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.

  • Review Article
  • Published:

CD8+ T cell metabolism in infection and cancer

Abstract

Cytotoxic CD8+ T cells play a key role in the elimination of intracellular infections and malignant cells and can provide long-term protective immunity. In the response to infection, CD8+ T cell metabolism is coupled to transcriptional, translational and epigenetic changes that are driven by extracellular metabolites and immunological signals. These programmes facilitate the adaptation of CD8+ T cells to the diverse and dynamic metabolic environments encountered in the circulation and in the tissues. In the setting of disease, both cell-intrinsic and cell-extrinsic metabolic cues contribute to CD8+ T cell dysfunction. In addition, changes in whole-body metabolism, whether through voluntary or disease-induced dietary alterations, can influence CD8+ T cell-mediated immunity. Defining the metabolic adaptations of CD8+ T cells in specific tissue environments informs our understanding of how these cells protect against pathogens and tumours and maintain tissue health at barrier sites. Here, we highlight recent findings revealing how metabolic networks enforce specific CD8+ T cell programmes and discuss how metabolism is integrated with CD8+ T cell differentiation and function and determined by environmental cues.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Organismal, niche and intrinsic metabolism dictate CD8+ T cell fate and function.
Fig. 2: Metabolism of CD8+ T cell activation and effector function.
Fig. 3: Metabolism of memory CD8+ T cell differentiation.
Fig. 4: Metabolism of tissue-resident memory CD8+ T cells.
Fig. 5: Metabolic insufficiency in chronic infection and cancer.

Similar content being viewed by others

References

  1. Kaech, S. M. & Cui, W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Jang, C. et al. Metabolite exchange between mammalian organs quantified in pigs. Cell Metab. 30, 594–606.e3 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Reina-Campos, M., Moscat, J. & Diaz-Meco, M. Metabolism shapes the tumor microenvironment. Curr. Opin. Cell Biol. 48, 47–53 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Troha, K. & Ayres, J. S. Metabolic adaptations to infections at the organismal level. Trends Immunol. 41, 113–125 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Haws, S. A., Leech, C. M. & Denu, J. M. Metabolism and the epigenome: a dynamic relationship. Trends Biochem. Sci. 45, 731–737 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Roos, D. & Loos, J. A. Changes in the carbohydrate metabolism of mitogenically stimulated human peripheral lymphocytes. Exp. Cell Res. 77, 127–135 (1973). This is an early landmark study exploring the metabolism of lymphocytes. It uses phytohaemagglutinin-activated human blood lymphocytes to quantify ATP production derived from glycolysis and oxidative phosphorylation.

    Article  CAS  PubMed  Google Scholar 

  8. Greiner, E. F., Guppy, M. & Brand, K. Glucose is essential for proliferation and the glycolytic enzyme induction that provokes a transition to glycolytic energy production. J. Biol. Chem. 269, 31484–31490 (1994).

    Article  CAS  PubMed  Google Scholar 

  9. Bauer, D. E. et al. Cytokine stimulation of aerobic glycolysis in hematopoietic cells exceeds proliferative demand. FASEB J. 18, 1303–1305 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. van der Windt, G. J. W. et al. Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell memory development. Immunity 36, 68–78 (2012).

    Article  PubMed  CAS  Google Scholar 

  12. Van Der Windt, G. J. W. et al. CD8 memory T cells have a bioenergetic advantage that underlies their rapid recall ability. Proc. Natl Acad. Sci. USA 110, 14336–14341 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Fox, C. J., Hammerman, P. S. & Thompson, C. B. Fuel feeds function: energy metabolism and the T-cell response. Nat. Rev. Immunol. 5, 844–852 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Pearce, E. L., Poffenberger, M. C., Chang, C. & Jones, R. G. Fueling immunity: insights into metabolism and lymphocyte function. Science 342, 1242454 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  16. 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). This study highlights the direct link between T cell activation and metabolism, showing TCR signalling tyrosine kinases directly interacting with the glycolytic enzyme PDHK1, as well as how glycolytic enzymes can moonlight as effector cytokine mRNA regulatory molecules, holding mRNA when T cells are quiescent and releasing it for translation when aerobic glycolysis is occurring.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gubser, P. M. et al. Rapid effector function of memory CD8+ T cells requires an immediate-early glycolytic switch. Nat. Immunol. 14, 1064–1072 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Pfeiffer, T., Schuster, S. & Bonhoeffer, S. Cooperation and competition in the evolution of ATP-producing pathways. Science 292, 504–507 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Cammann, C. et al. Early changes in the metabolic profile of activated CD8+ T cells. BMC Cell Biol. 17, 28 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Jacobs, S. R. et al. Glucose uptake is limiting in T cell activation and requires CD28-mediated Akt-dependent and independent pathways. J. Immunol. 180, 4476–4486 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Song, W., Li, D., Tao, L., Luo, Q. & Chen, L. Solute carrier transporters: the metabolic gatekeepers of immune cells. Acta Pharm. Sin. B 10, 61–78 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Wofford, J. a et al. IL-7 promotes Glut1 trafficking and glucose uptake via STAT5-mediated activation of Akt to support T cell survival. Blood 111, 2101–2112 (2007).

    Article  PubMed  CAS  Google Scholar 

  23. Tsai, S. et al. Insulin receptor-mediated stimulation boosts T cell immunity during inflammation and infection. Cell Metab. 28, 922–934.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Saucillo, D. C., Gerriets, V. A., Sheng, J., Rathmell, J. C. & Maciver, N. J. Leptin metabolically licenses T cells for activation to link nutrition and immunity. J. Immunol. 192, 136–144 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Rivadeneira, D. B. et al. Oncolytic viruses engineered to enforce leptin expression reprogram tumor-infiltrating T cell metabolism and promote tumor clearance. Immunity 51, 548–560.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, R. et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35, 871–882 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Klein-Hessling, S. et al. NFATc1 controls the cytotoxicity of CD8+ T cells. Nat. Commun. 8, 511 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Carr, E. L. et al. Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J. Immunol. 185, 1037–1044 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Fotiadis, D., Kanai, Y. & Palacín, M. The SLC3 and SLC7 families of amino acid transporters. Mol. Asp. Med. 34, 139–158 (2013).

    Article  CAS  Google Scholar 

  30. Sinclair, L. V. et al. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat. Immunol. 14, 500–508 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sinclair, L. V. et al. Antigen receptor control of methionine metabolism in T cells. eLife 8, 1–29 (2019).

    Article  Google Scholar 

  32. Bosshart, P. D., Kalbermatter, D., Bonetti, S. & Fotiadis, D. Mechanistic basis of L-lactate transport in the SLC16 solute carrier family. Nat. Commun. 10, 2649 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Brand, A. et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24, 657–671 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Powell, J. D. & Delgoffe, G. M. The mammalian target of rapamycin: linking T cell differentiation, function, and metabolism. Immunity 33, 301–311 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Charpentier, J. C. et al. Macropinocytosis drives T cell growth by sustaining the activation of mTORC1. Nat. Commun. 11, 180 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ma, E. H. et al. Serine is an essential metabolite for effector T cell expansion. Cell Metab. 25, 345–357 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Ma, E. H. et al. Metabolic profiling using stable isotope tracing reveals distinct patterns of glucose utilization by physiologically activated CD8+ T cells. Immunity 51, 856–870.e5 (2019). This study shows for the first time in vivo the patterns of glucose utilization by T cells in the context of physiological activation with Listeria infection, highlighting how in vitro T cell metabolic measurements do not fully capture the metabolic state of T cells in vivo, bringing into question conclusions derived from previous in vitro-centric metabolic studies.

    Article  CAS  PubMed  Google Scholar 

  38. Tan, H. et al. Integrative proteomics and phosphoproteomics profiling reveals dynamic signaling networks and bioenergetics pathways underlying T cell activation. Immunity 46, 488–503 (2017). This in-depth analysis of T cell whole proteome and phosphoproteome dynamics upon T cell activation provides a resource for understanding not only the early kinetics of T cell metabolism but also the essential link between mitochondrial function, carbon anabolism and cellular differentiation upon T cell activation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Muri, J. et al. The thioredoxin-1 system is essential for fueling DNA synthesis during T-cell metabolic reprogramming and proliferation. Nat. Commun. 9, 1851 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. 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 

  41. Pollizzi, K. N. et al. Asymmetric inheritance of mTORC1 kinase activity during division dictates CD8+ T cell differentiation. Nat. Immunol. 17, 704–711 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Verbist, K. C. et al. Metabolic maintenance of cell asymmetry following division in activated T lymphocytes. Nature 532, 389–393 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chisolm, D. A. & Weinmann, A. S. Connections between metabolism and epigenetics in programming cellular differentiation. Annu. Rev. Immunol. 36, 221–246 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Lee, J. et al. Regulator of fatty acid metabolism, acetyl coenzyme A carboxylase 1, controls T cell immunity. J. Immunol. 192, 3190–3199 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Ibitokou, S. A. et al. Early inhibition of fatty acid synthesis reduces generation of memory precursor effector T cells in chronic infection. J. Immunol. 200, 643–656 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Thurnher, M. & Gruenbacher, G. T lymphocyte regulation by mevalonate metabolism. Sci. Signal. 8, re4 (2015).

    Article  PubMed  CAS  Google Scholar 

  47. 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 

  48. Turner, M. & Díaz-Muñoz, M. D. RNA-binding proteins control gene expression and cell fate in the immune system. Nat. Immunol. 19, 120–129 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Zurli, V. et al. Phosphoproteomics of CD2 signaling reveals AMPK-dependent regulation of lytic granule polarization in cytotoxic T cells. Sci. Signal. 13, eaaz1965 (2020).

    Article  CAS  PubMed  Google Scholar 

  50. Chow, C.-W., Rincón, M. & Davis, R. J. Requirement for transcription factor NFAT in interleukin-2 expression. Mol. Cell. Biol. 19, 2300–2307 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. O’Sullivan, D. et al. Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 49, 375–376 (2014).

    Article  CAS  Google Scholar 

  53. Pearce, E. L. et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460, 103–107 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cui, G. et al. IL-7-induced glycerol transport and TAG synthesis promotes memory CD8+ T cell longevity. Cell 161, 750–761 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Buck, M. D. D. et al. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell 166, 63–76 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Saibil, S. D. et al. Activation of peroxisome proliferator-activated receptors α and δ synergizes with inflammatory signals to enhance adoptive cell therapy. Cancer Res. 79, 445–451 (2019).

    Article  CAS  PubMed  Google Scholar 

  57. Pollizzi, K. N. et al. mTORC1 and mTORC2 selectively regulate CD8+ T cell differentiation. J. Clin. Invest. 125, 2090–2108 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Raud, B. et al. Etomoxir actions on regulatory and memory T cells are independent of Cpt1a-mediated fatty acid oxidation. Cell Metab. 28, 504–515.e7 (2018). This study shows how loss of CPT1α, an essential enzyme for fatty acid oxidation, does not affect T cell homeostasis or memory formation, calling into question previous work showing memory T cells require fatty acid oxidation by using the CPR1α inhibitor etomoxir, where previous studies may have drawn conclusions from off-target effects.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Phan, A. T. et al. Constitutive glycolytic metabolism supports CD8+ T cell effector memory differentiation during viral infection. Immunity 45, 1024–1037 (2016). This study provides the first in vivo evidence that enforcing constitutive glycolytic metabolism through VHL deletion in CD8+ T cells does not limit, but increases, circulating memory CD8+ T cell differentiation in response to acute viral infection.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Xu, Y. et al. Glycolysis determines dichotomous regulation of T cell subsets in hypoxia. J. Clin. Invest. 126, 2678–2688 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Ma, R. et al. A Pck1-directed glycogen metabolic program regulates formation and maintenance of memory CD8+ T cells. Nat. Cell Biol. 20, 21–27 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Ghergurovich, J. M. et al. A small molecule G6PD inhibitor reveals immune dependence on pentose phosphate pathway. Nat. Chem. Biol. 16, 731–739 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gnanaprakasam, J. N. R., Wu, R. & Wang, R. Metabolic reprogramming in modulating T cell reactive oxygen species generation and antioxidant capacity. Front. Immunol. 16, 1075 (2018).

    Article  CAS  Google Scholar 

  64. Hermans, D. et al. Lactate dehydrogenase inhibition synergizes with IL-21 to promote CD8+ T cell stemness and antitumor immunity. Proc. Natl Acad. Sci. USA 117, 6047–6055 (2020). This study uncovers the metabolic reprogramming endowing the prostemness effect of IL-21 on CD8+ T cells and shows its synergy with LDH inhibition in promoting CD8+ T cell antitumour immunity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Bantug, G. R. et al. Mitochondria-endoplasmic reticulum contact sites function as immunometabolic hubs that orchestrate the rapid recall response of memory CD8+ T cells. Immunity 48, 542–555.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Simula, L. et al. Drp1 controls effective T cell immune-surveillance by regulating T cell migration, proliferation, and cMyc-dependent metabolic reprogramming. Cell Rep. 25, 3059–3073.e10 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Cogliati, S. et al. Mitochondrial cristae shape determines respiratory chain supercomplexes assembly and respiratory efficiency. Cell 155, 160–171 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Baixauli, F. et al. The mitochondrial fission factor dynamin-related protein 1 modulates T-cell receptor signalling at the immune synapse. EMBO J. 30, 1238–1250 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Champagne, D. P. et al. Fine-tuning of CD8+ T cell mitochondrial metabolism by the respiratory chain repressor MCJ dictates protection to influenza virus. Immunity 44, 1299–1311 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. He, S. et al. Characterization of the metabolic phenotype of rapamycin-treated CD8+ T cells with augmented ability to generate long-lasting memory cells. PLoS ONE 6, e20107 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Araki, K. et al. mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Rao, R. R., Li, Q., Odunsi, K. & Shrikant, P. A. The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and eomesodermin. Immunity 32, 67–78 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Sukumar, M. et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Invest. 123, 4479–4488 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Liu, G. Y. & Sabatini, D. M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol. 21, 183–203 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. O’Brien, T. F. et al. Regulation of T-cell survival and mitochondrial homeostasis by TSC1. Eur. J. Immunol. 41, 3361–3370 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Yang, K., Neale, G., Green, D. R., He, W. & Chi, H. The tumor suppressor Tsc1 enforces quiescence of naive T cells to promote immune homeostasis and function. Nat. Immunol. 12, 888–897 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Shrestha, S. et al. Tsc1 promotes the differentiation of memory CD8+ T cells via orchestrating the transcriptional and metabolic programs. Proc. Natl Acad. Sci. USA 111, 14858–14863 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Zhang, L. et al. TSC1/2 signaling complex is essential for peripheral naïve CD8+ T cell survival and homeostasis in mice. PLoS ONE 7, e30592 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Zhang, L. et al. Mammalian target of rapamycin complex 2 controls CD8 T cell memory differentiation in a Foxo1-dependent manner. Cell Rep. 14, 1206–1217 (2016).

    Article  CAS  PubMed  Google Scholar 

  80. Rolf, J. et al. AMPKα1: A glucose sensor that controls CD8 T-cell memory. Eur. J. Immunol. 43, 889–896 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Borges Da Silva, H. et al. The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8+ T cells. Nature 559, 264–268 (2018). This study shows that the purinergic receptor P2RX7 is required for the establishment of long-lived memory CD8+ T cells, finding commonalities between the sensing of extracellular ATP as a relay and danger signal of painful tissue damage to the nervous and immune systems.

    Article  CAS  PubMed  Google Scholar 

  82. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    Article  PubMed  Google Scholar 

  83. Pedicord, V. A., Cross, J. R., Montalvo-Ortiz, W., Miller, M. L. & Allison, J. P. Friends not foes: CTLA-4 blockade and mTOR inhibition cooperate during CD8+ T cell priming to promote memory formation and metabolic readiness. J. Immunol. 194, 2089–2098 (2015).

    Article  CAS  PubMed  Google Scholar 

  84. Lucas, C. L., Chandra, A., Nejentsev, S., Condliffe, A. M. & Okkenhaug, K. PI3Kδ and primary immunodeficiencies. Nat. Rev. Immunol. 16, 702–714 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Wu, J. et al. Loss of neurological disease HSAN-I-associated gene SPTLC2 impairs CD8+ T cell responses to infection by inhibiting T cell metabolic fitness. Immunity 50, 1218–1231.e5 (2019). This study uncovers how hereditary sensory neuropathy type 1-associated mutations in the palmitoyltransferase gene SPTLC2 dampen human immune responses to infection by blunting the sphingolipid biosynthetic capacity of CD8+ T cells, which chronically activates mTORC1, endoplasmic reticulum stress and CD8+ T cell death.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Patsoukis, N. et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat. Commun. 6, 6692 (2015).

    Article  CAS  PubMed  Google Scholar 

  87. Etchegaray, J. P. & Mostoslavsky, R. Interplay between metabolism and epigenetics: a nuclear adaptation to environmental changes. Mol. Cell 62, 695–711 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhang, D. et al. Metabolic regulation of gene expression by histone lactylation. Nature 574, 595–580 (2019).

    Article  Google Scholar 

  89. Tyrakis, P. A. et al. S-2-hydroxyglutarate regulates CD8+ T-lymphocyte fate. Nature 540, 236–241 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. 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 

  91. Masopust, D. & Soerens, A. G. Tissue-resident T cells and other resident leukocytes. Annu. Rev. Immunol. 37, 521–546 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. 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 

  93. Mackay, L. K. & Kallies, A. Transcriptional regulation of tissue-resident lymphocytes. Trends Immunol. 38, 94–103 (2017).

    Article  CAS  PubMed  Google Scholar 

  94. Li, C. et al. The transcription factor Bhlhe40 programs mitochondrial regulation of resident CD8+ T cell fitness and functionality. Immunity 17, 491–507.e7 (2019).

    Article  CAS  Google Scholar 

  95. Proietti, M. et al. ATP released by intestinal bacteria limits the generation of protective IgA against enteropathogens. Nat. Commun. 10, 250 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Stark, R. et al. TRM maintenance is regulated by tissue damage via P2RX7. Sci. Immunol. 3, eaau1022 (2018).

    Article  PubMed  Google Scholar 

  97. Borges da Silva, H. et al. Sensing of ATP via the purinergic receptor P2RX7 promotes CD8+ Trm cell generation by enhancing their sensitivity to the cytokine TGF-β. Immunity 53, 158–171.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  98. Grassi, F. The P2X7 receptor as regulator of T cell development and function. Front. Immunol. 11, 1179 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Rissiek, B., Haag, F., Boyer, O., Koch-Nolte, F. & Adriouch, S. P2X7 on mouse T cells: one channel, many functions. Front. Immunol. 6, 204 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Rissiek, B. et al. In vivo blockade of murine ARTC2.2 during cell preparation preserves the vitality and function of liver tissue-resident memory T cells. Front. Immunol. 9, 1580 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Fonseca, R. et al. Developmental plasticity allows outside-in immune responses by resident memory T cells. Nat. Immunol. 21, 412–421 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Masopust, D. et al. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 207, 553–564 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Konjar, Š. et al. Mitochondria maintain controlled activation state of epithelial-resident T lymphocytes. Sci. Immunol. 3, eaan2543 (2018). This study shows how the lipid composition of the mitochondria of tissue-resident memory CD8+ T cells of the intestine, specifically the cardiolipins, sustains their controlled activation state and empowers their proliferation and effector responses.

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. 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  CAS  PubMed  PubMed Central  Google Scholar 

  107. 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  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Smathers, R. L. & Petersen, D. R. The human fatty acid-binding protein family: evolutionary divergences and functions. Hum. Genomics 5, 170–191 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Frizzell, H. et al. Organ-specific isoform selection of fatty acid-binding proteins in tissue-resident lymphocytes. Sci. Immunol. 5, eaay9283 (2020). This study shows that tissue-resident memory CD8+ T cells metabolically adapt to tissues by varying FABP expression profiles, a mechanism required for lipid utilization and development of tissue-specific residency programmes.

    Article  CAS  PubMed  Google Scholar 

  111. Hotamisligil, G. S. & Bernlohr, D. A. Metabolic functions of FABPs - mechanisms and therapeutic implications. Nat. Rev. Endocrinol. 11, 592–605 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. 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 

  113. Holz, L. E. et al. CD8+ T cell activation leads to constitutive formation of liver tissue-resident memory T cells that seed a large and flexible niche in the liver. Cell Rep. 25, 68–79.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  114. Swadling, L. et al. Human liver memory CD8+ T cells use autophagy for tissue residence. Cell Rep. 30, 687–698.e6 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Pua, H. H., Dzhagalov, I., Chuck, M., Mizushima, N. & He, Y. W. A critical role for the autophagy gene Atg5 in T cell survival and proliferation. J. Exp. Med. 204, 25–31 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Han, S. J. et al. White adipose tissue is a reservoir for memory T cells and promotes protective memory responses to infection. Immunity 47, 1154–1168.e6 (2017). This study describes how the white adipose tissue becomes a reservoir of memory CD8+ T cells after acute infection and profiles their function, metabolic activities and interactions within the niche.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  118. Zajac, A. J. et al. Viral immune evasion due to persistence of activated T cells without effector function. J. Exp. Med. 188, 2205–2213 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  120. 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 

  121. Peng, X. et al. Molecular characterization and clinical relevance of metabolic expression subtypes in human cancers. Cell Rep. 23, 255–269 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Reznik, E. et al. A landscape of metabolic variation across tumor types. Cell Syst. 6, 301–313.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Goveia, J. et al. Meta-analysis of clinical metabolic profiling studies in cancer: challenges and opportunities. EMBO Mol. Med. 8, 1134–1142 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hu, J. et al. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat. Biotechnol. 31, 522–529 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ottensmeier, C. H. et al. Upregulated glucose metabolism correlates inversely with CD8+ T-cell infiltration and survival in squamous cell carcinoma. Cancer Res. 76, 4136–4148 (2016).

    Article  CAS  PubMed  Google Scholar 

  128. Ecker, C. et al. Differential reliance on lipid metabolism as a salvage pathway underlies functional differences of T cell subsets in poor nutrient environments. Cell Rep. 23, 741–755 (2018). This study profiles the adaptation of naive and circulating memory CD8+ T cell subsets to conditions of glucose limitation, and shows that, upon glucose restriction, CD8+ T cells can rely on fatty acid synthesis and glutamine usage, but this can come at the cost of IFNγ production and effector functions.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. DeVorkin, L. et al. Autophagy regulation of metabolism is required for CD8+ T cell anti-tumor immunity. Cell Rep. 27, 502–513.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  130. Bian, Y. et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 585, 277–282 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Quinn, W. J. et al. Lactate limits T cell proliferation via the NAD(H) redox state. Cell Rep. 33, 108500 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. García-Canaveras, J. C., Chen, L. & Rabinowitz, J. D. The tumor metabolic microenvironment: lessons from lactate. Cancer Res. 79, 3155–3162 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171, 358–371.e9 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Hui, S. et al. Glucose feeds the TCA cycle via circulating lactate. Nature 551, 115–118 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Liu, Y. et al. Tumor-repopulating cells induce PD-1 expression in CD8+ T cells by transferring kynurenine and AhR activation. Cancer Cell 33, 480–494.e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  136. Frumento, G. et al. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J. Exp. Med. 196, 459–468 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Sinclair, L. V., Neyens, D., Ramsay, G., Taylor, P. M. & Cantrell, D. A. Single cell analysis of kynurenine and system L amino acid transport in T cells. Nat. Commun. 9, 1981 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Minhas, P. S. et al. Macrophage de novo NAD+ synthesis specifies immune function in aging and inflammation. Nat. Immunol. 20, 50–63 (2019).

    Article  CAS  PubMed  Google Scholar 

  139. Ohta, A. et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl Acad. Sci. USA 103, 13132–13137 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Ohta, A. & Sitkovsky, M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414, 916–920 (2001).

    Article  CAS  PubMed  Google Scholar 

  141. Allard, B., Pommey, S., Smyth, M. J. & Stagg, J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin. Cancer Res. 19, 5626–5635 (2013).

    Article  CAS  PubMed  Google Scholar 

  142. Ohta, A. et al. A2A adenosine receptor may allow expansion of T cells lacking effector functions in extracellular adenosine-rich microenvironments. J. Immunol. 183, 5487–5493 (2009).

    Article  CAS  PubMed  Google Scholar 

  143. Baumann, T. et al. Regulatory myeloid cells paralyze T cells through cell–cell transfer of the metabolite methylglyoxal. Nat. Immunol. 21, 555–566 (2020).

    Article  CAS  PubMed  Google Scholar 

  144. Vodnala, S. K. et al. T cell stemness and dysfunction in tumors are triggered by a common mechanism. Science 363, eaau0135 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Czystowska-Kuzmicz, M. et al. Small extracellular vesicles containing arginase-1 suppress T-cell responses and promote tumor growth in ovarian carcinoma. Nat. Commun. 10, 3000 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Geiger, R. et al. L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167, 829–842.e13 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Hensley, C. T. et al. Metabolic heterogeneity in human lung tumors. Cell 164, 681–694 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Blank, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Russell, S. L. et al. Compromised metabolic reprogramming is an early indicator of CD8+ T cell dysfunction during chronic mycobacterium tuberculosis infection. Cell Rep. 29, 3564–3579.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. van Bruggen, J. A. C. et al. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8+ T cells and impede CAR T-cell efficacy. Blood 134, 44–58 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Gemta, L. F. et al. Impaired enolase 1 glycolytic activity restrains effector functions of tumor-infiltrating CD8+ T cells. Sci. Immunol. 4, eaap9520 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Siska, P. J. et al. Suppression of Glut1 and glucose metabolism by decreased Akt/mTORC1 signaling drives T cell impairment in B cell leukemia. J. Immunol. 197, 2532–2540 (2016).

    Article  CAS  PubMed  Google Scholar 

  153. He, W. et al. CD155T/TIGIT signaling regulates CD8+ T-cell metabolism and promotes tumor progression in human gastric cancer. Cancer Res. 77, 6375–6388 (2017).

    Article  CAS  PubMed  Google Scholar 

  154. Wang, T. et al. Inosine is an alternative carbon source for CD8+-T-cell function under glucose restriction. Nat. Metab. 2, 635–647 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Qiu, J. et al. Acetate promotes T cell effector function during glucose restriction. Cell Rep. 27, 2063–2074.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Leone, R. D. et al. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science 366, 1013–1021 (2019). This study demonstrates the inherent metabolic plasticity of T cells by using glutamine blockade in vivo to inhibit tumour metabolism while allowing for enhanced CD8+ TIL function due to T cells’ ability to utilize other types of metabolism in the face of glutamine inhibition.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Di Biase, S. et al. Creatine uptake regulates CD8 T cell antitumor immunity. J. Exp. Med. 216, 2869–2882 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  158. Gropper, Y. et al. Culturing CTLs under hypoxic conditions enhances their cytolysis and improves their anti-tumor function. Cell Rep. 20, 2547–2555 (2017).

    Article  CAS  PubMed  Google Scholar 

  159. Doedens, A. L. et al. Hypoxia-inducible factors enhance the effector responses of CD8+ T cells to persistent antigen. Nat. Immunol. 14, 1173–1182 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Najjar, Y. G. et al. Tumor cell oxidative metabolism as a barrier to PD-1 blockade immunotherapy in melanoma. JCI Insight 4, e124989 (2019).

    Article  PubMed Central  Google Scholar 

  161. Scharping, N. E., Menk, A. V., Whetstone, R. D., Zeng, X. & Delgoffe, G. M. Efficacy of PD-1 blockade is potentiated by metformin-induced reduction of tumor hypoxia. Cancer Immunol. Res. 5, 9–16 (2017).

    Article  CAS  PubMed  Google Scholar 

  162. Doedens, A. L. et al. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res. 70, 7465–7475 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Scharping, N. E. et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity 45, 374–388 (2016). CD8+ T cells lose mitochondrial mass and function in the TME due to repressed mitochondrial biogenesis, which cannot be rescued with checkpoint blockade immunotherapy, but instead engineering T cells to enforce mitochondrial biogenesis can improve their function, decrease tumour burden and increase overall survival.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Chinopoulos, C. Which way does the citric acid cycle turn during hypoxia? The critical role of α-ketoglutarate dehydrogenase complex. J. Neurosci. Res. 91, 1030–1043 (2013).

    Article  CAS  PubMed  Google Scholar 

  165. Sukumar, M. et al. Mitochondrial membrane potential identifies cells with enhanced stemness for cellular therapy. Cell Metab. 23, 63–76 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed Central  Google Scholar 

  167. Hurst, K. E. et al. Endoplasmic reticulum stress contributes to mitochondrial exhaustion of CD8+ T cells. Cancer Immunol. Res. 7, 476–486 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Haase, V. H. The VHL tumor suppressor: master regulator of HIF. Curr. Pharm. Des. 15, 3895–3903 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Yu, Y.-R. et al. Disturbed mitochondrial dynamics in CD8+ TILs reinforce T cell exhaustion. Nat. Immunol. 21, 1540–1551 (2020).

    Article  CAS  PubMed  Google Scholar 

  170. Bengsch, B. et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8+ T cell exhaustion. Immunity 45, 1–16 (2016). T cell exhaustion in chronic viral infection shows decreased glycolytic and mitochondrial metabolism, which can be rescued with checkpoint blockade immunotherapy or engineering T cells to improve their mitochondrial metabolism.

    Article  CAS  Google Scholar 

  171. Wolski, D. et al. Early transcriptional divergence marks virus-specific primary human CD8+ T cells in chronic versus acute infection. Immunity 47, 648–663.e8 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Scharping, N. E. et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat. Immunol. 22, 205–215 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Vardhana, S. A. et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat. Immunol. 21, 1022–1033 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Chakraborty, P. et al. Thioredoxin-1 improves the immunometabolic phenotype of antitumor T cells. J. Biol. Chem. 294, 9198–9212 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Cronin, S. J. F. et al. The metabolite BH4 controls T cell proliferation in autoimmunity and cancer. Nature 563, 564–568 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Kawalekar, O. U. et al. Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity 44, 380–390 (2016). This study highlights how engineering T cell via use of specific co-stimulatory domains in chimeric antigen receptor immunotherapeutic T cells can change the chimeric antigen receptor T cell metabolism, impacting differentiation state and function.

    Article  CAS  PubMed  Google Scholar 

  177. Zhang, Y. et al. Enhancing CD8+ T cell fatty acid catabolism within a metabolically challenging tumor microenvironment increases the efficacy of melanoma immunotherapy. Cancer Cell 32, 377–391.e9 (2017). This study shows that TILs fare better against tumours if their capacity to utilize environmental fatty acids is increased with agonists of PPARα, an effect that synergizes with PD1 blockade.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Herber, D. L. et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat. Med. 16, 880–886 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Manzo, T. et al. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J. Exp. Med. 217, e20191920 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  180. Chowdhury, P. S., Chamoto, K., Kumar, A. & Honjo, T. PPAR-induced fatty acid oxidation in T cells increases the number of tumor-reactive CD8+ T cells and facilitates anti-PD-1 therapy. Cancer Immunol. Res. 6, 1375–1387 (2018).

    Article  CAS  PubMed  Google Scholar 

  181. Chamoto, K. et al. Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proc. Natl Acad. Sci. USA 114, E761–E770 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Ma, X. et al. Cholesterol induces CD8+ T cell exhaustion in the tumor microenvironment. Cell Metab. 30, 143–156.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Yang, W. et al. Potentiating the antitumour response of CD8+ T cells by modulating cholesterol metabolism. Nature 531, 651–655 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Kanarek, N., Petrova, B. & Sabatini, D. M. Dietary modifications for enhanced cancer therapy. Nature 579, 507–517 (2020).

    Article  CAS  PubMed  Google Scholar 

  185. Di Biase, S. et al. Fasting-mimicking diet reduces HO-1 to promote T cell-mediated tumor cytotoxicity. Cancer Cell 30, 136–146 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  186. Pietrocola, F. et al. Caloric restriction mimetics enhance anticancer immunosurveillance. Cancer Cell 30, 147–160 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Contreras, N. A., Fontana, L., Tosti, V. & Nikolich-Žugich, J. Calorie restriction induces reversible lymphopenia and lymphoid organ atrophy due to cell redistribution. Geroscience 40, 279–291 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Collins, N. et al. The bone marrow protects and optimizes immunological memory during dietary restriction. Cell 178, 1088–1101.e15 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Rao, S. et al. Pathogen-mediated inhibition of anorexia promotes host survival and transmission. Cell 168, 503–516.e12 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Flint, T. R. et al. Tumor-induced IL-6 reprograms host metabolism to suppress anti-tumor immunity. Cell Metab. 24, 672–684 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Labarta-Bajo, L. et al. CD8 T cells drive anorexia, dysbiosis, and blooms of a commensal with immunosuppressive potential after viral infection. Proc. Natl Acad. Sci. USA 117, 24998–25007 (2020). This study shows that antiviral CD8+ T cell responses promote anorexia and blooms of a fasting-associated gut commensal, which can suppress the CD8+ T cell response.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Campbell, C., Marchildon, F., Michaels, A. J., Takemoto, N. & van der Veeken, J. FXR mediates T cell-intrinsic responses to reduced feeding during infection. Proc. Natl Acad. Sci. USA 117, 33446–33454 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. O’Keefe, S. J. The association between dietary fibre deficiency and high-income lifestyle-associated diseases: Burkitt’s hypothesis revisited. Lancet Gastroenterol. Hepatol. 4, 984–996 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  194. Desai, M. S. et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 167, 1339–1353.e21 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Kim, C. H., Park, J. & Kim, M. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw. 14, 277–288 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  196. Maslowski, K. M. et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 1282–1286 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Trompette, A. et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 20, 159–166 (2014).

    Article  CAS  PubMed  Google Scholar 

  198. Balmer, M. L. et al. Memory CD8+ T cells require increased concentrations of acetate induced by stress for optimal function. Immunity 44, 1312–1324 (2016). This study shows that systemic bacterial infections rapidly raise circulating levels of acetate, which is used by memory CD8+ T cells to increase their glycolytic capacity and mount an optimal recall response.

    Article  CAS  PubMed  Google Scholar 

  199. Bachem, A. et al. Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8+ T cells. Immunity 51, 285–297.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  200. Trompette, A. et al. Dietary fiber confers protection against flu by shaping Ly6c− patrolling monocyte hematopoiesis and CD8+ T cell metabolism. Immunity 48, 992–1005.e8 (2018).

    Article  CAS  PubMed  Google Scholar 

  201. Luu, M. et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat. Commun. 10, 760 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Scrimshaw, N. S. & SanGiovanni, J. P. Synergism of nutrition, infection, and immunity: an overview. Am. J. Clin. Nutr. 66, 464S–477S (1997).

    Article  CAS  PubMed  Google Scholar 

  203. Iyer, S. S. et al. Protein energy malnutrition impairs homeostatic proliferation of memory CD8 T cells. J. Immunol. 188, 77–84 (2012).

    Article  CAS  PubMed  Google Scholar 

  204. Chatraw, J. H., Wherry, E. J., Ahmed, R. & Kapasi, Z. F. Diminished primary CD8 T cell response to viral infection during protein energy malnutrition in mice is due to changes in microenvironment and low numbers of viral-specific CD8 T cell precursors. J. Nutr. 138, 806–812 (2008).

    Article  CAS  PubMed  Google Scholar 

  205. Ron-Harel, N. et al. T cell activation depends on extracellular alanine. Cell Rep. 28, 3011–3021.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Rebeles, J. et al. Obesity-induced changes in T-cell metabolism are associated with impaired memory T-cell response to influenza and are not reversed with weight loss. J. Infect. Dis. 219, 1652–1661 (2019).

    Article  CAS  PubMed  Google Scholar 

  207. Rausch, M. E., Weisberg, S., Vardhana, P. & Tortoriello, D. V. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int. J. Obes. 32, 451–463 (2008).

    Article  CAS  Google Scholar 

  208. Bhattacharjee, J. et al. Role of immunodeficient animal models in the development of fructose induced NAFLD. J. Nutr. Biochem. 25, 219–226 (2014).

    Article  CAS  PubMed  Google Scholar 

  209. Wolf, M. J. et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 26, 549–564 (2014).

    Article  CAS  PubMed  Google Scholar 

  210. Pollock, A. H. et al. Prolonged intake of dietary lipids alters membrane structure and T cell responses in LDLr−/− mice. J. Immunol. 196, 3993–4002 (2016).

    Article  CAS  PubMed  Google Scholar 

  211. Ringel, A. E. et al. Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell 183, 1848–1866.e26 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Lussier, D. M. et al. Enhanced immunity in a mouse model of malignant glioma is mediated by a therapeutic ketogenic diet. BMC Cancer 16, 310 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  213. Henning, A. N., Roychoudhuri, R. & Restifo, N. P. Epigenetic control of CD8+ T’cell differentiation. Nat. Rev. Immunol. 18, 340–356 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Yerinde, C., Siegmund, B., Glauben, R. & Weidinger, C. Metabolic control of epigenetics and its role in CD8+ T cell differentiation and function. Front. Immunol. 10, 2718 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Wellen, K. E. & Snyder, N. W. Should we consider subcellular compartmentalization of metabolites, and if so, how do we measure them? Curr. Opin. Clin. Nutr. Metab. Care 22, 347–354 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Desdín-Micó, G. et al. T cells with dysfunctional mitochondria induce multimorbidity and premature senescence. Science 368, 1371–1376 (2020).

    Article  PubMed  CAS  Google Scholar 

  217. Cham, C. M. & Gajewski, T. F. Glucose availability regulates IFN-γ production and p70S6 kinase activation in CD8+ effector T cells. J. Immunol. 174, 4670 LP–4674677 (2005).

    Article  Google Scholar 

  218. Wofford, J. A., Wieman, H. L., Jacobs, S. R., Zhao, Y. & Rathmell, J. C. IL-7 promotes Glut1 trafficking and glucose uptake via STAT5-mediated activation of Akt to support T-cell survival. Blood 111, 2101–2111 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Divakaruni, A. S., Paradyse, A., Ferrick, D. A., Murphy, A. N. & Jastroch, M. Analysis and interpretation of microplate-based oxygen consumption and pH data. Methods Enzymol. 547, 309–3540 (2014).

    Article  CAS  PubMed  Google Scholar 

  220. O’Neill, L. A. J., Kishton, R. J. & Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 16, 553–565 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  221. Wei, J. et al. Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature 576, 471–476 (2019). TILs face an immunosuppressive TME that restrains their mitochondrial activity, leading to an exhausted state. This study finds Regnase 1 acts as a negative regulator of potent antitumour responses by repressing mitochondrial function in effector T cells and restraining their long-lived potential.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Blagih, J. et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity 42, 41–54 (2015).

    Article  CAS  PubMed  Google Scholar 

  223. Miyajima, M. et al. Metabolic shift induced by systemic activation of T cells in PD-1-deficient mice perturbs brain monoamines and emotional behavior. Nat. Immunol. 18, 1342–1352 (2017).

    Article  CAS  PubMed  Google Scholar 

  224. Bu, D. X. et al. Statin-induced Kruppel-like factor 2 expression in human and mouse T cells reduces inflammatory and pathogenic responses. J. Clin. Invest. 120, 1961–1970 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Wang, H. et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 21, 298–308 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  226. Mehlhop-Williams, E. R. & Bevan, M. J. Memory CD8+ T cells exhibit increased antigen threshold requirements for recall proliferation. J. Exp. Med. 211, 345–356 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Davenport, B. et al. Aging of antiviral CD8+ memory T cells fosters increased survival, metabolic adaptations, and lymphoid tissue homing. J. Immunol. 202, 460–475 (2019).

    Article  CAS  PubMed  Google Scholar 

  228. Baazim, H. et al. CD8+ T cells induce cachexia during chronic viral infection. Nat. Immunol. 20, 701–710 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Lercher, A. et al. Type I interferon signaling disrupts the hepatic urea cycle and alters systemic metabolism to suppress T cell function. Immunity 51, 1074–1087.e9 (2019). The authors uncover specific metabolic alterations in the liver that induce systemic alterations in the levels of circulating metabolites, such as arginine and ornithine, which dampen antiviral T cell responses and liver disease.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Trefely, S. et al. Subcellular metabolic pathway kinetics are revealed by correcting for artifactual post harvest metabolism. Mol. Metab. 30, 61–71 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Binek, A. et al. Flow cytometry has a significant impact on the cellular metabolome. J. Proteome Res. 18, 169–181 (2019).

    CAS  PubMed  Google Scholar 

  232. Rappez, L. et al. Spatial single-cell profiling of intracellular metabolomes in situ. Preprint at bioRxiv https://doi.org/10.1101/510222 (2019).

    Article  Google Scholar 

  233. Hartmann, F. J. et al. Single-cell metabolic profiling of human cytotoxic T cells. Nat. Biotechnol. 39, 186–197 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  234. Pareek, V., Tian, H., Winograd, N. & Benkovic, S. J. Metabolomics and mass spectrometry imaging reveal channeled de novo purine synthesis in cells. Science 368, 283–290 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Stolp, B. et al. Salivary gland macrophages and tissue-resident CD8+ T cells cooperate for homeostatic organ surveillance. Sci. Immunol. 5, eaaz4371–00 (2020).

    Article  CAS  PubMed  Google Scholar 

  236. Winfree, S., Hato, T. & Day, R. N. Intravital microscopy of biosensor activities and intrinsic metabolic states. Methods 128, 95–104 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Argüello, R. J. et al. SCENITH: a flow cytometry-based method to functionally profile energy metabolism with single-cell resolution. Cell Metab. 32, 1063–1075.e7 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

M.R-C. is a Cancer Research Institute Irvington Fellow supported by the Cancer Research Institute (CRI2943), N.E.S is supported by the NCI Predoctoral to Postdoctoral Fellow Transition (F99/K00) Award (K00CA222711) and A.W.G. is supported by the NIH (AI067545, AI132122 and AI072117) and the University of California, San Diego Tata Chancellor Endowed Professorship. The authors thank L. Labarta-Bajo, A. Phan, M. Heeg, A. Ferry, K. Kennewick and A. Houk for insightful discussions and expert feedback on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Ananda W. Goldrath.

Ethics declarations

Competing Interests

M.R.-C. and N.E.S. declare no competing interests. A.W.G. serves on the scientific advisory boards of Pandion Therapeutics and Arsenal Bio.

Additional information

Peer review information

Nature Reviews Immunology thanks V. A. Boussiotis and the other, anonymous, reviewers for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

ATP

The molecule of energy currency inside of cells; it is dephosphorylated to AMP and adenosine by CD73 and CD39, respectively, when in the extracellular space to suppress immune function.

Oxidative phosphorylation

Metabolic process in which mitochondrial processes of the electron transport chain make ATP by consuming oxygen, making carbon dioxide and water as metabolic by-products.

Glycolysis

Metabolic process of breaking down glucose for energy, with lactic acid as a metabolic by-product.

Carbon anabolism

Metabolic biosynthetic process building larger molecules from smaller molecules.

Anaplerosis

Metabolic reactions whose products replenish tricarboxylic acid cycle intermediates.

Tricarboxylic acid (TCA) cycle

Also called the Krebs cycle and the citric acid cycle, a metabolic process of breaking down acetyl-CoA to carbon intermediates for energy and for synthesizing new metabolic products.

mTOR complex 1

(mTORC1). A protein complex that integrates signals from oxygen, energy, nutrients, stress and growth factors to control the anabolic output of the cell by stimulating protein synthesis and anabolic metabolism and restraining autophagy under conditions of nutrient abundance.

Fatty acid synthesis

Metabolic process of building fatty acids from acetyl-CoA in the cell cytoplasm.

Sterol regulatory element-binding proteins

Transcription factors from the basic helix–loop–helix leucine zipper family that translocate to the nucleus when cellular sterol concentration is low to upregulate enzymes involved in sterol biosynthesis.

Electron transport chain

(ETC). Series of protein complexes in the mitochondria that uses proton gradients to drive ATP production.

Extracellular acidification rate

(ECAR). Real-time measurement of medium acidification, used to interpret cellular glycolysis.

Oxygen consumption rate

(OCR). Real-time measurement of oxygen in media, used to interpret mitochondrial and cellular respiration.

Spare respiratory capacity

The maximum capacity of a cell to consume oxygen once the mitochondrial membrane has been uncoupled.

Fatty acid oxidation

Mitochondrial process of breaking down fatty acids into acetyl-CoA derivatives for energy.

Fatty acid

A carboxylic acid with an unsaturated or a saturated aliphatic tail.

NADPH/NADP+ ratios

Nicotinamide adenine dinucleotide phosphate in its oxidized (NADP+) and reduced forms (NADPH). A cofactor used by all forms of cellular life, it has a key role in central antioxidant systems and anabolism.

mTORC2

A protein complex that senses growth factors to control survival, proliferation and cytoskeletal rearrangements.

NADH/NAD+ ratios

Nicotinamide adenine dinucleotide in its oxidized (NAD+) and reduced (NADH) forms. A cofactor found in all living cells that is involved in redox reactions for the generation of ATP.

Fatty acid-binding proteins

(FABPs). A family composed of 12 different isoforms with distinct stoichiometries, affinities and specificities for fatty acids and heterogenous expression among CD8+ T cell subsets.

Glutaminolysis

Metabolic process to break down glutamine for energy and for carbon products such as α-ketoglutarate.

Short-chain fatty acids

Carboxylic acids with a small hydrocarbon chain, usually derived from the diet or bacterial fermentation.

Immune recall

The ability of memory lymphocytes to mount an immune response to a previously encountered antigen. A diet in which the most energy is derived from fat, rather than sugar or protein.

High-fat diet

A diet in which the most energy is derived from fat, rather than sugar or protein.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Reina-Campos, M., Scharping, N.E. & Goldrath, A.W. CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol 21, 718–738 (2021). https://doi.org/10.1038/s41577-021-00537-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41577-021-00537-8

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