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:

Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen

Abstract

The majority of tumor-infiltrating T cells exhibit a terminally exhausted phenotype, marked by a loss of self-renewal capacity. How repetitive antigenic stimulation impairs T cell self-renewal remains poorly defined. Here, we show that persistent antigenic stimulation impaired ADP-coupled oxidative phosphorylation. The resultant bioenergetic compromise blocked proliferation by limiting nucleotide triphosphate synthesis. Inhibition of mitochondrial oxidative phosphorylation in activated T cells was sufficient to suppress proliferation and upregulate genes linked to T cell exhaustion. Conversely, prevention of mitochondrial oxidative stress during chronic T cell stimulation allowed sustained T cell proliferation and induced genes associated with stem-like progenitor T cells. As a result, antioxidant treatment enhanced the anti-tumor efficacy of chronically stimulated T cells. These data reveal that loss of ATP production through oxidative phosphorylation limits T cell proliferation and effector function during chronic antigenic stimulation. Furthermore, treatments that maintain redox balance promote T cell self-renewal and enhance anti-tumor immunity.

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: Aerobic glycolysis is a hallmark of terminally exhausted T cells.
Fig. 2: Chronic antigen stimulation induces mitochondrial dysfunction and limits nucleotide biosynthesis.
Fig. 3: Inhibition of mitochondrial electron transport limits T cell proliferation.
Fig. 4: Endogenous antioxidants are required for T cell proliferation.
Fig. 5: Antioxidants reverse metabolic T cell dysfunction.
Fig. 6: Antioxidants restore the proliferation and self-renewal of chronically stimulated T cells.
Fig. 7: Antioxidants reverse chronic stimulation-driven loss of T cell effector function.

Similar content being viewed by others

Data availability

Datasets are deposited in the NCBI Gene Expression Omnibus using the following accession code: RNA-seq, GSE138459. Additional information can be found in the Nature Research Reporting Summary. Further information and requests for reagents may be directed to, and will be fulfilled by, the corresponding author. Source data are provided with this paper.

References

  1. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  2. Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. 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.e10 (2019).

    CAS  PubMed  Google Scholar 

  5. Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1CD8+ tumor-infiltrating T cells. Immunity 50, 181–194.e6 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Frauwirth, K. A. et al. The CD28 signaling pathway regulates glucose metabolism. Immunity 16, 769–777 (2002).

    CAS  PubMed  Google Scholar 

  10. Parry, R. V. et al. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell Biol. 25, 9543–9553 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Scharping, N. E. et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity 45, 374–388 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Siska, P. J. et al. Mitochondrial dysregulation and glycolytic insufficiency functionally impair CD8 T cells infiltrating human renal cell carcinoma. JCI Insight 2, e93411 (2017).

    PubMed Central  Google Scholar 

  13. Zhang, Y. et al. Enhancing CD8+ T cell fatty acid catabolism within a metabolically challenging tumor microenvironment increases the efficacy of melanoma immunotherapy. Cancer Cell 32, 377–391.e9 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Macian, F. et al. Transcriptional mechanisms underlying lymphocyte tolerance. Cell 109, 719–731 (2002).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  16. 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 

  17. Warburg, O. On the origin of cancer cells. Science 123, 309–314 (1956).

    CAS  PubMed  Google Scholar 

  18. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Google Scholar 

  19. Yost, K. E. et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 25, 1251–1259 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Yao, C. et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 20, 890–901 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Alfei, F. et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 571, 265–269 (2019).

    CAS  PubMed  Google Scholar 

  23. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Sena, L. A. et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 38, 225–236 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Cardenas, C. et al. Essential regulation of cell bioenergetics by constitutive InsP3 receptor Ca2+ transfer to mitochondria. Cell 142, 270–283 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Loffler, M., Jockel, J., Schuster, G. & Becker, C. Dihydroorotat-ubiquinone oxidoreductase links mitochondria in the biosynthesis of pyrimidine nucleotides. Mol. Cell Biochem. 174, 125–129 (1997).

    CAS  PubMed  Google Scholar 

  27. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Bauer, D. E., Hatzivassiliou, G., Zhao, F., Andreadis, C. & Thompson, C. B. ATP citrate lyase is an important component of cell growth and transformation. Oncogene 24, 6314–6322 (2005).

    CAS  PubMed  Google Scholar 

  29. Fraietta, J. A. et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 24, 563–571 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Titov, D. V. et al. Complementation of mitochondrial electron transport chain by manipulation of the NAD+/NADH ratio. Science 352, 231–235 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Flint, D. H., Tuminello, J. F. & Emptage, M. H. The inactivation of Fe-S cluster containing hydro-lyases by superoxide. J. Biol. Chem. 268, 22369–22376 (1993).

    CAS  PubMed  Google Scholar 

  32. Chandel, N. S. et al. Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc. Natl Acad. Sci. USA 95, 11715–11720 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Chang, C. H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Bevan, M. J., Epstein, R. & Cohn, M. The effect of 2-mercaptoethanol on murine mixed lymphocyte cultures. J. Exp. Med. 139, 1025–1030 (1974).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Mak, T. W. et al. Glutathione primes T cell metabolism for inflammation. Immunity 46, 675–689 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165–1169 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Shin, H. et al. A role for the transcriptional repressor Blimp-1 in CD8+ T cell exhaustion during chronic viral infection. Immunity 31, 309–320 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Rutishauser, R. L. et al. Transcriptional repressor Blimp-1 promotes CD8+ T cell terminal differentiation and represses the acquisition of central memory T cell properties. Immunity 31, 296–308 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Gautam, S. et al. The transcription factor c-Myb regulates CD8+ T cell stemness and antitumor immunity. Nat. Immunol. 20, 337–349 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bengsch, B. et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8+ T cell exhaustion. Immunity 45, 358–373 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Thommen, D. S. et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat. Med. 24, 994–1004 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Schietinger, A. et al. Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early during tumorigenesis. Immunity 45, 389–401 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Taylor, A., Rothstein, D. & Rudd, C. E. Small-molecule inhibition of PD-1 transcription is an effective alternative to antibody blockade in cancer therapy. Cancer Res. 78, 706–717 (2018).

    CAS  PubMed  Google Scholar 

  47. Ghosh, A. et al. Donor CD19 CAR T cells exert potent graft-versus-lymphoma activity with diminished graft-versus-host activity. Nat. Med. 23, 242–249 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Pilon-Thomas, S., Mackay, A., Vohra, N. & Mule, J. J. Blockade of programmed death ligand 1 enhances the therapeutic efficacy of combination immunotherapy against melanoma. J. Immunol. 184, 3442–3449 (2010).

    CAS  PubMed  Google Scholar 

  49. Liang, Y. et al. Targeting IFNɑ to tumor by anti-PD-L1 creates feedforward antitumor responses to overcome checkpoint blockade resistance. Nat. Commun. 9, 4586 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Naviaux, R. K., Costanzi, E., Haas, M. & Verma, I. M. The pCL vector system: rapid production of helper-free, high-titer, recombinant retroviruses. J. Virol. 70, 5701–5705 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Anders, S. et al. Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat. Protoc. 8, 1765–1786 (2013).

    PubMed  Google Scholar 

  53. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Liberzon, A. et al. The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Martindale, J. L. & Holbrook, N. J. Cellular response to oxidative stress: signaling for suicide and survival. J. Cell Physiol. 192, 1–15 (2002).

    CAS  PubMed  Google Scholar 

  57. Schaefer, C. F. et al. PID: the Pathway Interaction Database. Nucleic Acids Res. 37, D674–D679 (2009).

    CAS  PubMed  Google Scholar 

  58. van der Windt, G. J., Chang, C. H. & Pearce, E. L. Measuring bioenergetics in T cells using a Seahorse Extracellular Flux Analyzer. Curr. Protoc. Immunol. 113, 3.16B.1–3.16B.14 (2016).

    Google Scholar 

  59. Vardhana, S. A. et al. Glutamine independence is a selectable feature of pluripotent stem cells. Nat. Metab. 1, 676–687 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Millard, P., Letisse, F., Sokol, S. & Portais, J. C. IsoCor: correcting MS data in isotope labeling experiments. Bioinformatics 28, 1294–1296 (2012).

    CAS  PubMed  Google Scholar 

  61. Schworer, S. et al. Proline biosynthesis is a vent for TGFβ-induced mitochondrial redox stress. EMBO J. 39, e103334 (2020).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Thompson and Finley laboratories for discussion and critical feedback. S.A.V. is a Senior Fellow with the Parker Institute of Cancer Immunotherapy and is supported by a Burroughs Wellcome Fund Career Award for Medical Scientists. A.T.S. was supported by a Bridge Scholar Award from the Parker Institute for Cancer Immunotherapy and a Career Award for Medical Scientists from the Burroughs Wellcome Fund. This work was additionally supported by the Memorial Sloan Kettering Cancer Center Support Grant no. P30 CA008748 and R25 Training Grant no. AI140472-01A1.

Author information

Authors and Affiliations

Authors

Contributions

S.A.V. and C.B.T. conceived the study. S.A.V. performed all experiments with assistance from M.A.H. M.B. and J.R.C. assisted with LC–MS, extracellular flux and nutrient consumption experiments. D.K.W., B.K., A.T.S., H.Y.C. and K.E.Y. assisted with analysis of RNA-seq data. M.S., P.S.H. and M.R.M.v.d.B. assisted with CAR-T cell experiments. C.B.T. provided additional work in conception and study guidance. S.A.V. and C.B.T. wrote the manuscript.

Corresponding author

Correspondence to Craig B. Thompson.

Ethics declarations

Competing interests

C.B.T. is a founder of Agios Pharmaceuticals and a member of its scientific advisory board. He is also a former member of the Board of Directors and a stockholder of Merck and Charles River Laboratories. S.A.V. has received honoraria from Agios Pharmaceuticals and Rheos Pharmaceuticals, is an advisor for Immunai and has consulted for ADC Therapeutics. A.T.S. and D.K.W. are scientific founders and equity holders of, and receive consulting fees from, Immunai. A.T.S. received funding support from 10x Genomics and Arsenal Biosciences. K.E.Y. is an advisor for Immunai. H.Y.C. is a co-founder of Accent Therapeutics and Boundless Bio and is a consultant for 10x Genomics, Arsenal Biosciences and Spring Discovery.

Additional information

Peer review information Peer reviewer reports are available. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Chronic T cell stimulation induces T cell exhaustion.

All experimental analyses were conducted eight days after initial stimulation unless otherwise specified. ac, Expression of inhibitory immunoreceptors (PD-1, LAG-3, PD-L1) and intracellular cytokine production (IFN-γ and TNF) in acutely and chronically stimulated T cells following re-stimulation with PMA and ionomycin. d, Expression of Glut1 in acutely or chronically stimulated OT-I T cells with or without restimulation using bead-bound anti-CD3. Actin is used as a loading control. Experiment was repeated three times with similar results. Uncropped blot can be found within Source Data. e, Gene set enrichment plot showing that genes associated with chronically stimulated polyclonal T cells in vitro are enriched for genes upregulated in exhausted CD8 + T cells (Texh) but not anergic T cells15. f, Killing of peptide-pulsed B16 cells. Luciferase-expressing B16 cells pulsed with Ova peptide at the indicated doses for 4 h were co-cultured with acutely or chronically stimulated T cells for 24 h. The following day, cells were lysed and luciferase expression was assessed using a luminometer. g, Normalized isotopologue abundance of intracellular lactate in acutely and chronically stimulated T cells following 6 h of re-stimulation by plate-bound anti-CD3 in the presence of U-13C-Glucose. Abundance was normalized to cell number at the time of harvest. h, Median lactate excreted per molecule of glucose consumed in acutely and chronically stimulated T cells following initial stimulation. P values were calculated by unpaired, two-sided Student’s t-test (fh), relative to acutely stimulated T cells or based on 1,000 permutations by the GSEA algorithm and not adjusted for multiple comparisons (e). Data are presented as the mean ± s.d. of n = 3 biologically independent samples from a representative experiment. **P < 0.01.

Source data

Extended Data Fig. 2 Aerobic glycolysis is a hallmark of chronic stimulation-dependent terminal T cell dysfunction.

a, Extracellular acidification rate of acutely and chronically stimulated polyclonal T cells in media containing or lacking glucose as indicated. b, Extracellular acidification rate of acutely and chronically stimulated polyclonal T cells at baseline and in response to electron transport chain inhibition. c, Population doublings of acutely and chronically stimulated polyclonal CD8 + T cells following initial stimulation. d, Viability of acutely and chronically stimulated T cells as determined by forward scatter and DAPI exclusion. e, Intracellular TOX expression and proliferation as measured by dilution of Cell Trace Violet fluorescence of acutely or chronically stimulated T cells. f, Normalized expression of glycolytic genes in CD8 + T cell clusters from patients with basal and squamous cell carcinoma treated with immune checkpoint blockade19. g, Gene set enrichment plot showing that genes associated with terminally exhausted T cells isolated from murine B16 melanoma tumors8 are enriched for glycolytic genes. h, Correlation of glycolysis score (left) and TCA cycle score (right) with TCF7 expression in exhausted CD8+ T cell clusters from basal and squamous cell carcinoma patients treated with immune checkpoint inhibitors6. i, Gene set enrichment plot showing that chronically stimulated OT-I T cells in vitro significantly downregulate genes upregulated in progenitor Texh as compared to terminal Texh8. j, k, Intracellular cytokine production in acutely and chronically stimulated polyclonal T cells following re-stimulation. In (j), cells were cultured in the presence or absence of anti-PD-L1 (10 F.9G2) from D2-D8. In (k), “Chronic + 24 h rest” cells were rested in the absence of plate-bound anti-CD3 for 24 h prior to re-stimulation. Experiment was repeated three times with similar results. P values were calculated by unpaired, two-sided Student’s t-test (ac) relative to acutely stimulated T cells or based on 1,000 permutations by the GSEA algorithm and not adjusted for multiple comparisons (fi). Data are presented as the mean ± s.d. of n = 3 biologically independent samples from a representative experiment. ****P < 0.0001.

Extended Data Fig. 3 Chronic antigen stimulation impairs mitochondrial oxidation and ATP production.

a, Quantification of relative tricarboxylic acid cycle metabolite pool sizes in acutely and chronically stimulated T cells. Columns represent biological replicates for each condition. b, Oxygen consumption rate (OCR) of acutely or chronically stimulated T cells at baseline or in the presence of ATP synthase inhibition (Oligo), uncoupling agents (FCCP), inhibition of glucose uptake (2-DG), and complex III/IV inhibition (Rot/AA). c, Schematic depicting how oxidative metabolism of uniformly-labeled palmitate ([U-13C] palmitate) generates metabolites associated with the TCA cycle. Colored circles represent 13C-labeled carbons. d, Fractional labeling by [U-13C] palmitate of citrate, glutamate, fumarate, malate and aspartate in acutely and chronically stimulated T cells following re-stimulation. e, Proliferation of T cells acutely or chronically stimulated in the presence or absence of supplemental sodium acetate (5 mM), as measured by dilution of Cell Trace Violet fluorescence. f, Quantification of pool sizes of metabolite intermediates in nucleotide synthesis in acutely and chronically stimulated T cells. Heatmap depicts pool size relative to row median. Columns represent biological replicates for each condition. Experiment was repeated two times with similar results. P values were calculated by unpaired, two-sided Student’s t-test (a,b,d). Data are presented as the mean ± s.d. of n = 3 biologically independent samples from a representative experiment. **P < 0.01. ****P < 0.0001.

Extended Data Fig. 4 Oxidative stress limits T cell proliferative capacity.

a, Western blot depicting overexpression of FLAG-tagged recombinant NADH oxidase enzymes LbNOX and MitoLbNOX in T cells30. Experiment was repeated three times with similar results. Uncropped blot can be found within Source Data. b, Fluorescence intensity of acutely and chronically stimulated T cells expressing vector control, LbNOX, or MitoLbNOX following eight days in culture after loading with CM-H2DCFDA. c, Population doublings of acutely and chronically stimulated T cells expressing vector control, LbNOX, or MitoLbNOX. d, Fluorescence intensity of acutely and chronically stimulated T cells after loading with BODIPY-C11 to measure lipid peroxidation. Light-grey-shaded peak represents negative control. e, Fluorescence intensity of acutely or chronically stimulated T cells cultured with or without pharmacologic agents that impair ETC function following 2 days of initial stimulation. Cells were loaded with CM-H2DCFDA to measure ROS on D8 following initial stimulation. f, qRT-PCR of Myb and Tcf7 in acutely or chronically stimulated T cells with or without the addition of the indicated agents for 6 days following 2 days of primary stimulation. gi, Expression of oxidative stress-related metabolic genes (“ROS score”) in tumor-infiltrating CD8 + T cells from basal and squamous cell carcinoma patients treated with immune checkpoint inhibitors19. In (g), ROS score in independent CD8 + T cell clusters is shown. In (h), ROS score in exhausted and memory T cell populations is shown according to clone size as measured by TCR sequencing; box center line=median, box limits=upper and lower quartiles, box whiskers=1.58 x interquartile range. In (i), correlation of ROS score with TCF7 expression in exhausted CD8 + T cells is shown. Only cells with non-zero TCF7 expression were included. P values were calculated by one-way ANOVA with Sidak’s multiple comparisons post-test (g, i), or one-sided Student’s t-test relative to base mean (g, h). Data are presented as the mean ± s.d. of n = 3 biologically independent samples from a representative experiment. **P < 0.01. ***P < 0.001. ****P < 0.0001.

Source data

Extended Data Fig. 5 Endogenous anti-oxidant production is limiting for T cell proliferation.

a, Motif analysis of sites with increased accessibility in tumor-infiltrating CD8 + T cells (L7) as compared to T cells from Listeria-infected mice (E7) showing NFATc1 as among the motifs whose accessibility was most preferentially increased in L7 cells15. b, Intracellular calcium flux as measured by ratio of bound to unbound Indo-1-AM in acutely and chronically stimulated T cells, at baseline, in response to monomeric anti-CD3, and in response to receptor clustering (streptavidin). c, Gene set enrichment plot showing that chronically stimulated OT-I T cells are enriched for NFAT target genes. d, Expression of NFAT target genes (“nfat score”) in independent CD8 + T cell clusters. e, Correlation of expression of NFAT target genes (“nfat score”) with expression of oxidative stress-related metabolic genes (“ROS score”) in tumor-infiltrating CD8 + T cells from melanoma patients treated with immune checkpoint inhibitors. f, Fluorescence intensity of acutely and chronically stimulated T cells cultured with or without βME supplementation after loading with CM-H2DCFDA to measure ROS. Light-grey-shaded peak represents negative control. g, Proliferation of T cells acutely stimulated in the presence or absence of BSO or diamide as measured by dilution of Cell Trace Violet fluorescence. h, Expression of TCF-1 and TOX in chronically stimulated T cells cultured in the presence or absence of BSO. P values were calculated by one-sided Student’s t-test relative to base mean (d, e). ****P < 0.0001.

Extended Data Fig. 6 N-acetylcysteine reverses oxidative stress in chronically stimulated T cells.

a, Quantification of relative metabolite pool sizes as measured by LC-MS in chronically stimulated T cells cultured with or without N-AC. Colored dots represent intermediates in glutathione synthesis as indicated. Dashed lines represent cutoffs of p < 0.01 and log2 fold change > 0.5. b, ATP production by acutely or chronically stimulated T cells cultured with or without N-AC. P values were calculated by one-way ANOVA with Sidak’s multiple comparisons post-test compared to acutely stimulated T cells (b). Data are presented as the mean ± s.d. of n = 4 biologically independent samples from a representative experiment (b). *P < 0.05.

Extended Data Fig. 7 Antioxidants restore T cell self-renewal during chronic stimulation.

a, Population doublings of chronically stimulated T cells with or without N-AC supplementation under normoxic (left) or hypoxic (right) conditions. Experiment was repeated two times with similar results. b, qRT-PCR of Tcf, Myb, and Prdm1 in acutely or chronically stimulated T cells with or without the addition of N-AC as indicated. c, Intracellular calcium flux as measured by ratio of bound to unbound Indo-1-AM in acutely and chronically stimulated T cells cultured with or without N-AC. d, Gene set enrichment plot showing that the addition of N-AC during chronic stimulation reduces expression of NFAT target genes. P values were calculated by unpaired, two-sided Student’s t-test relative to cells cultured without N-AC (a), one-way ANOVA with Sidak’s multiple comparisons post-test (b) or based on 1,000 permutations by the GSEA algorithm and not adjusted for multiple comparisons (d). Data are presented as the mean ± s.d. of n = 3 biologically independent samples from a representative experiment. *P < 0.05. **P < 0.01. ***P < 0.0001.

Extended Data Fig. 8 Antioxidants reverse endogenous tumor-associated T cell dysfunction.

a, Production of IFN-γ and TNF following re-stimulation with PMA and ionomycin in chronically stimulated T cells with or without N-AC supplementation under normoxic or hypoxic conditions. Experiment was repeated two times with similar results. b, c, Oxygen consumption rate (OCR) of OT-I T cells chronically co-cultured with B16 melanoma cells with or without anti-PD-L1 antibodies and with or without N-AC supplementation at baseline or in the presence of ATP synthase inhibition (Oligo), uncoupling agents (FCCP), or complex III/IV inhibition (Rot/AA). d, Production of IFN-γ and TNF following re-stimulation with PMA and ionomycin in chronically stimulated T cells with or without MitoTEMPO or Trolox supplementation as indicated. Experiment was repeated two times with similar results. P values were calculated by unpaired, two-sided Student’s t-test (c). Data are presented as the mean ± s.d. of n = 4 biologically independent samples from a representative experiment (b, c). *P < 0.05.

Extended Data Fig. 9 Gating strategy for fluorescence activated cell sorting analysis.

For both polyclonal and OT-I transgenic T cells, gating was perform as shown. First, doublet exclusion was performed on cells gated by FSC-H versus FSC-W. Then, doublet exclusion was performed on cells gated by SSC-H versus SSC-W. Viable cells were identified by FSC-A and Live/Dead Blue exclusion. Finally, CD8 positivity was assessed by fluorescence in the BV-786 channel.

Supplementary information

Source data

Source Data Fig. 3

Images depict uncropped images for western blots in Fig. 3b. Red boxes contain cropped area shown in main figure.

Source Data Extended Data Fig. 1

Images depict uncropped images for western blots in Extended Data Fig. 1d. Red boxes contain cropped area shown in main figure.

Source Data Extended Data Fig. 4

Images depict uncropped images for western blots in Extended Data Fig. 4a. Red boxes contain cropped area shown in main figure.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vardhana, S.A., Hwee, M.A., Berisa, M. et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat Immunol 21, 1022–1033 (2020). https://doi.org/10.1038/s41590-020-0725-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0725-2

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