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:

Metabolic programs of T cell tissue residency empower tumour immunity

Abstract

Tissue resident memory CD8+ T (TRM) cells offer rapid and long-term protection at sites of reinfection1. Tumour-infiltrating lymphocytes with characteristics of TRM cells maintain enhanced effector functions, predict responses to immunotherapy and accompany better prognoses2,3. Thus, an improved understanding of the metabolic strategies that enable tissue residency by T cells could inform new approaches to empower immune responses in tissues and solid tumours. Here, to systematically define the basis for the metabolic reprogramming supporting TRM cell differentiation, survival and function, we leveraged in vivo functional genomics, untargeted metabolomics and transcriptomics of virus-specific memory CD8+ T cell populations. We found that memory CD8+ T cells deployed a range of adaptations to tissue residency, including reliance on non-steroidal products of the mevalonate–cholesterol pathway, such as coenzyme Q, driven by increased activity of the transcription factor SREBP2. This metabolic adaptation was most pronounced in the small intestine, where TRM cells interface with dietary cholesterol and maintain a heightened state of activation4, and was shared by functional tumour-infiltrating lymphocytes in diverse tumour types in mice and humans. Enforcing synthesis of coenzyme Q through deletion of Fdft1 or overexpression of PDSS2 promoted mitochondrial respiration, memory T cell formation following viral infection and enhanced antitumour immunity. In sum, through a systematic exploration of TRM cell metabolism, we reveal how these programs can be leveraged to fuel memory CD8+ T cell formation in the context of acute infections and enhance antitumour 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: Functional genetics in vivo, metabolomics and transcriptional analysis of ex vivo populations of memory CD8+ T cells identify a graded upregulation of SREBP2-dependent metabolic programming across TRM cells.
Fig. 2: An SREBP2-dependent metabolic program controls SI TRM formation.
Fig. 3: Non-steroidal products of the mevalonate–cholesterol synthesis pathway mediate TRM cell adaptations.
Fig. 4: Pdss2 expression is required and sufficient to promote mitochondrial respiration and CoQ synthesis by CD8+ T cells.
Fig. 5: Increased production of non-steroidal products of the mevalonate–cholesterol synthesis pathway is a common requirement for SI TRM cells and TILs.

Similar content being viewed by others

Data availability

Bulk RNA-seq and single-cell RNA-seq data have been deposited to the Gene Expression Omnibus under the reference series GSE207044. In addition, the following published datasets were used: GSE107278 (ref. 17), GSE106107 (ref. 17), GSE182276 (ref. 24), GSE70813 (ref. 64), GSE131847 (ref. 65), GSE125527 (ref. 66), GSE137015 (ref. 28), GSE70813 (ref. 64) and GSE157072 (ref. 67). Source data are provided with this paper.

References

  1. Masopust, D. & Soerens, A. G. Tissue-resident T cells and other resident leukocytes. Annu. Rev. Immunol. https://doi.org/10.1146/annurev-immunol-042617-053214 (2019).

  2. Park, S. L., Gebhardt, T. & Mackay, L. K. Tissue-resident memory T cells in cancer immunosurveillance. Trends Immunol. 40, 735–747 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Byrne, A. et al. Tissue-resident memory T cells in breast cancer control and immunotherapy responses. Nat. Rev. Clin. Oncol. https://doi.org/10.1038/s41571-020-0333-y (2020).

  4. Konjar, Š. et al. Mitochondria maintain controlled activation state of epithelial-resident T lymphocytes. Sci. Immunol. 3, eaan2543 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Steinert, E. M. et al. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell 161, 737–749 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wijeyesinghe, S. et al. Expansible residence decentralizes immune homeostasis. Nature 592, 457–462 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Soerens, A. G. et al. Functional T cells are capable of supernumerary cell division and longevity. Nature 614, 762–766 (2023).

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Gebhardt, T. et al. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 10, 524–530 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Iijima, N. & Iwasaki, A. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 346, 93–98 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jiang, X. et al. Skin infection generates non-migratory memory CD8+ TRM cells providing global skin immunity. Nature 483, 227–231 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Teijaro, J. R. et al. Cutting edge: Tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. J. Immunol. 187, 5510–5514 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Ariotti, S. et al. Skin-resident memory CD8+ T cells trigger a state of tissue-wide pathogen alert. Science 346, 101–105 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Schenkel, J. M. et al. Resident memory CD8 T cells trigger protective innate and adaptive immune responses. Science 346, 98–101 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Mackay, L. K. et al. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc. Natl Acad. Sci. USA 109, 7037–7042 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shin, H. & Iwasaki, A. A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature 491, 463–467 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wu, T. et al. Lung-resident memory CD8 T cells (TRM) are indispensable for optimal cross-protection against pulmonary virus infection. J. Leukoc. Biol. 95, 215–224 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Milner, J. J. et al. Runx3 programs CD8+ T cell residency in non-lymphoid tissues and tumours. Nature 552, 253–257 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nizard, M. et al. Induction of resident memory T cells enhances the efficacy of cancer vaccine. Nat. Commun. 8, 15221 (2017).

    Article  Google Scholar 

  19. Malik, B. T. et al. Resident memory T cells in the skin mediate durable immunity to melanoma. Sci. Immunol. 2, 6346 (2017).

    Article  Google Scholar 

  20. Ganesan, A. P. et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat. Immunol. 18, 940–950 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Djenidi, F. et al. CD8+CD103+ tumor–infiltrating lymphocytes are tumor-specific tissue-resident memory T cells and a prognostic factor for survival in lung cancer patients. J. Immunol. 194, 3475–3486 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Reina-Campos, M., Scharping, N. E. & Goldrath, A. W. CD8+ T cell metabolism in infection and cancer. Nat. Rev. Immunol. 21, 718–738 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Huang, H. et al. In vivo CRISPR screening reveals nutrient signaling processes underpinning CD8+ T cell fate decisions. Cell 184, 1245–1261 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Crowl, J. et al. Tissue-resident memory CD8+ T cells possess unique transcriptional, epigenetic and functional adaptations to different tissue environments. Nat. Immunol. 23, 1121–1131 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Pan, Y. et al. Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism. Nature 543, 252–256 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Milner, J. J. & Goldrath, A. W. Transcriptional programming of tissue-resident memory CD8+ T cells. Curr. Opin. Immunol. 51, 162–169 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Frizzell, H. et al. Organ-specific isoform selection of fatty acid-binding proteins in tissue-resident lymphocytes. Sci. Immunol. 5, 9283 (2020).

    Article  Google Scholar 

  28. Wei, J. et al. Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature 576, 471–476 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Locke, F. L., Zha, Y., Zheng, Y., Driessens, G. & Gajewski, T. F. Conditional deletion of PTEN in peripheral T cells augments TCR-mediated activation but does not abrogate CD28 dependency or prevent anergy induction. J. Immunol. 191, 1677–1685 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Pauls, S. D. & Marshall, A. J. Regulation of immune cell signaling by SHIP1: a phosphatase, scaffold protein, and potential therapeutic target. Eur. J. Immunol. 47, 932–945 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Kidani, Y. et al. Sterol regulatory element-binding proteins are essential for the metabolic programming of effector T cells and adaptive immunity. Nat. Immunol. 14, 489–499 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yu, B. et al. Epigenetic landscapes reveal transcription factors that regulate CD8+ T cell differentiation. Nat. Immunol. 18, 573–582 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fernandez-Ruiz, D. et al. Liver-resident memory CD8+ T cells form a front-line defense against malaria liver-stage infection. Immunity 45, 889–902 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Shimano, H. & Sato, R. SREBP-regulated lipid metabolism: convergent physiology – divergent pathophysiology. Nat. Rev. Endocrinol. 13, 710–730 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Zeiser, R. Immune modulatory effects of statins. Immunology 154, 69–75 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sullivan, Z. A. et al. γδ T cells regulate the intestinal response to nutrient sensing. Science 371, eaba8310 (2021).

  37. Menk, A. V. et al. 4-1BB costimulation induces T cell mitochondrial function and biogenesis enabling cancer immunotherapeutic responses. J. Exp. Med. 215, 1091–1100 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Scharping, N. E. et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity https://doi.org/10.1016/j.immuni.2016.07.009 (2016).

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

  40. Lanterna, C. et al. The administration of drugs inhibiting cholesterol/oxysterol synthesis is safe and increases the efficacy of immunotherapeutic regimens in tumor-bearing mice. Cancer Immunol. Immunother. 65, 1303–1315 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Shires, J., Theodoridis, E. & Hayday, A. C. Biological insights into TCRγδ+ and TCRαβ+ intraepithelial lymphocytes provided by serial analysis of gene expression (SAGE). Immunity 15, 419–434 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Fahrer, A. M. et al. Attributes of γδ intraepithelial lymphocytes as suggested by their transcriptional profile. Proc. Natl Acad. Sci. USA 98, 10261–10266 (2001).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hoytema van Konijnenburg, D. P. et al. Intestinal epithelial and intraepithelial T cell crosstalk mediates a dynamic response to infection. Cell 171, 783–794 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Passi, S., Stancato, A., Aleo, E., Dmitrieva, A. & Littarru, G. P. Statins lower plasma and lymphocyte ubiquinol/ubiquinone without affecting other antioxidants and PUFA. BioFactors 18, 113–124 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Xu, S. et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity 54, 1561–1577 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ma, X. et al. Cholesterol induces CD8+ T cell exhaustion in the tumor microenvironment. Cell Metab. 30, 143–156 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. O’Connor, A., Quizon, P. M., Albright, J. E., Lin, F. T. & Bennett, B. J. Responsiveness of cardiometabolic-related microbiota to diet is influenced by host genetics. Mamm. Genome 25, 583–599 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc. 9, 209–222 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. McCausland, M. M. & Crotty, S. Quantitative PCR technique for detecting lymphocytic choriomeningitis virus in vivo. J. Virol. Methods 147, 167–176 (2008).

    Article  CAS  PubMed  Google Scholar 

  50. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-Seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Brinkman, E. K., Chen, T., Amendola, M. & Van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kushnareva, Y. et al. Functional analysis of immune signature genes in Th1* memory cells links ISOC1 and pyrimidine metabolism to IFN-γ and IL-17 production. J. Immunol. 206, 1181–1193 (2021).

    Article  CAS  PubMed  Google Scholar 

  62. Ye, Q. et al. HORMA domain proteins and a Trip13-like ATPase regulate bacterial cGAS-like enzymes to mediate bacteriophage immunity. Mol. Cell 77, 709–722 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Mackay, L. K. et al. Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes. Science 352, 459–463 (2016).

    Article  ADS  CAS  PubMed  Google Scholar 

  65. Kurd, N. S. et al. Early precursors and molecular determinants of tissue-resident memory CD8+ T lymphocytes revealed by single-cell RNA sequencing. Sci. Immunol. 5, eaaz6894 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Boland, B. S. et al. Heterogeneity and clonal relationships of adaptive immune cells in ulcerative colitis revealed by single-cell analyses. Sci. Immunol. 5, eabb4432 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Milner, J. J. et al. Delineation of a molecularly distinct terminally differentiated memory CD8 T cell population. Proc. Natl Acad. Sci. USA 117, 25667–25678 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  68. Trindade, B. C. et al. The cholesterol metabolite 25-hydroxycholesterol restrains the transcriptional regulator SREBP2 and limits intestinal IgA plasma cell differentiation. Immunity 54, 2273–2287 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Pipkin for sharing the shSrebf2 plasmid, A. Reboldi for sharing the nSREBP2-OE plasmid, S. Crotty and K. Faliti for sharing the LsgA vector backbone, and G. Garcia and M. Sevilla for technical histological assistance. We also thank the Center for Advanced Technology at UCSF for sequencing support, L. Labarta-Bajo and the Goldrath laboratory members for technical advice, helpful discussion and critical reading of the manuscript, and the Immunological Genome Project (ImmGen) for reagents and sample and data processing. We thank A. M. Pinto and the Mass Spectrometry Core of the Salk Institute (supported by NIH-NCI CCSG: P30 014195 and the Helmsley Center for Genomic Medicine). The MS data described here were gathered on a Thermo Fisher Q Exactive hybrid quadrupole–Orbitrap mass spectrometer (supported by 1S10OD021815-01). Funding is as follows: National Institutes of Health (NIH) grants R01 AI067545, P01 AI132122, R01 AI150282 and R01 AI072117 (A.W.G.); NIH grant R01 CA197363 (M.F.K.); NIH grants P01 HL146358 and R01 HL157710 (S.J.B.); Cancer Research Institute Postdoctoral Fellowship CRI2943 (M.R.-C.); Cancer Research Institute Postdoctoral Fellowship CRI4145 (G.G.); HHMI fellowship GT14887 (K.K.); NCI grant P30 CA030199 (SBP Flow Cytometry Core); NIH grant P30 DK063720 (UCSF Parnassus Flow CoLab); NIH SIG grant S10 OD026929 (UCSD IGM Core).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: M.R.-C., A.W.G. Methodology: M.R.-C., K.K., I.T.M., K.H.H., G.G., A.W.G., S.S., M.J., M.F.K., H.C. Investigation: M.R.-C., K.K., I.T.M., V.L., Q.N., G.G., H.H., J.J.M., K.H.H., A.V., N.S., B.S.B. Visualization: M.R.-C., M.H., K.K., I.T.M., G.G. Funding acquisition: M.R.-C., A.W.G., J.T.C., S.S., M.F.K., H.C., S.J.B. Project administration: A.W.G. Supervision: M.R.-C., A.W.G. Writing – original draft: M.R.-C., A.W.G. Writing – review and editing: M.R.-C., M.H., G.G., H.C., S.S., S.J.B., A.W.G.

Corresponding author

Correspondence to Ananda W. Goldrath.

Ethics declarations

Competing interests

A.W.G. serves on the scientific advisory boards of ArsenalBio and Foundery Innovations. H.C. is a consultant for Kumquat Biosciences. M.F.K. is a founder and shareholder of PIONYR Immunotherapeutic and Foundery Innovations. A provisional patent application has been filed by M.R.-C., G.G. and A.W.G. covering the methods of use of FDFT1 and PDSS2 manipulation for “Enhancing CAR-T cell performance for cancer immunotherapy” (US Provisional Application Serial No. 63/449,701). The rest of the authors declare no competing interests.

Peer review

Peer review information

Nature thanks Martin Giera and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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 figures and tables

Extended Data Fig. 1 Functional genetics in vivo, metabolomics and transcriptional analysis of ex vivo populations of memory CD8+ T cells identify a graded upregulation of a Srebp2-dependent metabolic programming across TRM.

a, Unsupervised hierarchical clustering heatmap of scaled GSVA scores for metabolic signatures across samples, p.i., post-infection. GSE107278. b, sgRNA library heterogeneity reported as Gini Index values. c, Upset plot showing intersection sizes and set sizes for positive and negative regulators of each P14 CD8+ T cell population in the in vivo CRISPR/Cas9-mediated loss-of-function screen. d, Sample acquisition and analysis workflow for LC-MS/MS-based untargeted metabolomics of ex vivo populations of CD8+ T cells in the context of LCMV infection. Gating strategy included for each cell type analyzed. e, Tandem mass spectra matching against commercial standards of mevalonate and mevalonolactone. Tandem mass spectra derived from the most abundant ion for each compound. Black spectra are derived from cell lysate, red mirrored spectra from commercial standard. f, Unsupervised hierarchical clustering of scaled relative abundances of identified annotated metabolites of P14 CD8+ T cells profiled by untargeted metabolomics at day 13 after LCMV infection. The images of the mice in d have been modified from ref. 17.

Extended Data Fig. 2 Upregulation of the mevalonate-cholesterol synthesis pathway in mouse and human TRM.

a, Mevalonate-cholesterol synthesis GSVA scores from averaged single cell expression of spleen and SI P14 CD8+ T cells at indicated time points after LCMV infection profiled by scRNA-seq, GSE131847. b, Mevalonate-cholesterol synthesis GSVA scores for indicated subsets of CD8+ T cells in the spleen or SI in the context of LCMV infection, GSE157072. c, Mevalonate-cholesterol synthesis GSVA scores for indicated subsets of CD8+ T cells at the indicated tissues in the context of LCMV and Herpes Simplex Virus (HSV), GSE70813. d, Gene expression of indicated genes in P14 CD8+ T cell populations from indicated tissues >30 days after LCMV infection, GSE182276e, Mevalonate-cholesterol synthesis pathway scores for all human immune cells grouped by indicated tissues (left), cell types (middle), or cell types of the Jejunum SI (right), from the tissue immune cell scRNA-seq atlas. f, Expression of selected genes on human CD8+ T cells from PBMC, rectum, and SI samples, pooled from 13 healthy donors, GSE125527. g, HMGCR protein expression in CD8+ T cells from human spleen, SI, and colon by immunofluorescence staining. Quantification of total HMGCR by CD8+ T cell area. Donor ID (#). Scale bar, 20 μm. h, GSEA of the mevalonate-cholesterol synthesis in Runx3 KO or control CD8+ T cells GSE106107. Data are pseudo-bulk averaged values (a), mean +/− s.e.m. (b, c, and d), or geometric distribution (f and g), with a total of n = 2 (b), n = 2–4 (c), n = 2–5 (d) mice, and n = 1-2 (g) samples. Two-sided unpaired t-Test (b, c, d, f, and g), and Gene Set Enrichment Analysis (h). *P < 0.05, **P < 0.01, ***P < 0.005.

Source data

Extended Data Fig. 3 A Srebp2-dependent metabolic program controls SI TRM formation.

a, Quantification of gene expression by qPCR of shRNAmir control (shCd19) or shSrebf2 shRNAmir P14 CD8+ T cells before adoptive transfer. b, Frequency of effector shRNAmir control (shCd19) or shSrebf2 shRNAmir P14 CD8+ T cell populations in the spleen 7 days after LCMV infection. c, Frequency of memory shRNAmir control (shCd19) or shSrebf2 shRNAmir P14 CD8+ T cell populations in the spleen 21 days after LCMV infection. d, CRISPR/Cas9-mediated indel efficiency of the Srebf2 sgRNA construct on sorted transduced P14 CD8+ T cells before adoptive transfer. e, Frequency of effector sgCd19 or sgSrebf2 P14 CD8+ T cell populations in the spleen 7 days after LCMV infection. f, Frequency of memory sgCd19 or sgSrebf2 P14 CD8+ T cell populations in the spleen 21 days after LCMV infection. g, Total cell quantification of CD44high CD8+ T cells (left) and Tet+ CD8+ T cells (right) in the blood of Scap WT or Scap KO at different time points after LCMV infection. h, Total cell quantification of Tet+ CD8+ T cells (upper) and CD44high CD8+ T cells (lower) in SI, kidney, and liver of Scap WT or Scap KO at different time points after LCMV infection. i, Ratio of congenically distinct P14 CD8+ T cells transduced with a constitutively active nuclear form of human Srebp268 (nSrebp2) or an empty vector (Empty) from indicated tissues 7 days after LCMV infection. j, Flow cytometry analysis of cholesterol content by Filipin stain (left), and LDLR expression in P14 CD8+ T cells transduced with a bicistronic construct encoding Thy1.1 alone or Thy1.1 and nSrebp2 (right). P14 CD8+ T cells from the kidney, liver, WAT, and SI were gated on the IV population (h and i). Data are mean +/− s.e.m. and representative of at least two independent experiments, with a total of n = 2 (a), n = 4 (b-e), n = 5 (f), n = 6 (WT) and n = 7 (Scap KO) (g and h), n = 4 (i and j) mice and n = 2 (d) cell replicates. Two-sided unpaired (a, d, g, and h) and paired (j) t-Test. *P < 0.05, **P < 0.01, ***P < 0.005. Two-sided one-sample t-Test (i) #P < 0.05, ##P < 0.01, ###P < 0.005. Two-way ANOVA (h) ##P < 0.01.

Source data

Extended Data Fig. 4 Limited effect of Srebp2 on transcriptional programs of CD8+ T cell differentiation.

a, Srebf2 expression values from the bulk Ultralow-input RNA-seq analysis of shRNAmir control (shCd19) and Srebf2 shRNAmir P14 CD8+ T cells from the spleen, kidney, liver, and SI from day 7 and 14 after LCMV infection. b, Mevalonate-cholesterol synthesis pathway GSVA scores from the bulk Ultralow-input RNA-seq analysis of shRNAmir control (shCd19) and Srebf2 shRNAmir P14 CD8+ T cells from the spleen, kidney, liver, and SI from day 7 and 14 after LCMV infection. c, Unsupervised hierarchically clustered heatmap of mevalonate-cholesterol synthesis pathway gene expression values from the bulk Ultralow-input RNA-seq analysis of shRNAmir control (shCd19) and Srebf2 shRNAmir P14 CD8+ T cells from the spleen, kidney, liver, and SI from day 7 and 14 after LCMV infection. d, Core TRM GSVA scores from the bulk Ultralow-input RNA-seq analysis of shRNAmir control (shCd19) and Srebf2 shRNAmir P14 CD8+ T cells from the spleen, kidney, liver, and SI from day 7 and 14 after LCMV infection. e, Circulating GSVA scores from the bulk Ultralow-input RNA-seq analysis of shRNAmir control (shCd19) and Srebf2 shRNAmir P14 CD8+ T cells from the spleen, kidney, liver, and SI from day 7 and 14 after LCMV infection. f, Volcano plots of differentially expressed genes between shCd19 and shSrebf2 SI P14 CD8+ T cells at days 7 and 14 after LCMV infection, adjusted (adj) p-value. P14 CD8+ T cells from the kidney, liver and SI were gated on the IV population (a-f). TE were defined as KLRG1highCD127low P14 CD8+ T cells. Data are mean +/− s.e.m.. (a, b, d, and e) from one experiment with n = 2-3 (a-f), where every sample is 1×103 cells pooled from 2~3 mice. Two-sided unpaired t-Test (a, b, d, and e), and Deseq2 DEG testing with Benjamini-Hochberg multiple test correction. *P < 0.05, **P < 0.01, ***P < 0.005.

Source data

Extended Data Fig. 5 SI TRM uptake and sense dietary cholesterol.

a, Quantification by flow cytometry of in vivo incorporation of Bodipy-cholesterol on indicated cell populations after 10 minutes after oral administration of Bodipy-cholesterol to memory P14 mice. LPL, lamina propia lymphocytes. b, Quantification by flow cytometry of incorporation of Bodipy-LDL on indicated cell types of the spleen 20 minutes (min) after IV delivery to memory P14 mice. c, Quantification of intracellular cholesterol by filipin staining in Tet+ populations of CD8+ T cells for various tissues >30 days post LCMV infection measured by imaging flow cytometry. d, Gene expression analysis by qPCR of selected SREBP2 targets on CD8+ T cells from the spleen and SI of mice on regular chow or subjected to low and high cholesterol-containing diets. e, Experimental design to measure transcriptional changes induced by dietary cholesterol on established SI TRM by Ultralow-input RNA-seq, f, SI TRM frequencies from total CD8+ T cells after 7 days of dietary intervention. g, Food consumption per day per mouse subjected to a low or high-cholesterol diet. h, Bodyweight of mice subjected to low or high cholesterol-containing diets. i, Total cholesterol in serum in mice subjected to low or high cholesterol-containing diets. j, Volcano plot of differentially expressed genes in SI TRM from mice fed a high vs a low cholesterol-containing diet. k, Mevalonate-cholesterol synthesis GSVA scores in SI TRM from mice fed a high versus low cholesterol-containing diet. l, Gene pathway analysis of top upregulated and top downregulated genes between SI TRM from mice fed a high versus low cholesterol-containing diet (MsigDb, Hallmark gene sets). P14 CD8+ T cells and CD8β+/CD44high/Tet+ from the kidney, liver and SI were gated on the IV population (a, b, c, e, f, j, k, and l). Data are mean +/− s.e.m. and representative of at least two independent experiments, with a total of n = 4 (a), n = 3 (b, d), n = 5 (c), n = 3 (f) mice, or pooled from at least two independent experiments with a total of n = 5–10 (g-i) mice, or performed once (j-l) with a total of n = 3 samples each sample is 1 × 103 SI TRM pooled from 2~3 mice (j, k). Two-sided unpaired t-Test (a, b, d, f, i, and k), and Deseq2 DEG testing with Benjamini-Hochberg multiple test correction (j). *P < 0.05, **P < 0.01, ***P <  0.005. The images of the mice in a,b,e have been modified from ref. 17.

Source data

Extended Data Fig. 6 Mevalonate-cholesterol synthesis pathway intermediates sustain the production of non-steroidal metabolites.

a, The mevalonate-cholesterol synthesis pathway with adjacent and downstream metabolic routes. Enzymes not included in the first CRISPR/Cas9-mediated loss-of-function screen in grey, downstream products and/or fates of cholesterol are boxed, key measured metabolites in bold, non-steroidal metabolites in blue, key enzymes are bold and colored by in vivo effect of deletion in P14 CD8+ T cells, gain-of-function in red, loss-of-function in blue, dashed lines indicate indirect or unknown mode of regulation. b, Abundance of 4-hydroxybenzaldehyde profiled by LC-MS/MS of ex vivo populations of P14 CD8+ T cells 13 days after LCMV infection. c, Tandem mass spectra matching against commercial standards of 4-hydroxybenzaldehyde. Tandem mass spectra derived from the most abundant ion for each compound: m/z = 121.0295 for 4-hydroxybenzaldehyde [M-H], black spectra are derived from cell lysate, red mirrored spectra from commercial standard. d, CRISPR/Cas9-mediated indel efficiency of the Hpd sgRNA construct on sorted transduced P14 CD8+ T cells before adoptive transfer. e, CRISPR/Cas9-mediated indel efficiency of the Fdft1 sgRNA construct on sorted transduced P14 CD8+ T cells before adoptive transfer. f, Gini index scores of sgRNA representation in each library. g, Schematic illustration of the contribution of mevalonate-cholesterol pathway intermediates to non-steroidal products. h, Quantification of Pdss2 mRNA by qPCR in transduced Empty and Pdss2 OE P14 CD8+ T cells. Data are mean +/− s.e.m. (b and h), and representative of at least two independent experiments (d, e, and h), with a total of n = 2 (d, e), and n = 3 (h) cell replicates, or one experiment (b), with n = 3 (SI), n = 4 (TN), and n = 5 (rest) (b) samples, where each sample contains cells pooled from 2 to 5 mice. Two-sided unpaired t-Test (b, d, e and h). *P < 0.05, **P < 0.01, ***P < 0.005.

Source data

Extended Data Fig. 7 Increased production of non-steroidal products of the mevalonate-cholesterol synthesis pathway is a common requirement between SI TRM and TIL.

a, Dimensionality reduction plot by UMAP of P14 CD8+ T cells profiled from spleens and subcutaneously implanted MC38-GP33–41 tumors by scRNAseq 7 days after ACT. b, Mevalonate-cholesterol synthesis signature values and expression of indicated genes in P14 CD8+ T cells from the spleen and MC38-GP33–41 tumors profiled by scRNAseq. MAGIC-imputed gene values are shown. c, Hierarchically clustered heatmap of scaled averaged gene expression values by cluster of P14 CD8+ T cells profiled from spleens and MC38-GP33–41 tumors by scRNAseq 7 days after adoptive transfer. d, Dimensionality reduction plot by UMAP of CD8+ T cell metaclusters (left) with “meta.cluster.coarse” labels and hierarchically clustered heatmap of mevalonate-cholesterol synthesis pathway scores grouped by “meta.cluster.coarse” labels and cell type. NPC, nasopharingeal carcinoma, ESCA, esophageal cancer, LC, lung cancer, HCC, hepatocellular carcinoma, CHOL, cholangiocarcinoma, RC, renal carcinoma, CRC, colorectal cancer, AML, acute myeloid leukemia, BCL, B-cell lymphoma, MM, multiple myeloma, HNSCC, head and neck squamous cell carcinoma, THCA, thyroid carcinoma, MELA, melanoma, BCC, basal cell carcinoma, SCC, squamous cell carcinoma, BRCA, breast cancer, STAD, stomach adenocarcinoma, PACA, pancreatic cancer, OV, ovarian cancer, FTC, fallopian tube carcinoma, UCEC, uterine corpus endometrial carcinoma. EMRA, effector memory CD45RA+, mem, memory. e, sgRNA library heterogeneity reported as Gini Index values. f, CRISPR/Cas9-mediated indel efficiency of the Pdss2 sgRNA construct on sorted transduced P14 CD8+ T cells. g, Frequency of PD1highTim3high and PD1highTim3low subpopulations for sgCd19 and sgFdft1 Cas9 P14 CD8+ T cells in MC38-GP33–41 tumors at day 5–7 after adoptive transfer. h, Frequency of PD1highTim3high and PD1highTim3low subpopulations for Empty and Pdss2 OE P14 CD8+ T cells in MC38-GP33–41 tumors at day 5–7 after adoptive transfer. i, Quantification of gene expression by qPCR of control (shNT) and Fdft1 deficient (shFdft1) P14 CD8+ T cells before adoptive transfer. j, Restimulation capacity of control (shNT) and Fdft1 deficient (shFdft1) P14 TIL 4 days after in vivo transfer treated with GP33–41 peptide ex vivo. k, MC38-GP33–41 tumor growth curves of mice receiving ZAA or vehicle. l, Survival of mice implanted with MC38-GP33–41 tumors receiving ZAA or vehicle. m, MC38-GP33–41 tumor growth curves of mice receiving ZAA in combination with CD8+ T cell depletion. Data are mean +/− s.e.m. (g, h, i, j, k, and m), raw values (d), geometrical distributions of single-cell gene expression values (b) or averaged gene expression (c and e) and pooled or representative of at least two independent experiments, with a total of n = 2 (f) cell replicates, and n = 8 (g), n = 9 (h), n = 3 (i), n = 10 (j), n = 12/group (k, l), n = 5/group (m) mice. Two-sided unpaired t-Test (b, f and i). Log-rank test (l). Two-way ANOVA (m). *P < 0.05, **P < 0.01, ***P < 0.005.

Source data

Extended Data Fig. 8 A requirement for the mevalonate-cholesterol synthesis enzymes for TIL persistence and heightened antitumor activity in a melanoma model.

a, GSEA of the mevalonate-cholesterol synthesis pathway in genes pre-ranked by their impact on TIL accumulation in B16-OVA tumors from a published CRISPR/Cas9-mediated loss-of-function screen. b, Dimensionality reduction plot by UMAP of WT and Regnase-1 KO OT-I cells profiled from MC38-GP33–41 tumors by scRNA-seq 7 days after adoptive transfer (left) and unsupervised hierarchical clustering heatmap of selected genes related to CD8+ T cell cytotoxicity (Ifng) stemness (Tcf7), proliferation (Mki67), and the mevalonate-cholesterol synthesis pathway (Srebf2, Hmgcs1, Fdft1, Hmgcr, and Ldlr) grouped by cell type and cluster. c, Comparison of the effect of targeting the 50 genes of the mevalonate-cholesterol pathway included in our targeted screen in WT TIL or Regnase-1 KO TIL (sgZc3h12a) from a published CRISPR/Cas9-mediated loss-of-function screen in B16 melanoma tumors32. Relevant enzymes are labeled.

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

Gene-level enrichment scores for all samples versus the input for each sample and library.

Supplementary Table 2

Curated list of metabolic signatures.

Supplementary Table 3

Pathway-level enrichment scores for all samples versus the input for each sample and library for the metabolic CRISPR–Cas9 screen.

Supplementary Table 4

Polar metabolites identified by LC–MS/MS.

Supplementary Table 5

Statin usage information for the human cohort profiled by single-cell RNA-seq.

Supplementary Table 6

Differentially expressed genes in P14 CD8+ T cells isolated from the SI of mice fed a high- or low-cholesterol diet for 7 days and profiled by ultralow-input RNA-seq.

Supplementary Table 7

Targeted library sgRNA information.

Supplementary Table 8

Gene-level enrichment scores for all sgCd19 and sgFdft1 samples versus corresponding inputs in the mevalonate–cholesterol synthesis pathway-targeted CRISPR–Cas9-mediated loss-of-function screen.

Supplementary Table 9

Gene-level enrichment scores for TIL, dLN and SI TRM samples versus corresponding inputs in the mevalonate–cholesterol synthesis pathway-targeted CRISPR–Cas9-mediated loss-of-function screen.

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

Reina-Campos, M., Heeg, M., Kennewick, K. et al. Metabolic programs of T cell tissue residency empower tumour immunity. Nature 621, 179–187 (2023). https://doi.org/10.1038/s41586-023-06483-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06483-w

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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