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:

Rebalancing TGFβ1/BMP signals in exhausted T cells unlocks responsiveness to immune checkpoint blockade therapy

Abstract

T cell dysfunctionality prevents the clearance of chronic infections and cancer. Furthermore, epigenetic programming in dysfunctional CD8+ T cells limits their response to immunotherapies, including immune checkpoint blockade (ICB). However, it is unclear which upstream signals drive acquisition of dysfunctional epigenetic programs, and whether therapeutically targeting these signals can remodel terminally dysfunctional T cells to an ICB-responsive state. Here we innovate an in vitro model system of stable human T cell dysfunction and show that chronic TGFβ1 signaling in posteffector CD8+ T cells accelerates their terminal dysfunction through stable epigenetic changes. Conversely, boosting bone morphogenetic protein (BMP) signaling while blocking TGFβ1 preserved effector and memory programs in chronically stimulated human CD8+ T cells, inducing superior responses to tumors and synergizing the ICB responses during chronic viral infection. Thus, rebalancing TGFβ1/BMP signals provides an exciting new approach to unleash dysfunctional CD8+ T cells and enhance T cell immunotherapies.

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: Posteffector TGFβ1 drives severe dysfunction of chronically stimulated human CD8+ T cells.
Fig. 2: Chronic TGFβ1 and TCR signals establish a stable dysfunctional program in CD8+ T cells.
Fig. 3: BMP4 agonist treatment limits exhaustion features and enhances survival of human CD8+ T cells.
Fig. 4: Boosting BMP signaling while targeting TGFβ1 modulates terminal T cell dysfunction.
Fig. 5: Rebalancing TGFβ1/BMP signals remodels gene expression programs in dysfunctional human T cells.
Fig. 6: Epigenetic remodeling of dysfunctional human T cells by targeting TGFβ1 and boosting BMP.
Fig. 7: Treated CD8+ T cells exhibit superior antitumor cytotoxic functions.
Fig. 8: Combined TGFβR1i and BMP4a treatment synergizes CD8+ T cell responses to PD-L1 blockade.

Similar content being viewed by others

Data availability

WGBS datasets and RNA-seq data have been deposited in GEO under the accession codes GSE217087 and GSE217072, respectively. Published datasets accessed throughout this study include Supplementary Table 1 (Fig.1i; see table for dataset accession numbers), ChIP-Atlas Enrichment Analysis (https://chip-atlas.org/enrichment_analysis; Figs. 5d,6i), C7 Immune Signature Database GSE41867 and GSE9650 (Fig. 5f), WebGestalt (http://www.webgestalt.org/# for GO and ORA; Fig. 5g–i), Enrichr using NCI-Nature 2016 database for pathway enrichment analysis (http://amp.pharm.mssm.edu/Enrichr; Fig. 6e and Extended Data Fig. 6b), PRJNA546023, GSE149810 and GSE84105 (Extended Data Fig. 3a,b), Hallmark gene signature MSigDB database for GSEA from UC San Diego and Broad Institute (https://www.gsea-msigdb.org/gsea/msigdb/index.jsp; Extended Data Fig. 6d), Yellow fever vaccination RNA-seq data (GSE100745; Extended Data Fig. 5b,d), Pan-Cancer TILs expression RNA-seq data (GSE156728; Extended Data Fig. 7) and scATAC-seq data (GSE129785; Extended Data Fig. 8d,e). Source data are provided with this paper.

Code availability

The code generated and used for the analysis of sequencing data is available from the corresponding author on reasonable request.

References

  1. McLane, L. M., Abdel-Hakeem, M. S. & Wherry, E. J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 37, 457–495 (2019).

    Article  CAS  Google Scholar 

  2. Sharma, P. & Allison, J. P. Dissecting the mechanisms of immune checkpoint therapy. Nat. Rev. Immunol. 20, 75–76 (2020).

    Article  CAS  Google Scholar 

  3. van der Leun, A. M., Thommen, D. S. & Schumacher, T. N. CD8+ T cell states in human cancer: insights from single-cell analysis. Nat. Rev. Cancer 20, 218–232 (2020).

    Article  Google Scholar 

  4. Møller, S. H., Hsueh, P.-C., Yu, Y.-R., Zhang, L. & Ho, P.-C. Metabolic programs tailor T cell immunity in viral infection, cancer, and aging. Cell Metab. 34, 378–395 (2022).

    Article  Google Scholar 

  5. Hudson, W. H. et al. Proliferating transitory t cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic infection. Immunity 51, 1043–1058 (2019).

    Article  CAS  Google Scholar 

  6. Chen, Y. et al. BATF regulates progenitor to cytolytic effector CD8+ T cell transition during chronic viral infection. Nat. Immunol. 22, 996–1007 (2021).

    Article  CAS  Google Scholar 

  7. Raju, S. et al. Identification of a T-bethi quiescent exhausted CD8 T cell subpopulation that can differentiate into TIM3+ CX3CR1+ effectors and memory-like cells. J. Immunol. 206, 2924–2936 (2021).

    Article  CAS  Google Scholar 

  8. Henning, A. N., Roychoudhuri, R. & Restifo, N. P. Epigenetic control of CD8+ T cell differentiation. Nat. Rev. Immunol. 18, 340–356 (2018).

    Article  CAS  Google Scholar 

  9. Saadey, A. A., Yousif, A. & Ghoneim, H. E. Chapter Seven - Epigenetic programming of the immune responses in cancer. Cancer Immunology and Immunotherapy (eds Amiji, M. M. & Milane, L. S.) 197–235 (Academic Press, 2022).

  10. Saini, A., Ghoneim, H. E., Lio, C.-W., Collins, P. L. & Oltz, E. M. Gene regulatory circuits in innate and adaptive immune cells. Annu. Rev. Immunol. 40, 387–411 (2022).

    Article  Google Scholar 

  11. Ghoneim, H. E. et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142–157 (2017).

    Article  CAS  Google Scholar 

  12. Abdel-Hakeem, M. S. et al. Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation. Nat. Immunol. 22, 1008–1019 (2021).

    Article  CAS  Google Scholar 

  13. Yates, K. B. et al. Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans. Nat. Immunol. 22, 1020–1029 (2021).

    Article  CAS  Google Scholar 

  14. Hensel, N. et al. Memory-like HCV-specific CD8+ T cells retain a molecular scar after cure of chronic HCV infection. Nat. Immunol. 22, 229–239 (2021).

    Article  CAS  Google Scholar 

  15. Tonnerre, P. et al. Differentiation of exhausted CD8+ T cells after termination of chronic antigen stimulation stops short of achieving functional T cell memory. Nat. Immunol. 22, 1030–1041 (2021).

    Article  CAS  Google Scholar 

  16. Yousif, A. & Ghoneim, H. E. T cell exhaustion—a memory locked behind scars. Nat. Immunol. 22, 938–940 (2021).

    Article  CAS  Google Scholar 

  17. Collier, J. L., Weiss, S. A., Pauken, K. E., Sen, D. R. & Sharpe, A. H. Not-so-opposite ends of the spectrum: CD8+ T cell dysfunction across chronic infection, cancer and autoimmunity. Nat. Immunol. 22, 809–819 (2021).

    Article  CAS  Google Scholar 

  18. Abdelsamed, H. A. et al. Beta cell-specific CD8+ T cells maintain stem cell memory-associated epigenetic programs during type 1 diabetes. Nat. Immunol. 21, 578–587 (2020).

    Article  CAS  Google Scholar 

  19. Page, N. et al. Persistence of self-reactive CD8+ T cells in the CNS requires TOX-dependent chromatin remodeling. Nat. Commun. 12, 1009 (2021).

    Article  CAS  Google Scholar 

  20. Li, Y. & Kurlander, R. J. Comparison of anti-CD3 and anti-CD28-coated beads with soluble anti-CD3 for expanding human T cells: differing impact on CD8 T cell phenotype and responsiveness to restimulation. J. Transl. Med. 8, 104 (2010).

    Article  Google Scholar 

  21. Belk, J. A. et al. Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Cancer Cell 40, 768–786 (2022).

    Article  CAS  Google Scholar 

  22. Gorelik, L. & Flavell, R. A. Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat. Med. 7, 1118–1122 (2001).

    Article  CAS  Google Scholar 

  23. Tauriello, D. V. F., Sancho, E. & Batlle, E. Overcoming TGFβ-mediated immune evasion in cancer. Nat. Rev. Cancer 22, 25–44 (2022).

    Article  CAS  Google Scholar 

  24. Moreau, J. M., Velegraki, M., Bolyard, C., Rosenblum, M. D. & Li, Z. Transforming growth factor-β1 in regulatory T cell biology. Sci. Immunol. 7, eabi4613 (2022).

    Article  CAS  Google Scholar 

  25. Christo, S. N. et al. Discrete tissue microenvironments instruct diversity in resident memory T cell function and plasticity. Nat. Immunol. 22, 1140–1151 (2021).

    Article  CAS  Google Scholar 

  26. Chen, C.-H. et al. Transforming growth factor beta blocks Tec kinase phosphorylation, Ca2+ influx, and NFATc translocation causing inhibition of T cell differentiation. J. Exp. Med. 197, 1689–1699 (2003).

    Article  CAS  Google Scholar 

  27. Duhen, T. et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat. Commun. 9, 2724 (2018).

    Article  Google Scholar 

  28. Kwoczek, J. et al. Cord blood-derived T cells allow the generation of a more naïve tumor-reactive cytotoxic T-cell phenotype. Transfusion 58, 88–99 (2018).

    Article  CAS  Google Scholar 

  29. West, E. E. et al. Tight regulation of memory CD8+ T cells limits their effectiveness during sustained high viral load. Immunity 35, 285–298 (2011).

    Article  CAS  Google Scholar 

  30. Nolz, J. C. & Harty, J. T. Protective capacity of memory CD8+ T cells is dictated by antigen exposure history and nature of the infection. Immunity 34, 781–793 (2011).

    Article  CAS  Google Scholar 

  31. Good, C. R. et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 184, 6081–6100 (2021).

    Article  CAS  Google Scholar 

  32. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).

    Article  CAS  Google Scholar 

  33. Weber, E. W. et al. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science 372, eaba1786 (2021).

    Article  CAS  Google Scholar 

  34. Beltra, J.-C. et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52, 825–841 (2020).

    Article  CAS  Google Scholar 

  35. David, C. J. & Massagué, J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat. Rev. Mol. Cell Biol. 19, 419–435 (2018).

    Article  CAS  Google Scholar 

  36. Mariathasan, S. et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554, 544–548 (2018).

    Article  CAS  Google Scholar 

  37. Rachidi, S. et al. Platelets subvert T cell immunity against cancer via GARP-TGFβ axis. Sci. Immunol. 2, eaai7911 (2017).

    Article  Google Scholar 

  38. Martin, C. J. et al. Selective inhibition of TGFβ1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape. Sci. Transl. Med. 12, eaay8456 (2020).

    Article  CAS  Google Scholar 

  39. Gunderson, A. J. et al. TGFβ suppresses CD8+ T cell expression of CXCR3 and tumor trafficking. Nat. Commun. 11, 1749 (2020).

    Article  CAS  Google Scholar 

  40. Hore, T. A. et al. Retinol and ascorbate drive erasure of epigenetic memory and enhance reprogramming to naïve pluripotency by complementary mechanisms. Proc. Natl Acad. Sci. USA 113, 12202–12207 (2016).

    Article  CAS  Google Scholar 

  41. Giles, J. R. et al. Human epigenetic and transcriptional T cell differentiation atlas for identifying functional T cell-specific enhancers. Immunity 55, 557–574 (2022).

    Article  CAS  Google Scholar 

  42. Woehrle, T. et al. Pannexin-1 hemichannel-mediated ATP release together with P2X1 and P2X4 receptors regulate T-cell activation at the immune synapse. Blood 116, 3475–3484 (2010).

    Article  CAS  Google Scholar 

  43. Thaker, Y. R., Raab, M., Strebhardt, K. & Rudd, C. E. GTPase-activating protein Rasal1 associates with ZAP-70 of the TCR and negatively regulates T-cell tumor immunity. Nat. Commun. 10, 4804 (2019).

    Article  Google Scholar 

  44. O’Connell, P. et al. SLAMF7 signaling reprograms T cells toward exhaustion in the tumor microenvironment. J. Immunol. 206, 193–205 (2021).

    Article  Google Scholar 

  45. Zheng, L. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021).

    Article  Google Scholar 

  46. Youngblood, B. et al. Effector CD8 T cells dedifferentiate into long-lived memory cells. Nature 552, 404–409 (2017).

    Article  CAS  Google Scholar 

  47. Oki, S. et al. ChIP-Atlas: a data-mining suite powered by full integration of public ChIP-seq data. EMBO Rep. 19, e46255 (2018).

    Article  Google Scholar 

  48. Yamada, T., Park, C. S., Mamonkin, M. & Lacorazza, D. The transcription factor ELF4 controls proliferation and homing of CD8+ T cells via the Krüppel-like factors KLF4 and KLF2. Nat. Immunol. 10, 618–626 (2009).

    Article  CAS  Google Scholar 

  49. Satpathy, A. T. et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat. Biotechnol. 37, 925–936 (2019).

    Article  CAS  Google Scholar 

  50. Philip, M. et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545, 452–456 (2017).

    Article  CAS  Google Scholar 

  51. Kalbasi, A. & Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 20, 25–39 (2020).

    Article  CAS  Google Scholar 

  52. Tinoco, R., Alcalde, V., Yang, Y., Sauer, K. & Zuniga, E. I. Cell-intrinsic transforming growth factor-beta signaling mediates virus-specific CD8+ T cell deletion and viral persistence in vivo. Immunity 31, 145–157 (2009).

    Article  CAS  Google Scholar 

  53. Gabriel, S. S. et al. Transforming growth factor-β-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection. Immunity 54, 1698–1714 (2021).

    Article  CAS  Google Scholar 

  54. Matloubian, M., Concepcion, R. J. & Ahmed, R. CD4+ T cells are required to sustain CD8+ cytotoxic T-cell responses during chronic viral infection. J. Virol. 68, 8056–8063 (1994).

    Article  CAS  Google Scholar 

  55. Mouillesseaux, K. P. et al. Notch regulates BMP responsiveness and lateral branching in vessel networks via SMAD6. Nat. Commun. 7, 13247 (2016).

    Article  CAS  Google Scholar 

  56. Pauken, K. E. et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 354, 1160–1165 (2016).

    Article  CAS  Google Scholar 

  57. Angelosanto, J. M., Blackburn, S. D., Crawford, A. & Wherry, E. J. Progressive loss of memory T cell potential and commitment to exhaustion during chronic viral infection. J. Virol. 86, 8161–8170 (2012).

    Article  CAS  Google Scholar 

  58. Hu, Y. et al. TGF-β regulates the stem-like state of PD-1+ TCF-1+ virus-specific CD8 T cells during chronic infection. J. Exp. Med. 219, e20211574 (2022).

    Article  CAS  Google Scholar 

  59. Ponomarev, L. C., Ksiazkiewicz, J., Staring, M. W., Luttun, A. & Zwijsen, A. The BMP pathway in blood vessel and lymphatic vessel biology. Int. J. Mol. Sci. 22, 6364 (2021).

    Article  CAS  Google Scholar 

  60. Jeong, S. et al. BMP4 and perivascular cells promote hematopoietic differentiation of human pluripotent stem cells in a differentiation stage-specific manner. Exp. Mol. Med. 52, 56–65 (2020).

    Article  CAS  Google Scholar 

  61. Goldman, D. C. et al. BMP4 regulates the hematopoietic stem cell niche. Blood 114, 4393–4401 (2009).

    Article  CAS  Google Scholar 

  62. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  Google Scholar 

  63. Nakashima, H. et al. Modeling tumor immunity of mouse glioblastoma by exhausted CD8+ T cells. Sci. Rep. 8, 208 (2018).

    Article  Google Scholar 

  64. Abdelsamed, H. A. et al. Human memory CD8 T cell effector potential is epigenetically preserved during in vivo homeostasis. J. Exp. Med. 214, 1593–1606 (2017).

    Article  CAS  Google Scholar 

  65. Sakamoto, Y. et al. Long-read whole-genome methylation patterning using enzymatic base conversion and nanopore sequencing. Nucleic Acids Res. 49, e81 (2021).

    Article  CAS  Google Scholar 

  66. Xi, Y. & Li, W. BSMAP: whole genome bisulfite sequence MAPping program. BMC Bioinf. 10, 232 (2009).

    Article  Google Scholar 

  67. Wu, H. et al. Detection of differentially methylated regions from whole-genome bisulfite sequencing data without replicates. Nucleic Acids Res. 43, e141 (2015).

    Google Scholar 

  68. Liao, Y., Wang, J., Jaehnig, E. J., Shi, Z. & Zhang, B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res. 47, W199–W205 (2019).

    Article  CAS  Google Scholar 

  69. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful for the insightful discussions, helpful suggestions, and editing of the manuscript by E. Oltz at the College of Medicine. We would like to thank P. Collins, J. Lio and A. Zayed at the Ohio State University for their expert assistance. We thank the human cord blood donors and the Leukemia Tissue Bank at the Ohio State University Comprehensive Cancer Center, Columbus, OH, for blood sample collection. We would also like to thank A. Wetzel and the team of the IGM Genomic Services Lab of the Research Institute at Nationwide Children’s Hospital, Columbus, OH, for their help with genomic sequencing. We thank the NIH Tetramer Facility at Emory University in Atlanta, GA, for providing the monomers used in this study for the detection of LCMV-specific tetramer+ CD8+ T cells. Research reported in this publication was funded by the Ohio State University Comprehensive Cancer Center and College of Medicine.

Author information

Authors and Affiliations

Authors

Contributions

H.E.G. conceived and designed the study and developed methodologies. A.A.S., A.Y., R.S., B.L., M.R. and Y.-L.C. performed in vitro experiments. A.A.S., A.Y., N.O., P.B., B.L., R.S. and H.E.G. performed in vivo infection and tumor experiments. H.E.G., A.W. and A.Y. participated in bioinformatics data analysis and interpretation. B.L. and A.Y. participated in library preparation for NGS sequencing. H.E.G., A.A.S. and A.Y. drafted the manuscript. All authors contributed to data analysis, figure preparation and manuscript editing.

Corresponding author

Correspondence to Hazem E. Ghoneim.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Maike Hofmann and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: N. 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 TGFβ1 signaling accelerates dysfunction in chronically stimulated human CD8+ T cells.

(a) Gating strategy showing the phenotype of freshly isolated human CBMCs before ex vivo stimulation. (b) Summary bar graphs showing MFI of IFNG, (c) TNFα, (d) CD107a, (e) T-bet, and (f) TCF-1 for ‘Weak’ TCR stimulated IFNG + CD8+ T cells after in vitro PMA/Ionomycin stimulation on day 28. (g) Longitudinal tracking of CD8+ T cell numbers per 100 ul (left y-axis, solid lines) and frequency of dead CD8+ T cells (right y-axis, red dotted lines) under ‘Strong’ TCR stimulation from day 0–28. (h) Bar graphs showing frequency of dead CD8+ T cells on day 28, or MFI of (i) CD39, (j) LAG3, and (k) CD103 on CD8+ T cells under ‘Strong’ TCR and TGFβ1 after PMA/Ionomycin stimulation on day 28. (l) Schematic of human adult naïve CD8+ T cell isolation followed by acute or chronic stimulation for 28 days. (m) Bar graphs showing frequency of IFNG + CD107a+, (n) IFNG + TNFα + adult CD8+ T cells, (o) MFI of CD107a on IFNG + CD8+ T cells, and (p) MFI of Perforin, (q) CD101, (r) CD103, (s) CD39, or (t) PD-1 on adult CD8+ T cells after PMA/Ionomycin stimulation on day 28. (u) Bar graphs showing MFI of CD103 and (v) frequencies of IFNG + TNFa+ CD8+ T cells in adult polyclonal effector memory CD8+ T cells (TEM) after PMA/Ionomycin stimulation on day 28. All n = 3–4 biological replicates per group, representative of 2–3 independent experiments. * P-value < 0.05, ** p < 0.01, *** p < 0.001; comparisons were made using the Mann-Whitney U test (unpaired, two-sided)-(panels b-f), or Adjusted P-value ** p < 0.01, *** p < 0.001, **** p < 0.0001 using one-way ANOVA with Tukey’s multiple comparison (panels h-k) as indicated, or one-way ANOVA with Bonferroni’s multiple comparisons relative to Ch.TCR group (panels m-v). Error bars = mean ± SEM.

Source data

Extended Data Fig. 2 Chronic TGFβ1 and TCR signals establish a stable dysfunctional program in CD8+ T cells.

(a) Summary bar graphs showing MFI of IFNG, (b) T-bet, (c) TNFα, and (d) CD107a in IFNG + CD8+ T cells after 7 days of resting from chronic or acute stimulation (day 35) as show in Fig. 2a. (e) Bar graphs showing MFI of TNFα, and (f) CD107a in IFNG + CD8+ T cells after 14 days of rest (say 42). (g) Bar graphs showing MFI of GZMB, and (h) CD11a in CD8+ T cells after 14 days of rest (Day 42). N = 3 biological replicates per group, representative of 2–3 independent experiments. Adjusted P-value ** p < 0.01, *** p < 0.001, **** p < 0.0001 comparisons were made using one-way ANOVA with Bonferroni’s multiple comparisons relative to Dysf. group. Error bars = mean ± SEM.

Source data

Extended Data Fig. 3 Boosting BMP while blocking TGFβ1 recovers effector function in dysfunctional CD8+ T cells.

(a) IGV Snapshots showing: DNA methylation levels (top panel) at individual CpG sites within mouse naïve (black), antigen-specific exhausted WT (red) or Dnmt3a-deficient (green) CD8+ T cells on day 35 post-chronic LCMV infection. Blue-to-red ratio indicates % of unmethylated versus methylated reads (GSE99450); open chromatin peaks (middle panel) detected in naïve (black), progenitor (PD-1+ Tim-3-; green), or terminally exhausted (PD-1+ Tim-3 + ; red) CD8+ T cells from chronically infected mice (PRJNA546023); and levels of H3K27 acetylation marks (bottom panel) in the progenitor (green), cytolytic (blue; Cx3cr1+PD-1+), and terminally exhausted (Cx3cr1- PD-1+ Tim-3+) CD8 T cells during chronic LCMV infection (GSE149810) at the Smad1, Smad5, and Tgfbr3 loci. (b) Heatmap of RNA levels in exhausted T cell subsets during chronic LCMV infection (GSE84105). (c) tSNE plot visualization of human CD8+ T cells for the described conditions in Fig. 4a after PMA/ionomycin stimulation on day 28, and representative plots showing protein expression levels of IFNG, TNFα, CD107a, T-bet, and CD103 in PMA/Ionomycin-stimulated CD8+ T cells on day 28 (n = 4 wells per condition). (d) Bar graphs showing expression levels of TOX in adult blood-derived CD8+ T cells on day 21 of chronic ‘Strong’ or (e) ‘Weak’ TCR stimulation. (f) Bar graphs showing MFI of IL-7R and (g) CD103 in adult CD8+ T cells on day 21 of ‘Weak’ TCR stimulation. For panels d-g, n = 3–4 biological replicates per group per experiment, data pooled from 2 independent experiments. Adjusted P-value ** p < 0.01, *** p < 0.001, **** p < 0.0001 comparisons were made using one-way ANOVA with Tukey’s multiple comparisons as indicated (panels d,f) or one-way ANOVA with Bonferroni’s multiple comparisons relative to Dysf. group (panels e,g). Error bars = mean ± SEM.

Source data

Extended Data Fig. 4 Treated CD8+ T cells maintain a heritable functional memory-like program.

(a) Representative FACS plots showing the gating strategy for sorting dysfunctional human CD8+ T cells on day 14 after chronic TCR plus TGFβ1 stimulation, and day 28 expression of IFNG and CD103 in PMA/ionomycin-stimulated CD8+ T cells after treatment. (b) Summary bar graphs showing frequency of IFNG + TNFα + CD8+ T cells, and (c) MFI of CD101 in sorted CD8+ T cells after PMA/ionomycin stimulation on day 28. (d) Representative FACS plots showing CD8+ T cell divisions and expression levels of IL-7R in CFSE-labeled CD8+ T cells from each condition as described in Fig. 4a on day 35 (after 7 day-rest under homeostatic conditions). (e) MFI of IL-7R in divided CD8+ T cells on day 35. (f) Representative FACS plots showing CD8+ T cell divisions and expression levels of CD103 in CFSE-labeled CD8+ T cells on day 35. (g) MFI of CD103 in divided CD8+ T cells on day 35. (h) Bar graphs showing frequencies of divided CD8+ T cells (≥3 proliferation cycles), and (i) MFI of IL-7R and (j) CD103 in CD8+ T cells on day 35. (k) Bar graphs showing MFI of TNFα and (l) Perforin in IFNG + CD8+ T cells after PMA/Ionomycin stimulation on day 35. N = 3 (panels a-c) or 4 (panels d-l) biological replicates per group, representative of 2–3 independent experiments. Adjusted P-value ** p < 0.01, *** p < 0.001, **** p < 0.0001 comparisons were made using one-way ANOVA with Tukey’s multiple comparisons as indicated (panels b, c, h–l) or one-way ANOVA with Bonferroni’s multiple comparisons relative to Dysf. group (panels e, g). Error bars = mean ± SEM.

Source data

Extended Data Fig. 5 Transcriptional re-wiring of dysfunctional human CD8+ T cells.

(a) Volcano plot showing DEGs in acute TCR stimulated versus naïve human CD8+ T cells, or (c) in chronic TCR stimulated versus Dysf. human CD8+ T cells. (b) Gene set enrichment analysis for Ac.TCR versus Naïve CD8+ T cells or (d) Ac.TCR versus Dysf. CD8+ T cells compared against common upregulated genes in human CD8+ T cell subsets following yellow fever vaccination (GSE100745) (e) Heatmap of DEGs between each condition based on relative Z-score and organized into clusters: C1 (yellow; up in Naïve and Dysf.), C2 (orange; up in Dysf. only), C3 (blue; up in Ch.TCR and Ac.TCR), C4 (green; up in Treated.I-III), C5 (purple; up in Ch.TCR, Ac.TCR, and Treated.I-III), and C6 (pink; up in Naïve, Ch.TCR, Ac.TCR, and Treated.I-III). Example genes with significant fold change in expression (fold change >2, p < 0.05) are listed per cluster. N = 2 biological replicates per condition. Panels (5a,5c,5e) were statistically analyzed by DESeq2 from Partek. Panels (5b and 5d) were statistically analyzed by GSEA using Partek.

Extended Data Fig. 6 BMP4 agonist treatment promotes transcriptional recovery of effector and memory programs.

(a) Venn diagram of overlapping genes that are downregulated in Dysf. versus Treated.I-III CD8+ T cells on day 28. (b) Pathway enrichment analysis of genes that are upregulated in Treated.II versus Dysf. CD8+ T cells or overlap with the upregulated genes in Treated.III versus Dysf. CD8+ T cells (NCI-Nature 2016 pathway database). Significance was determined by Enrichr62. (c) Venn diagram of overlapping genes upregulated in Dysf. versus Treated.I-III CD8+ T cells. (d) Gene set enrichment analysis of biological pathways linked to the upregulated genes in Dysf. versus Treated.I-III CD8+ T cells using Hallmark gene signature database. (e) Interaction plots from RNA-seq data showing average expression levels of KLF3, (f) S1PR1, (g) GZMK, (h) FCGR3A, (i) TGFBR3, (j) CX3CR1, (k) PRF1, (l) CD28, (m) CD109, (n) MYO7A, (o) ITGAE, and (p) SMAD6 transcripts among chronically (Ch.TCR, Dysf., Treated.I-III) or acutely (Ac.TCR) stimulated CD8+ T cells on day 28.

Source data

Extended Data Fig. 7 TGFβ1 exposure drives transcriptional similarity to exhausted CD8+ T cells in human cancer.

(a) UMAP visualization of CD8+ T cell meta-clusters identified in single-cell RNA-seq analysis of human CD8+ T cell subsets including tumor-infiltrating lymphocytes (TILs) from 21 types of human cancer (GSE156728)45 (b, c) UMAP visualization showing RNA expression levels of some selected signature genes in human TILs and memory CD8+ T cell clusters that overlap with transcriptional features of in vitro-differentiated Dysfunctional and Treated human CD8+ T cells.

Extended Data Fig. 8 BMP signals maintain effector and memory programs in human CD8+ T cells.

(a) Bar graph showing numbers of methylated (top) or demethylated (bottom) DMRs in Dysf. CD8+ T cells relative to all other conditions on day 28. (b) Venn diagrams showing transcription factor (TF)-binding motifs enriched in hypermethylated or (c) hypomethylated genomic regions in in Dysf. versus Treated.I, II, or III human CD8+ T cells on day 28. (d) Snapshots from scATAC-seq database (GSE129785) showing chromatin accessibility changes at selected loci of effector and memory, or (e) dysfunctional programs in human CD8+ T cell subsets from patients with cancer49.

Extended Data Fig. 9 Treatment of dysfunctional CD8+ T cells improves cytotoxicity against tumor cell lines.

(a) Representative FACS plots showing E-cadherin and PD-L1 expression on THP-1 (AML) and MDA-MB-231 (breast adenocarcinoma) tumor cells. (b) Summary bar graphs showing frequencies of PD-1 + LAG3+, (c) total LAG3 + CD8+ T cells, or (d) MFI of PD-1 and (e) GZMB on CD8+ T cells following 18-hour co-culture with AML cells on day 28. (f) Bar graph showing MFI of CD103 on CD8+ T cells following co-culture with MDA-MB-231 cells at day 28. (g) Bar graph showing frequency of LAG3 + CD101 + CD8+ T cells under ‘Strong’ TCR stimulation following co-culture with AML cells at day 28. (h) Summary of dead AML cells per 100 viable CD8+ T cells under ‘Strong’ TCR following co-culture with AML cells at varying T cell: tumor cell ratios on day 28. (i) Bar graph of dead AML cells per 100 viable CD8+ T cells after rest from ‘Strong’ TCR under homeostatic conditions, following co-culture with AML cells on day 35. (j) MFI of LAG3 in CD8+ T cells after PMA/Ionomycin stimulation on day 35. For panels b–f, n = 3–4 biological replicates per group per experiment, with data pooled from 2 independent experiments. For panels g–j, n = 4 biological replicates per group, representative of 2–3 independent experiments. Adjusted P-value ** p < 0.01, *** p < 0.001, **** p < 0.0001; comparisons were made using one-way ANOVA with Tukey’s multiple comparison (panels b, d, j, k) as indicated, one-way ANOVA with Bonferroni’s multiple comparisons relative to Dysf. group (panels c, e–g, i), or two-way ANOVA with Tukey’s multiple comparisons (panel h). Error bars = mean ± SEM.

Source data

Extended Data Fig. 10 Combined TGFβR1i and BMP4 agonist enhances ICB response during chronic LCMV.

(a) Summary bar graphs showing numbers of polyclonal LCMV-specific (CD44hi PD-1+), (b) proliferating (Ki67+) CD44hi PD-1+, (c) Cytolytic (CD44hi Cx3cr1+ PD-1+) and (d) Terminally Exhausted (Tim3+ PD-1 + Cx3cr1-) CD8+ T cells in spleen on day 33 following primary treatment using RepSox, BMP4a, or combined treatment as described in Fig. 8a. (e) Summary dot plot of LCMV viral titers (PFU per ml) in serum from chronically LCMV-infected mice on day 33 following primary treatment. (f) Summary bar graphs showing numbers of proliferating (Ki67 + CD44hi PD-1+) CD8+ T cells, and (g) proliferating P14 (Ki67 + Thy1.1 + GP33-tetramer+) CD8+ T cells in spleen following anti-PD-L1 treatment. All N = 4–6 mice per treatment group per experiment, with data pooled from 2–3 independent experiments. Adjusted P-value ** p < 0.01, *** p < 0.001, **** p < 0.0001; comparisons were made using one-way ANOVA with Tukey’s multiple comparison (panels a–d), or Kruskal-Wallis Test with Dunn’s multiple comparisons (panels f, g) as indicated. Error bars = mean ± SEM.

Source data

Supplementary information

Supplementary Information

Supplementary data for CD103 MFI on P14 cells from Fig. 7n (raw flow histograms and data/statistics).

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1: IPA datasets used in Fig. 1i. Supplementary Table 2: RNA-seq normalized counts. Supplementary Table 3: Differentially expressed genes from DESEq2 analysis of RNA-seq data. Supplementary Table 4: DMRs from WGBS data

Source data

Source Data Fig. 1

Raw and statistical source data.

Source Data Fig. 2

Raw and statistical source data.

Source Data Fig. 3

Raw and statistical source data.

Source Data Fig. 4

Raw and statistical source data.

Source Data Fig. 5

Raw and statistical source data.

Source Data Fig. 6

Raw and statistical source data.

Source Data Fig. 7

Raw and statistical source data.

Source Data Fig. 8

Raw and statistical source data.

Source Data Extended Data Fig. 1

Raw and statistical source data.

Source Data Extended Data Fig. 2

Raw and statistical source data.

Source Data Extended Data Fig. 3

Raw and statistical source data.

Source Data Extended Data Fig. 4

Raw and statistical source data.

Source Data Extended Data Fig. 6

Raw and statistical source data.

Source Data Extended Data Fig. 9

Raw and statistical source data.

Source Data Extended Data Fig. 10

Raw and 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

Saadey, A.A., Yousif, A., Osborne, N. et al. Rebalancing TGFβ1/BMP signals in exhausted T cells unlocks responsiveness to immune checkpoint blockade therapy. Nat Immunol 24, 280–294 (2023). https://doi.org/10.1038/s41590-022-01384-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01384-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