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:

Distinct metabolic programs established in the thymus control effector functions of γδ T cell subsets in tumor microenvironments

Abstract

Metabolic programming controls immune cell lineages and functions, but little is known about γδ T cell metabolism. Here, we found that γδ T cell subsets making either interferon-γ (IFN-γ) or interleukin (IL)-17 have intrinsically distinct metabolic requirements. Whereas IFN-γ+ γδ T cells were almost exclusively dependent on glycolysis, IL-17+ γδ T cells strongly engaged oxidative metabolism, with increased mitochondrial mass and activity. These distinct metabolic signatures were surprisingly imprinted early during thymic development and were stably maintained in the periphery and within tumors. Moreover, pro-tumoral IL-17+ γδ T cells selectively showed high lipid uptake and intracellular lipid storage and were expanded in obesity and in tumors of obese mice. Conversely, glucose supplementation enhanced the antitumor functions of IFN-γ+ γδ T cells and reduced tumor growth upon adoptive transfer. These findings have important implications for the differentiation of effector γδ T cells and their manipulation in cancer immunotherapy.

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: Intratumoral γδ T cell subsets display distinct metabolic profiles.
Fig. 2: Peripheral γδ T cell subsets show different mitochondrial and metabolic phenotypes.
Fig. 3: γδ T cell subsets are metabolically programmed in the thymus.
Fig. 4: Distinct mitochondrial activities underlie effector fate of thymic γδ T cell progenitors.
Fig. 5: γδ17 cells show higher lipid uptake and lipid droplet content than γδIFN cells.
Fig. 6: HFD promotes the expansion of pro-tumoral γδ17 cells in lymph nodes and within tumors.
Fig. 7: Glucose supplementation enhances the antitumor effector functions of γδIFN cells.

Similar content being viewed by others

Data availability

The GEO public repository accession codes are GSE150585 for single-cell RNA sequencing; and GSE156782 for bulk RNA sequencing. The data that support the findings of this study are available from the corresponding author upon request.

References

  1. Buck, M. D., Sowell, R. T., Kaech, S. M. & Pearce, E. L. Metabolic instruction of immunity. Cell 169, 570–586 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Almeida, L., Lochner, M., Berod, L. & Sparwasser, T. Metabolic pathways in T cell activation and lineage differentiation. Semin. Immunol. 28, 514–524 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Geltink, R. I. K., Kyle, R. L. & Pearce, E. L. Unraveling the complex interplay between T cell metabolism and function. Annu Rev. Immunol. 36, 461–488 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Cham, C. M., Driessens, G., O’Keefe, J. P. & Gajewski, T. F. Glucose deprivation inhibits multiple key gene expression events and effector functions in CD8+ T cells. Eur. J. Immunol. 38, 2438–2450 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chang, C. H. et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 162, 1229–1241 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. O’Sullivan, D., Sanin, D. E., Pearce, E. J. & Pearce, E. L. Metabolic interventions in the immune response to cancer. Nat. Rev. Immunol. 19, 324–335 (2019).

    Article  PubMed  Google Scholar 

  8. Silva-Santos, B., Mensurado, S. & Coffelt, S. B. γδ T cells: pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer 19, 392–404 (2019).

    Article  CAS  PubMed  Google Scholar 

  9. Sebestyen, Z., Prinz, I., Déchanet-Merville, J., Silva-Santos, B. & Kuball, J. Translating gammadelta (γδ) T cells and their receptors into cancer cell therapies. Nat. Rev. Drug Discov. 19, 169–184 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Chien, Y. H., Meyer, C. & Bonneville, M. γδ T cells: first line of defense and beyond. Annu Rev. Immunol. 32, 121–155 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Hayday, A. C. γδ T cells and the lymphoid stress-surveillance response. Immunity 31, 184–196 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Hayday, A. C. γδ T cell update: adaptate orchestrators of immune surveillance. J. Immunol. 203, 311–320 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Ravens, S. et al. Human γδ T cells are quickly reconstituted after stem-cell transplantation and show adaptive clonal expansion in response to viral infection. Nat. Immunol. 18, 393–401 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Kohlgruber, A. C. et al. γδ T cells producing interleukin-17A regulate adipose regulatory T cell homeostasis and thermogenesis. Nat. Immunol. 19, 464–474 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ribeiro, M. et al. Meningeal γδ T cell–derived IL-17 controls synaptic plasticity and short-term memory. Sci. Immunol. https://doi.org/10.1126/sciimmunol.aay5199 (2019).

  16. Papotto, P. H., Ribot, J. C. & Silva-Santos, B. IL-17+ γδ T cells as kick-starters of inflammation. Nat. Immunol. 18, 604–611 (2017).

    Article  CAS  PubMed  Google Scholar 

  17. Jensen, K. D. et al. Thymic selection determines γδ T cell effector fate: antigen-naive cells make interleukin-17 and antigen-experienced cells make interferon γ. Immunity 29, 90–100 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ribot, J. C. et al. CD27 is a thymic determinant of the balance between interferon-γ- and interleukin 17–producing γδ T cell subsets. Nat. Immunol. 10, 427–436 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sumaria, N., Grandjean, C. L., Silva-Santos, B. & Pennington, D. J. Strong TCRγδ signaling prohibits thymic development of IL-17A-secreting γδ T cells. Cell Rep. 19, 2469–2476 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Muñoz-Ruiz, M. et al. TCR signal strength controls thymic differentiation of discrete proinflammatory γδ T cell subsets. Nat. Immunol. 17, 721–727 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Muñoz-Ruiz, M., Sumaria, N., Pennington, D. J. & Silva-Santos, B. Thymic determinants of γδ T cell differentiation. Trends Immunol. 38, 336–344 (2017).

    Article  PubMed  Google Scholar 

  22. Silva-Santos, B., Serre, K. & Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 15, 683–691 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Argüello, R. J. et al. SCENITH: a flow cytometry based method for functional profiling energy metabolism with single cell resolution. Cell Metab. 32, 1063–1075.e7 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Gleyzer, N., Vercauteren, K. & Scarpulla, R. C. Control of mitochondrial transcription specificity factors (TFB1M and TFB2M) by nuclear respiratory factors (NRF-1 and NRF-2) and PGC-1 family coactivators. Mol. Cell. Biol. 25, 1354–1366 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Scarpulla, R. C. Nuclear control of respiratory chain expression in mammalian cells. J. Bioenerg. Biomembr. 29, 109–119 (1997).

    Article  CAS  PubMed  Google Scholar 

  26. Dang, C. V. et al. The c-Myc target gene network. Semin. Cancer Biol. 16, 253–264 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Guo, Q. M. et al. Identification of c-myc responsive genes using rat cDNA microarray. Cancer Res. 60, 5922–5928 (2000).

    CAS  PubMed  Google Scholar 

  28. In, T. S. H. et al. HEB is required for the specification of fetal IL-17-producing γδ T cells. Nat. Commun. 8, 2004 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Coffey, F. et al. The TCR ligand-inducible expression of CD73 marks γδ lineage commitment and a metastable intermediate in effector specification. J. Exp. Med. 211, 329–343 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Turchinovich, G. & Hayday, A. C. Skint-1 identifies a common molecular mechanism for the development of interferon-γ-secreting versus interleukin-17-secreting γδ T cells. Immunity 35, 59–68 (2011).

    Article  CAS  PubMed  Google Scholar 

  31. Lu, Y., Cao, X., Zhang, X. & Kovalovsky, D. PLZF controls the development of fetal-derived IL-17+Vγ6+ γδ T cells. J. Immunol. 195, 4273–4281 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Tan, L. et al. Single-cell transcriptomics identifies the adaptation Scart1+ Vγ6+ T cells to skin residency as activated effector cells. Cell Rep. 27, 3657–3671.e4 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. McCully, M. L. et al. Skin metabolites define a new paradigm in the localization of skin tropic memory T cells. J. Immunol. 195, 96–104 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bartz, R. et al. Lipidomics reveals that adiposomes store ether lipids and mediate phospholipid traffic. J. Lipid Res. 48, 837–847 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Le Goffe, C., Vallette, G., Jarry, A., Bou-Hanna, C. & Laboisse, C. L. The in vitro manipulation of carbohydrate metabolism: a new strategy for deciphering the cellular defence mechanisms against nitric oxide attack. Biochem. J. 344, 643–648 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Bustamante, E. & Pedersen, P. L. High aerobic glycolysis of rat hepatoma cells in culture: role of mitochondrial hexokinase. Proc. Natl Acad. Sci. USA 74, 3735–3739 (1977).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Buck, M. D., O’Sullivan, D. & Pearce, E. L. T cell metabolism drives immunity. J. Exp. Med. 212, 1345–1360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Menk, A. V. et al. Early TCR signaling induces rapid aerobic glycolysis enabling distinct acute T cell effector functions. Cell Rep. 22, 1509–1521 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Shin, B. et al. Mitochondrial oxidative phosphorylation regulates the fate decision between pathogenic TH17 and regulatory T cells. Cell Rep. 30, 1898–1909.e4 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Di Luccia, B., Gilfillan, S., Cella, M., Colonna, M. & Huang, S. C. ILC3s integrate glycolysis and mitochondrial production of reactive oxygen species to fulfill activation demands. J. Exp. Med. 216, 2231–2241 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Schmolka, N. et al. Epigenetic and transcriptional signatures of stable versus plastic differentiation of proinflammatory γδ T cell subsets. Nat. Immunol. 14, 1093–1100 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Schmolka, N., Wencker, M., Hayday, A. C. & Silva-Santos, B. Epigenetic and transcriptional regulation of γδ T cell differentiation: programming cells for responses in time and space. Semin. Immunol. 27, 19–25 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Legut, M., Cole, D. K. & Sewell, A. K. The promise of γδ T cells and the γδ T cell receptor for cancer immunotherapy. Cell Mol. Immunol. 12, 656–668 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nakamizo, S. et al. High fat diet exacerbates murine psoriatic dermatitis by increasing the number of IL-17-producing γδ T cells. Sci. Rep. 7, 14076 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Goldberg, E. L. et al. Ketogenic diet activates protective γδ T cell responses against influenza virus infection. Sci. Immunol. https://doi.org/10.1126/sciimmunol.aav2026 (2019).

  47. Michelet, X. et al. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat. Immunol. 19, 1330–1340 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Fischer, K. et al. Inhibitory effect of tumor cell–derived lactic acid on human T cells. Blood 109, 3812–3819 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Wang, Z. et al. Glycolysis and oxidative phosphorylation play critical roles in natural killer cell receptor-mediated natural killer cell functions. Front Immunol. 11, 202 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Almeida, A. R. et al. Delta one T cells for immunotherapy of chronic lymphocytic leukemia: clinical-grade expansion/differentiation and preclinical proof of concept. Clin. Cancer Res. 22, 5795–5804 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Lynch, L. et al. Regulatory iNKT cells lack expression of the transcription factor PLZF and control the homeostasis of Treg cells and macrophages in adipose tissue. Nat. Immunol. 16, 85–95 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. Vu, J. P. et al. Long-term intake of a high-protein diet affects body phenotype, metabolism, and plasma hormones in mice. J. Nutr. 147, 2243–2251 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kohlgruber, A. C. et al. γδ T cells producing interleukin-17A regulate adipose regulatory T cell homeostasis and thermogenesis. Nat. Immunol. 19, 464–474 (2018).

  54. McKenzie, D. R. et al. IL-17-producing γδ T cells switch migratory patterns between resting and activated states. Nat. Commun. 8, 15632 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Stuart, T. et al. Comprehensive integration of single-cell data resource comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

  57. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Liberzon, A. et al. Molecular Signatures Database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Linderman, G. C., Zhao, J. & Kluger, Y. Zero-preserving imputation of scRNA-seq data using low-rank approximation. Preprint at bioRxiv https://doi.org/10.1101/397588 (2018).

Download references

Acknowledgements

We are grateful for the valuable assistance of the staff of the flow cytometry, bioimaging and animal facilities at our institutions. We thank J.-W. Taanman, A. Magalhães, J. Ribot, K. Serre and N. Sousa for technical suggestions and administrative help. This work was supported by the Wellcome Trust (092973/Z/10/Z to D.J.P.), Biotechnology and Biological Sciences Research Council (BBSRC) UK (BB/R017808/1 to D.J.P.), European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 646701 to B.S.-S.; StG_679173 to L.L.), Science Foundation Ireland (SFI) (16/FRL/3865 to L.L.), NIH (NS115064, HG008155, AG062377 to M.K.), NIH (R01 AI134861 and metabolic core grant S10 OD020100 to L.L.), Fundação Astrazeneca (Prémio FAZ Ciência 2019 to B.S.-S. and N.L.) and PAC-PRECISE LISBOA-01-0145-FEDER-016394, co-funded by FEDER (POR Lisboa 2020 (Programa Operacional Regional de Lisboa, do Portugal 2020)) and Fundação para a Ciência e a Tecnologia (Portugal). N.L. is supported by a postdoctoral fellowship from EMBO (ALTF 752-2018); S.M. was supported by a studentship from the Medical Research Council (MRC) UK; G.F. is supported by the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement no. 752932; and A.D., S.C., L.D. and H.P. are supported by Irish Research Council fellowships.

Author information

Authors and Affiliations

Authors

Contributions

N.L., C.M., S.M. and M.R. performed most of the experiments and analyzed the data. G.J.F. designed and performed some experiments. N.S., A.C.K., L.D., H.K., A.D., S.C., H.P., R.L. and C.C. provided technical assistance in some experiments. M.K. and L.Z.A. performed bioinformatic analysis, and M.B. provided reagents, materials and support. P.P. and R.J.A. provided key assistance with the SCENITHTM methodology. B.S.-S., D.J.P. and L.L. conceived and supervised the study. N.L., C.M., B.S.-S., D.J.P. and L.L. wrote the manuscript.

Corresponding author

Correspondence to Daniel J. Pennington.

Ethics declarations

Competing interests

B.S.-S. is an inventor of the patented ‘Delta One T cell’ technology, which has been acquired by GammaDelta Therapeutics (London, UK).

Additional information

Peer review information Nature Immunology thanks Juan Carlos Zúñiga-Pflücker and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 SCENITHTM methodology for analysis of cell metabolism.

a, Experimental design: E0771 breast or MC38 colon cancer cell lines were injected in WT mice; 6 and 15 days later, tumors were extracted for metabolic analysis of gd T cells using SCENITHTM. b, SCENITHTM assesses the impact of metabolic inhibitors on protein synthesis. Mean fluorescence intensity (MFI) of puromycin is analysed in each condition (Co: control-no inhibition; DG: 2-deoxyglucose inhibiting glycolysis; O: oligomycin inhibiting OXPHOS; and DGO: DG+O inhibitors). Glucose dependence, fatty acid and amino acid oxidation capacity, mitochondrial dependence and glycolytic capacity are calculated as detailed in the Methods and reference #23. Error bars show mean+SEM. Data are representative of 3 independent experiments (n=3 mice in triplicates per group and per experiment).

Extended Data Fig. 2 In vitro expanded γδ17 and γδIFN γδ T cells retain their mitochondrial and lipid phenotypes.

a, Representative flow plots of CD3 and TCRγδ expression on γδ17 and γδIFN T cells expanded in vitro from total spleen/LN cells. b, CD27 expression on in vitro expanded γδ17 and γδIFN γδ T cells. c, IL-17 and IFNγ production by in vitro expanded γδ17 and γδIFN T cells respectively, following activation with PMA/ionomycin. d, Vγ1 and Vγ4 expression on in vitro expanded γδ17 and γδIFN T cells. e, Representative staining of in vitro expanded γδ17 and γδIFN T cells for mitotracker, TMRM, lipidTOX and Bodipy-FL-C16. f, MFI of mitotracker, TMRM, lipidTOX and Bodipy-FL-C16 staining in vitro expanded γδ17 and γδIFN T cells. n=3, data representative of 3 independent experiments. Mitotracker p=0.0026; TMRM p=0.0003; LipidTOX p<0.0001; Bodipy FL-C16 p=0.036. Error bars show mean+SD, **p < 0.01, ***p<0.001, ****p < 0.0001, using two-tailed unpaired Student’s t-test.

Extended Data Fig. 3 γδTN cells can generate γδ17 and γδIFN T cells.

Flow cytometry profiles of thymic γδ T cells from E15 thymic lobes that had been cultured for 7-days in fetal thymic organ culture (E15 + 7dFTOC). CD24+ (γδ24+) precursors downregulate CD24 to become a CD24-CD44-CD45RB- (γδTN) population. γδTN cells are able to become either IL-17-secreting CD44+CD45RB- γδ17 cells, or IFN-γ-producing CD44+CD45RB+ γδIFN cells.

Extended Data Fig. 4 Thymic γδ17 cells are increased upon inhibition of glucose uptake.

Flow cytometry profiles of thymic γδTN (CD44-CD45RB-), γδ17 (CD44+CD45RB-) and γδIFN (CD44+CD45RB+) cells in γδ24- cells from E15 thymic lobes in 7-day FTOC with media containing or not Fasentin. Histograms show the number of γδ17 T cells (p<0.0001) and γδ17/γδIFN ratio (p=0.0028). Data are representative of 2 independent experiments (at least 4 lobes pooled per group per experiment). Error bars show mean±SEM, **p < 0.01, ****p < 0.0001, using two-tailed unpaired Student’s t-test.

Extended Data Fig. 5 Mitochondrial activity identifies Vγ4+ progenitors with distinct effector fates at very early stages.

a, Flow cytometry plots pre-sort, and after sorted TMRElo and TMREhi Vγ4+γδ24+ cells were cultured for 5-days on OP9DL1 cells. Percentage of thymic γδ17 and γδIFN cells generated are displayed in the graph on right. Data are representative of 3 independent experiments (cells sorted from n = 4 independent mice pooled per group per experiment). b, Flow cytometry plots for pre- and post-sort TMREhi and TMRElo Vγ4+γδTN cells that were cultured on OP9-DL1 cells for a further 5-days (plots on right). Histogram shows the percentage of each γδ T cell subset generated from cultured TMRElo and TMREhi Vγ4+γδTN cells. Error bars show mean + SD. Data are representative of 2 independent experiments (at least 4 lobes pooled per group per experiment). Error bars show mean±SD, *p < 0.05, ***p < 0.01, using two-tailed unpaired Student’s t-test.

Extended Data Fig. 6 Distinct mitochondrial activities underlie effector fate of thymic γδ T cell progenitors.

a, Experimental design for single-cell RNAseq (10x Genomics) on TMRElo and TMREhi gd24+ cells from E15 + 2d FTOC. b, Heatmap of differentially upregulated genes from comparison of TMRElo and TMREhi gd24+ cells. Genes are grouped in relation to their function in either OxPhos or glucose metabolism.

Extended Data Fig. 7 Enriched lipid metabolism and higher lipid uptake in γδ17 cells.

a, Experimental set up for bulk RNA-sequencing of PLZF+ (gd17) and PLZF (gdIFN) cells isolated from PLZF-GFP (Zbtb16GFP) mice. b, LipidTOX MFI in γδ17 (CD27-) and γδIFN (CD27+) T cells from LN cells activated in vitro with IL-1β+IL-23 and IL-12+IL-18 respectively. n=9, data pooled from 3 independent experiments. c, Representative plots of LipidTOX staining and IL-17A, IL-17F or RORγt expression in γδ27- T cells from LNs activated in vitro with IL-1β+IL-23 for 6h. Data representative of 3 independent experiments. d, Bodipy-FL-C16 MFI in γδ17 (CD27-) and γδIFN (CD27+) T cells T cells unstimulated or stimulated in vitro with IL-12+IL-18 or IL-1β+IL-23.(n=3, data from 1 experiment; γδ17 p= 0.0044; γδIFN p=0.8035). Error bars show mean+SD, **p < 0.01, ***p < 0.001, ****p < 0.0001 using one-way ANOVA.

Extended Data Fig. 8 Inhibition of dietary fat uptake reduces tumour growth and γδ17 cells in the tumour.

B16F10-tumour bearing mice were given daily injections of either vehicle or orlistat on days 6-9, and tumours were analysed on day 10. a, Percentage body weight following tumor cell injection; arrows indicate when orlistat or vehicle were administered. b, Tumor volume on days 8-10 following B16F10 inoculation. Absolute numbers c, and LipidTOX staining d, of tumor-infiltrating γδ17 cells on day 10. n=8 biologically independent animals, data from 1 independent experiment. Data represents mean+SD, *p<0.06, **p < 0.01 using unpaired Student’s t-test or one-way ANOVA.

Extended Data Fig. 9 Glucose supplementation diminishes γδ17 cell numbers and proliferation.

a, Flow cytometry profiles of peripheral γδ17 T cells cultured with media containing low (5mM) or high (50mM) doses of glucose. Graph depicts total numbers of γδ17 T cells (p=0.0028). b, Number of proliferating Ki-67+ γδ17 T cells cultured with low or high glucose (p=0.0034). n=6 biologically independent animals, data from 2 independent experiments. Error bars show mean±SEM, **p < 0.01, using unpaired two-tailed Student’s t-test.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lopes, N., McIntyre, C., Martin, S. et al. Distinct metabolic programs established in the thymus control effector functions of γδ T cell subsets in tumor microenvironments. Nat Immunol 22, 179–192 (2021). https://doi.org/10.1038/s41590-020-00848-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-00848-3

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