell. We calculated a signature enrichment score of the up-regulated (d) or down-regulated (e) genes obtained from the bulk RNA sequencing data using the AddModuleScore_UCell function of the UCell package. The score varies from 0 to 1, with 1 as the maximum enrichment of the signature.

Extended Data Fig. 6 TI NFAT5KO P14 Tpex cells showed a reduced exhaustion phenotype when co-transferred with NFAT5WT P14 cells.

a) Representative histograms of NFAT5-mCherry expression in P14 CD8+ T cells from acute (Armstrong) or chronic (clone 13) LCMV-infected mice at day seven or day 28 post-infection. b) Frequency of NFAT5WT and NFAT5KO P14 TI CD8+ among co-transferred cells from mice bearing tumors 7 days post-ACT. c) Frequency of Tpex and Tex among NFAT5WT and NFAT5KO P14 from the tumor 7 days post-ACT. d) GMFI of indicated protein expression in Tpex and Tex among NFAT5WT and NFAT5KO P14 from the tumor 7 days post-ACT. b-d) Two independent experiments were pooled. Paired students t-test, two sided. Each pair of dots is representing Tils analyzed from one mouse.

Source data

Extended Data Fig. 7 Constructs used to report NFAT5 transcriptional activity in vitro and in vivo.

a) Fold change of mCherry for FK506 treated over DMSO treated animals during indicated periods in the tdLN and ndLN with n = 10 mice per timepoint. Mann-Whitney test, two sided. b) Schematic representation of the luciferase NFAT5 activity reporter plasmid. The blue sequence represents the NFAT5 binding site (TonE). c) Schematic representation of the mCherry NFAT5 activity reporter plasmid. The blue sequence represents the NFAT5 binding site (TonE). d) Frequencies of GFP+ mCherry+ NFAT5WT and NFAT5KO P14 CD8+ T cells from the tdLN or the tumor from B16-GP33 bearing mice. Each pair of dots is representing cells analyzed from one mouse. Paired student t-test, two sided.

Source data

Extended Data Table 1 Gene list of commonly expressed genes
Extended Data Table 2 List of Ab and PCR primers

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

Gene list of differentially expressed genes between TI P14 NFAT5KO and TI P14 NFAT5WT 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. 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. 6

Statistical source data.

Source Data Extended Data Fig. 7

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

Tillé, L., Cropp, D., Charmoy, M. et al. Activation of the transcription factor NFAT5 in the tumor microenvironment enforces CD8+ T cell exhaustion. Nat Immunol 24, 1645–1653 (2023). https://doi.org/10.1038/s41590-023-01614-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01614-x

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