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:

CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors

Abstract

Depleting regulatory T cells (Treg cells) to counteract immunosuppressive features of the tumor microenvironment (TME) is an attractive strategy for cancer treatment; however, autoimmunity due to systemic impairment of their suppressive function limits its therapeutic potential. Elucidating approaches that specifically disrupt intratumoral Treg cells is direly needed for cancer immunotherapy. We found that CD36 was selectively upregulated in intrautumoral Treg cells as a central metabolic modulator. CD36 fine-tuned mitochondrial fitness via peroxisome proliferator-activated receptor-β signaling, programming Treg cells to adapt to a lactic acid-enriched TME. Genetic ablation of Cd36 in Treg cells suppressed tumor growth accompanied by a decrease in intratumoral Treg cells and enhancement of antitumor activity in tumor-infiltrating lymphocytes without disrupting immune homeostasis. Furthermore, CD36 targeting elicited additive antitumor responses with anti-programmed cell death protein 1 therapy. Our findings uncover the unexplored metabolic adaptation that orchestrates the survival and functions of intratumoral Treg cells, and the therapeutic potential of targeting this pathway for reprogramming the TME.

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 Treg cells elevate the expression of CD36 and genes involved in lipid metabolism.
Fig. 2: Disruption of CD36 selectively impairs the accumulation and suppressive function of intratumoral Treg cells.
Fig. 3: CD36 expression selectively supports suppressive activity of intratumoral Treg cells.
Fig. 4: CD36 deficiency stimulates apoptosis in intratumoral Treg cells.
Fig. 5: PPAR-β signaling is required for metabolic adaptation in intratumoral Treg cells.
Fig. 6: CD36 targeting impairs intratumoral Treg cells and primes tumors to PD-1 blockade.

Similar content being viewed by others

Data availability

The RNA-Seq data for intratumoral Treg cells are available in the Gene Expression Omnibus database under accession code GSE139325. All relevant data are available from the corresponding author upon request.

References

  1. Roychoudhuri, R., Eil, R. L. & Restifo, N. P. The interplay of effector and regulatory T cells in cancer. Curr. Opin. Immunol. 33, 101–111 (2015).

    CAS  PubMed  Google Scholar 

  2. Delgoffe, G. M. et al. Stability and function of regulatory T cells is maintained by a neuropilin-1–semaphorin-4a axis. Nature 501, 252–256 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Saito, T. et al. Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat. Med. 22, 679–684 (2016).

    CAS  PubMed  Google Scholar 

  4. Rech, A. J. et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci. Transl. Med. 4, 134ra162 (2012).

    Google Scholar 

  5. Sutmuller, R. P. et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25+ regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J. Exp. Med. 194, 823–832 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Marabelle, A. et al. Depleting tumor-specific Treg cells at a single site eradicates disseminated tumors. J. Clin. Invest. 123, 2447–2463 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Teng, M. W. et al. Conditional regulatory T-cell depletion releases adaptive immunity preventing carcinogenesis and suppressing established tumor growth. Cancer Res. 70, 7800–7809 (2010).

    CAS  PubMed  Google Scholar 

  8. Walter, S. et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat. Med. 18, 1254–1261 (2012).

    CAS  PubMed  Google Scholar 

  9. Nishikawa, H. & Sakaguchi, S. Regulatory T cells in tumor immunity. Int. J. Cancer 127, 759–767 (2010).

    CAS  PubMed  Google Scholar 

  10. Simpson, T. R. et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 210, 1695–1710 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Curtin, J. F. et al. Treg depletion inhibits efficacy of cancer immunotherapy: implications for clinical trials. PLoS ONE 3, e1983 (2008).

    PubMed  PubMed Central  Google Scholar 

  12. Arce Vargas, F. et al. Fc effector function contributes to the activity of human anti-CTLA-4 antibodies. Cancer Cell 33, 649–663.e4 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Miragaia, R. J. et al. Single-cell transcriptomics of regulatory T cells reveals trajectories of tissue adaptation. Immunity 50, 493–504.e7 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Arpaia, N. et al. A distinct function of regulatory T cells in tissue protection. Cell 162, 1078–1089 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Burzyn, D. et al. A special population of regulatory T cells potentiates muscle repair. Cell 155, 1282–1295 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Cipolletta, D. et al. PPAR-γ is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 486, 549–553 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Panduro, M., Benoist, C. & Mathis, D. Tissue Treg cells. Annu. Rev. Immunol. 34, 609–633 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Vignali, D. A., Collison, L. W. & Workman, C. J. How regulatory T cells work. Nat. Rev. Immunol. 8, 523–532 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Chaudhry, A. & Rudensky, A. Y. Control of inflammation by integration of environmental cues by regulatory T cells. J. Clin. Invest. 123, 939–944 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Campbell, D. J. & Koch, M. A. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat. Rev. Immunol. 11, 119–130 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zeng, H. et al. mTORC1 couples immune signals and metabolic programming to establish Treg-cell function. Nature 499, 485–490 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, K. et al. Homeostatic control of metabolic and functional fitness of Treg cells by LKB1 signalling. Nature 548, 602–606 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Huynh, A. et al. Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability. Nat. Immunol. 16, 188–196 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Gerriets, V. A. et al. Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat. Immunol. 17, 1459–1466 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Weinberg, S. E. et al. Mitochondrial complex III is essential for suppressive function of regulatory T cells. Nature 565, 495–499 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Angelin, A. et al. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. 25, 1282–1293 e1287 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, H., Franco, F. & Ho, P. C. Metabolic regulation of Treg cells in cancer: opportunities for immunotherapy. Trends Cancer 3, 583–592 (2017).

    CAS  PubMed  Google Scholar 

  28. Li, X. et al. Navigating metabolic pathways to enhance antitumour immunity and immunotherapy. Nat. Rev. Clin. Oncol. 16, 425–441 (2019).

    CAS  PubMed  Google Scholar 

  29. Plitas, G. et al. Regulatory T cells exhibit distinct features in human breast cancer. Immunity 45, 1122–1134 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Piconese, S. et al. Human OX40 tunes the function of regulatory T cells in tumor and nontumor areas of hepatitis C virus-infected liver tissue. Hepatology 60, 1494–1507 (2014).

    CAS  PubMed  Google Scholar 

  31. Zappasodi, R. et al. Rational design of anti-GITR-based combination immunotherapy. Nat. Med. 25, 759–766 (2019).

    CAS  PubMed  Google Scholar 

  32. He, N. et al. Metabolic control of regulatory T cell (Treg) survival and function by Lkb1. Proc. Natl Acad. Sci. USA 114, 12542–12547 (2017).

    CAS  PubMed  Google Scholar 

  33. Beier, U. H. et al. Essential role of mitochondrial energy metabolism in Foxp3+ T-regulatory cell function and allograft survival. FASEB J. 29, 2315–2326 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Ho, P. C. et al. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 162, 1217–1228 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Siska, P. J. & Rathmell, J. C. T cell metabolic fitness in antitumor immunity. Trends Immunol. 36, 257–264 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Vannini, N. et al. The NAD-booster nicotinamide riboside potently stimulates hematopoiesis through increased mitochondrial clearance. Cell Stem Cell 24, 405–418 e407 (2019).

    CAS  PubMed  Google Scholar 

  37. Ravnskjaer, K. et al. PPARδ is a fatty acid sensor that enhances mitochondrial oxidation in insulin-secreting cells and protects against fatty acid-induced dysfunction. J. Lipid Res. 51, 1370–1379 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Morino, K. et al. Regulation of mitochondrial biogenesis by lipoprotein lipase in muscle of insulin-resistant offspring of parents with type 2 diabetes. Diabetes 61, 877–887 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Fan, W. et al. PPARδ promotes running endurance by preserving glucose. Cell Metab. 25, 1186–1193.e4 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Pascual, G. et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 541, 41–45 (2017).

    CAS  PubMed  Google Scholar 

  41. Ladanyi, A. et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 37, 2285–2301 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Miska, J. et al. HIF-1α is a metabolic switch between glycolytic-driven migration and oxidative phosphorylation-driven immunosuppression of Treg cells in glioblastoma. Cell Rep. 27, 226–237.e4 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Phan, A. T., Goldrath, A. W. & Glass, C. K. Metabolic and epigenetic coordination of T cell and macrophage immunity. Immunity 46, 714–729 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Liu, P. S. & Ho, P. C. Mitochondria: a master regulator in macrophage and T cell immunity. Mitochondrion 41, 45–50 (2018).

    CAS  PubMed  Google Scholar 

  45. Pacella, I. et al. Fatty acid metabolism complements glycolysis in the selective regulatory T cell expansion during tumor growth. Proc. Natl Acad. Sci. USA 115, E6546–E6555 (2018).

    CAS  PubMed  Google Scholar 

  46. Son, N. H. et al. Endothelial cell CD36 optimizes tissue fatty acid uptake. J. Clin. Invest. 128, 4329–4342 (2018).

    PubMed  PubMed Central  Google Scholar 

  47. Dammone, G. et al. PPARγ controls ectopic adipogenesis and cross-talks with myogenesis during skeletal muscle regeneration. Int. J. Mol. Sci. 19, E2044 (2018).

    PubMed  Google Scholar 

  48. Meeth, K., Wang, J. X., Micevic, G., Damsky, W. & Bosenberg, M. W. The YUMM lines: a series of congenic mouse melanoma cell lines with defined genetic alterations. Pigment Cell Melanoma Res. 29, 590–597 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Ho, P. C. et al. Immune-based antitumor effects of BRAF inhibitors rely on signaling by CD40L and IFNγ. Cancer Res. 74, 3205–3217 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Driscoll, W. S., Vaisar, T., Tang, J., Wilson, C. L. & Raines, E. W. Macrophage ADAM17 deficiency augments CD36-dependent apoptotic cell uptake and the linked anti-inflammatory phenotype. Circ. Res. 113, 52–61 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Cheng, W. C. et al. Uncoupling protein 2 reprograms the tumor microenvironment to support the anti-tumor immune cycle. Nat. Immunol. 20, 206–217 (2019).

    CAS  PubMed  Google Scholar 

  52. Picelli, S. et al. Full-length RNA-Seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    CAS  PubMed  Google Scholar 

  53. Nelson, J. W., Sklenar, J., Barnes, A. P. & Minnier, J. The START App: a web-based RNAseq analysis and visualization resource. Bioinformatics 33, 447–449 (2017).

    CAS  PubMed  Google Scholar 

  54. Liu, P. S. et al. α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 18, 985–994 (2017).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank L.-F. Lu and W.-L. Lo for critical reading and comments. We also thank Y. Maeda and H. Nishikawa for helpful discussion. P.-C.H. was supported in part by the SNSF (project grants 31003A_163204 and 31003A_182470), the Swiss Cancer Foundation (KFS-3949-08-2016), the Swiss Institute for Experimental Cancer Research (ISREC 26075483), a European Research Council Staring Grant (802773-MitoGuide), the Cancer Research Institute Clinic and Laboratory Integration Program award and the SITC-MRA Young Investigator Award. C.J. is supported by the SNSF (project grants PMPDP3_129022 and PZ00P3_161459). A.Z. is supported by the SNSF (project grant 320030_162575) and Cancer League Switzerland (KFS-3394-02-2014). R.S. and I.G. are supported by NIH funding (P01 HL46403, P01 HL087018 and R01 HL142152 to R.S. and HL45095 and HL073029 to I.G.). E.M. acknowledges funding from the Swiss Cancer Research Foundation (KFS-3681-08-2015-R). S.-M.F. acknowledges funding from an FWO grant and projects, as well as KU Leuven Methusalem co-funding. J.F.-G. is supported by an FWO postdoctoral fellowship. J.D.W and T.M. are supported by NIH funding (P30 CA008748 and R01 CA056821), Swim Across America, the Ludwig Institute for Cancer Research, the Parker Institute for Cancer Immunotherapy and the Breast Cancer Research Foundation. R.Z. is supported by the Parker Institute for Cancer Immunotherapy Bridge Scholar Award. We also appreciate the support provided by the Electron Microscopy Facility at the University of Lausanne and the Biomedical Sequencing Facility at the Research Center for Molecular Medicine of the Austrian Academy of Sciences.

Author information

Authors and Affiliations

Authors

Contributions

H.W. and P.-C.H. contributed to overall project design and wrote the manuscript. H.W., F.F., Y.-C.T., C.-H.T. and F.P. performed the in vitro and in vivo animal works and data analysis. X.X. and S.R.M. performed analysis of the RNA-Seq results. H.W., M.P.T., R.Z., J.D.W., T.M., C.J., I.S. and A.Z. conducted collection and flow cytometry analysis of the human samples. J.F.-G. and S.-M.F. supported the metabolomite analysis. R.S. and I.G. provided the hybridoma clone for anti-CD36 antibody production and CD36flox mice, respectively. E.M. provided samples of NSCLC murine models.

Corresponding author

Correspondence to Ping-Chih Ho.

Ethics declarations

Competing interests

H.W. and P.-C.H. are inventors on a patent application related to targeting CD36 in cancer immunotherapy. P.-C.H. is serving as a member of the scientific advisory board for Elixiron Immunotherapeutics and receiving research grants from Roche and Idorsia. J.D.W. is serving as a consultant for Adaptive Biotechnologies, Advaxis, Amgen, Apricity, Array BioPharma, Ascentage Pharma, Astellas, Bayer, BeiGene, Bristol-Myers Squibb. Celgene, Chugai, Elucida, Eli Lilly, F-Star, Genentech, Imvaq, Janssen, Kyowa Hakko Kirin, Kleo Pharmaceuticals, Linnaeus, MedImmune, Merck, Neon Therapeutics, Northern Biologics, Ono, Polaris Pharma, Polynoma, PsiOxus, PureTech, Recepta, Takara Bio, Trieza, Sellas Life Sciences, Serametrix, Surface Oncology, Syndax and Synthologic. J.D.W. received research support from Bristol-Myers Squibb, MedImmune, Merck and Genentech and has equity in Potenza Therapeutics, Tizona Pharmaceuticals, Adaptive Biotechnologies, Elucida, Imvaq, BeiGene, Trieza and Linnaeus. P.-C.H. received an honorarium from Pfizer and Chugai. J.D.W. received an honorarium from Esanex.

Additional information

Peer review information 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 Lipid accumulation and increased CD36 expression in intratumoral Treg cells.

a, b, Quantitative results of geometric mean (GeoMean) fluorescent intensity of Bodipy FL C12 (a) and Bodipy 493/503 (Bodipy) (b) in Treg cells from paired TILs and PBMCs of non-small cell lung cancer (NSCLC) patients (n=6 per group). c, d, Quantitative results of GeoMean fluorescent intensity of Bodipy FL C12 (c) and Bodipy 493/503 (d) in Treg cells from paired PBMC and tumor infiltrated lymph nodes (TILNs) of melanoma patients (n=19). e, f, Quantitative results of fluorescent intensity of Bodipy FL C12 in Treg cells from indicated tissues of B16 melanoma-bearing mice (dLN, n=7; Spleen, n=6; Thymus, n=7; Tumor, n=6) (e), and MC38 colon carcinoma-bearing mice (n=8) (f). g, Quantitative result of GeoMean fluorescent intensity of CD36 surface staining in Treg cells from paired TILs and PBMCs of NSCLC patients (n=6) h, i, j, k, Quantitative results of surface expression of CD36 in Treg cells of indicated tissues from B16-OVA melanoma-bearing B6 mice (n=6, one outlier was removed from dLN) (h), inducible Braf/Pten melanoma-bearing mice (n=9) (i), K-rasLSL-G12D/+/p53fl/fl mouse model of NSCLC (Blood, n=8; Tumor, n=13) (j), and MC38 colon cancer (n=8) (k). l, CD36 expression in iTreg cells cultured in different indicated conditions for 48h. (RPMI: normal cell culture RPMI 1640 medium indicated in methods; CM, cancer cell conditioned medium, n=4 per group). m, CD36 expression in iTreg cells cultured in cancer cell-conditioned medium treated with control procedure or lipid removal procedure for 48h. (n=6 per group). Data are representative result of at least two independent experiments with similar results (l, m) or cumulative results from at least two independent experiments (a, b, c, d, e, f, g, h, i, j, k). Each symbol represents one individual. Data are mean ± S.D. and were analyzed by two-tailed, unpaired Student’s t-test (e, f, h, i, j, k, l, m) or two-tailed, paired Student’s t-test (a, b) or one-tailed, paired Student’s t-test (c, d, g).

Source data

Extended Data Fig. 2 CD36 expression supports the accumulation and suppressive function of intratumoral Treg cells.

a, Body weight of WT and TregCd36-/- mice at the age of 21–23 weeks (WT male, n=4; TregCd36-/- male, n=6; WT female, n=6; TregCd36-/- female, n=5;). b, Representative plots (left) and quantitative frequency of CD44hi/CD62Llow CD4+ or CD8+ T cells (right) in aged WT and TregCd36-/- mice (n=7 per group). c, Representative images of hematoxylin and eosin (H&E) staining for indicated tissues from WT or TregCd36-/- mice at the age of 21–23 weeks. Scale bar, 200 µm. d, e, Tumor growth of B16-OVA melanoma (n=7 per group) (d) or MC38 colon carcinoma (n=6 per group) (e) from WT or TregCd36-/- mice. f, g, h, Absolute number of FoxP3+ Treg cells per gram tumor (f), percentage of CD8+ T cells out of CD3+ T cells among tumor-infiltrating T cells (g), and the ratio of CD8+ to Treg cell TIL density (h) of YUMM1.7 melanoma-bearing WT and TregCd36-/- mice (n=11 per group). i, Representative plots (left) and percentage of indicated cytokine-producing CD4+ T cells among total tumor-infiltrating CD4+ T cells from indicated mice (right) (n=5 per group). Data are representative result of at least two independent experiments with similar results (c, d, e, i) or cumulative results from at least two independent experiments (a, b, f, g, h) Each symbol represents one individual. Data are mean ± S.D. (a, b, f, g, h, i) or ± S.E.M. (d, e) and were analyzed by two-tailed, unpaired Student’s t-test.

Source data

Extended Data Fig. 3 Effects of CD36 in expression of activation markers and stability of intratumoral Treg cells.

a, Representative images of guts (a) and spleens (b) from indicated group of Rag1-/- mice. c, d, e, Expression of CD44 (n=17) (c), CD103 (n=5) (d), and KLRG1 (n=5) (e) in intratumoral Treg cells of YUMM1.7 melanoma-bearing WT and TregCd36-/- mice. f, The expression of YFP in intratumoral Treg cells of YUMM1.7 melanoma-bearing WT and TregCd36-/- mice (n=15). g, h, i, Representative plots of IFNγ and TNF production among total intratumoral Treg cells from indicated mice (g), and quantitative result of percentage of IFNγ-producing (n=19 per group) (h) and TNF-producing (n=18, one outlier was removed from TregCd36-/-) (i) Treg cells among total intratumoral Treg cells of indicated mice. j, Expression of Ki67 in intratumoral Treg cells of YUMM1.7 melanoma-bearing WT and TregCd36-/- mice (n=10 per group). k, Representative histograms (left) and quantitative analysis (right) of Annexin V staining in intratumoral Treg cells from WT and TregCd36-/- tumor-bearing mice (n=14 per group). l, Quantitative analysis of cleaved caspase-3 levels in Treg cells of indicated tissues from WT (n=13 per group except for thymus, n=9) and TregCd36-/- (n=14 per group except for thymus, n=9) tumor-bearing mice. LN: non-draining lymph node; DLN: draining lymph node. Data are representative results of two independent experiments with similar results (a, b, d, e) or cumulative results from at least three independent experiments (c, f, g, h, i, j, k, l). Each symbol represents one individual. Data are mean ± S.D. (c, d, e, f, h, i, j, k, l) and were analyzed by two-tailed, unpaired Student’s t-test.

Source data

Extended Data Fig. 4 CD36-deficiency results in a metabolic shift and elevated apoptosis in Treg cells.

a, Indicated iTreg cells cultured in cancer cell-conditioned medium for 48 hrs (n=3 per group). Oxygen consumption rate (OCR) of indicated iTreg cells was measured and then followed by treatment with oligomycin, FCCP, and antimycin A plus Rotenone (n≥4 per group). b, Indicated iTreg cells cultured in cancer cell-conditioned medium for 48 hrs (n≥4 per group) and then media were refreshed with Seahorse Flux assay media without glucose. Basel extracellular acidification rate (ECAR) of indicated iTreg cells was measured and then followed by treatment with glucose, oligomycin, FCCP and 2-DG (n=4 per group). c, Quantitative result of glycolysis and glycolytic capacity based on the measurement of b. d, The viability of either WT or TregCd36-/- iTreg cells cultured under indicated conditions for 72 hrs (n=6 per group). Data are representative results of three independent experiments with similar results (a, b, c, d). Data are mean ± S.D. and were analyzed by two-tailed, unpaired Student’s t-test (c, d).

Source data

Extended Data Fig. 5 Intratumoral Treg cells require PPAR-β, not PPAR-γ, signaling for metabolic adaptation.

a, Enrichment plots of signals controlling mitochondrial matrix (left) and mitochondrial envelope in intratumoral Treg (n=4) compared to PBMC Treg cells (n=6), identified by GSEA computational method. ES: enrichment score; NES: normalized enrichment score; FDR: false discovery rate; NOM p-val: Nominal p value. b, c, d, Percentage of FoxP3+ Treg cells among CD4+ tumor-infiltrating T lymphocytes (n=5) (b), tumor growth (n=5) (c) and tumor weight (n=7) (d) from tumor-bearing WT and TregPPARγ-/- mice. e, Percentage of CD8+ T cells among tumor-infiltrating T cells from tumor-bearing WT and TregPPARβ-/- mice (n=10). f, Quantitative result of CD36+ intratumoral Treg cells from YUMM1.7 melanoma-bearing WT and TregPPARβ-/- mice (WT, n=14; TregPPARβ-/-, n=11). g, NAD/NADH ratio of indicated iTreg cells cultured in cancer cell-conditioned medium with DMSO or PPAR-β agonist for 48h (DMSO, n=8; PPAR-β agonist, n=10). Data are representative results of at least two independent experiments with similar results (b, c, d) or cumulative results from at least two independent experiments (e, f, g). Each symbol represents one individual. Data are mean ± S.D. (b, d, e, f, g) or ± S.E.M. (c) and were analyzed by two-tailed, unpaired Student’s t-test.

Source data

Extended Data Fig. 6 CD36-targeting unleashes host antitumor immunity.

a, b, c, d, Absolute number of FoxP3+ Treg cells per gram tumor (n=10 per group) (a), percentage of CD8+ T cells among tumor-infiltrating T cells (n=10 per group) (b) and representative plots and percentage of indicated cytokine-producing CD8+ T cells among total tumor-infiltrating CD8+ T cells (c) and CD4+ T cells among total tumor-infiltrating CD4+ T cells (d) from YUMM1.7 melanoma-bearing mice treated with indicated treatments (n=10 per group). e, f, Tumor growth (e) and survival curves (f) of YUMM1.7 melanoma-bearing B6 mice treated with indicated treatments (Ctrl, n = 10; α-PD1, n = 10; α-CD36, n = 11; α-CD36 + αPD-1, n = 11). Arrows indicate the date of treatment. Dotted lines indicate the tumor volume of 800 mm3. Data are cumulative results from at least two independent experiments. Each symbol represents one individual. Data are mean ± S.D. and were analyzed by two-tailed, unpaired Student’s t-test (a, b, c, d). Difference between survival curves was analyzed by Log-rank (Mantel-Cox) test (f).

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 6

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, H., Franco, F., Tsui, YC. et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat Immunol 21, 298–308 (2020). https://doi.org/10.1038/s41590-019-0589-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0589-5

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