DNA; NAD; T cells; acetylation; epigentics; histone; metabolism
Résumé :
[en] T cell subsets adapt and rewire their metabolism according to their functions and surrounding microenvironment. Whereas naive T cells rely on mitochondrial metabolic pathways characterized by low nutrient requirements, effector T cells induce kinetically faster pathways to generate the biomass and energy needed for proliferation and cytokine production. Recent findings support the concept that alterations in metabolism also affect the epigenetics of T cells. In this review we discuss the connections between T cell metabolism and epigenetic changes such as histone post-translational modifications (PTMs) and DNA methylation, as well as the 'extra-metabolic' roles of metabolic enzymes and molecules. These findings collectively point to a new group of potential therapeutic targets for the treatment of T cell-dependent autoimmune diseases and cancers.
Disciplines :
Immunologie & maladie infectieuse
Auteur, co-auteur :
Soriano-Baguet, Leticia ; Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg, Immunology and Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
BRENNER, Dirk ; University of Luxembourg > Luxembourg Centre for Systems Biomedicine (LCSB) > Immunology and Genetics
Co-auteurs externes :
no
Langue du document :
Anglais
Titre :
Metabolism and epigenetics at the heart of T cell function.
FNR; PRIDE/11012546/NEXTIMMUNE,; FNR CORE Grants C21/BM/15796788 and C18/BM/12691266
Subventionnement (détails) :
We apologize to researchers whose work could not be referenced here owing to space limitations. We thank Anouk Ewen for the design of the figures using BioRender.com . L.S-B. and D.B. are supported by the Fonds National de la Recherche Luxembourg ( FNR ; PRIDE/11012546/NEXTIMMUNE). D.B. is supported by FNR CORE grants ( C21/BM/15796788 and C18/BM/12691266 ).
Buck, M.D., et al. T cell metabolism drives immunity. J. Exp. Med. 212 (2015), 1345–1360.
Franchina, D.G., et al. Reactive oxygen species: involvement in T cell signaling and metabolism. Trends Immunol. 39 (2018), 489–502.
Guerra, L., et al. Metabolic modulation of immunity: a new concept in cancer immunotherapy. Cell Rep., 32, 2020, 107848.
Kurniawan, H., et al. Regulatory T cell metabolism at the intersection between autoimmune diseases and cancer. Eur. J. Immunol. 50 (2020), 1626–1642.
Rangel Rivera, G.O., et al. Fundamentals of T cell metabolism and strategies to enhance cancer immunotherapy. Front. Immunol., 12, 2021, 645242.
Kurniawan, H., et al. Glutathione restricts serine metabolism to preserve regulatory T cell function. Cell Metab. 31 (2020), 920–936.
Hochrein, S.M., et al. The glucose transporter GLUT3 controls T helper 17 cell responses through glycolytic-epigenetic reprogramming. Cell Metab. 34 (2022), 516–532.e11.
Soriano-Baguet, L., et al. Pyruvate dehydrogenase fuels a critical citrate pool that is essential for Th17 cell effector function. Cell Rep., 42(2), 2023.
Qiu, J., et al. Acetate promotes T cell effector function during glucose restriction. Cell Rep 27 (2019), 2063–2074.
Sugiura, A., et al. MTHFD2 is a metabolic checkpoint controlling effector and regulatory T cell fate and function. Immunity 55 (2022), 65–81.
Dutta, A., et al. New insights into epigenetic regulation of T cell differentiation. Cells, 10, 2021, 3459.
Rhodes, C.T., Lin, C.-H.A., Role of the histone methyltransferases Ezh2 and Suv4-20h1/Suv4-20h2 in neurogenesis. Neural Regen. Res. 18 (2023), 469–473.
Ducker, G.S., Rabinowitz, J.D., One-carbon metabolism in health and disease. Cell Metab. 25 (2017), 27–42.
Kurniawan, H., et al. The emerging role of one-carbon metabolism in T cells. Curr. Opin. Biotechnol. 68 (2021), 193–201.
Ma, E.H., et al. Serine is an essential metabolite for effector T cell expansion. Cell Metab. 25 (2017), 345–357.
Meiser, J., et al. Serine one-carbon catabolism with formate overflow. Sci. Adv., 2, 2016, e1601273.
Tran, K.A., et al. The role of α-ketoglutarate-dependent proteins in pluripotency acquisition and maintenance. J. Biol. Chem. 294 (2019), 5408–5419.
Li, J., et al. KDM6B-dependent chromatin remodeling underpins effective virus-specific CD8+ T cell differentiation. Cell Rep., 34, 2021, 108839.
Xu, T., et al. Kdm6b regulates the generation of effector CD8+ T cells by inducing chromatin accessibility in effector-associated genes. J. Immunol. 206 (2021), 2170–2183.
Russ, B.E., et al. Regulation of H3K4me3 at transcriptional enhancers characterizes acquisition of virus-specific CD8+ T cell-lineage-specific function. Cell Rep. 21 (2017), 3624–3636.
LaMere, S.A., et al. H3K27 methylation dynamics during CD4 T cell activation: regulation of JAK/STAT and IL12RB2 expression by JMJD3. J. Immunol. 199 (2017), 3158–3175.
Li, Q., et al. Critical role of histone demethylase Jmjd3 in the regulation of CD4+ T-cell differentiation. Nat. Commun., 5, 2014, 5780.
Carbonneau, M., et al. The oncometabolite 2-hydroxyglutarate activates the mTOR signalling pathway. Nat. Commun., 7, 2016, 12700.
Tyrakis, P.A., et al. The immunometabolite S-2-hydroxyglutarate regulates CD8+ T-lymphocyte fate. Nature 540 (2016), 236–241.
Notarangelo, G., et al. Oncometabolite D-2HG alters T cell metabolism to impair CD8+ T cell function. Science 377 (2022), 1519–1529.
Sun, X.-J., et al. The role of histone acetyltransferases in normal and malignant hematopoiesis. Front. Oncol., 5, 2015, 108.
Ellmeier, W., Seiser, C., Histone deacetylase function in CD4+ T cells. Nat. Rev. Immunol. 18 (2018), 617–634.
Liotti, A., et al. Epigenetics: an opportunity to shape innate and adaptive immune responses. Immunology 167 (2022), 451–470.
Glozak, M.A., et al. Acetylation and deacetylation of non-histone proteins. Gene 363 (2005), 15–23.
Kwon, H.-S., et al. Three novel acetylation sites in the Foxp3 transcription factor regulate the suppressive activity of regulatory T cells. J. Immunol. 188 (2012), 2712–2721.
van Loosdregt, J., et al. Regulation of Treg functionality by acetylation-mediated Foxp3 protein stabilization. Blood 115 (2010), 965–974.
Kuroda, S., et al. Basic leucine zipper transcription factor, ATF-like (BATF) regulates epigenetically and energetically effector CD8 T-cell differentiation via Sirt1 expression. Proc. Natl. Acad. Sci. U. S. A. 108 (2011), 14885–14889.
Møller, S.H., et al. Metabolic programs tailor T cell immunity in viral infection, cancer, and aging. Cell Metab. 34 (2022), 378–395.
Bricker, D.K., et al. A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila, and humans. Science 337 (2012), 96–100.
Herzig, S., et al. Identification and functional expression of the mitochondrial pyruvate carrier. Science 337 (2012), 93–96.
Wenes, M., et al. The mitochondrial pyruvate carrier regulates memory T cell differentiation and antitumor function. Cell Metab. 34 (2022), 731–746.
Peng, M., et al. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science 354 (2016), 481–484.
Xu, K., et al. Glycolytic ATP fuels phosphoinositide 3-kinase signaling to support effector T helper 17 cell responses. Immunity 54 (2021), 976–987.
Mak, T.W., et al. Glutathione primes T cell metabolism for inflammation. Immunity 46 (2017), 675–689.
Xie, N., et al. NAD+ metabolism: pathophysiologic mechanisms and therapeutic potential. Signal. Transduct. Target. Ther., 5, 2020, 227.
Hamaidi, I., Kim, S., Sirtuins are crucial regulators of T cell metabolism and functions. Exp. Mol. Med. 54 (2022), 207–215.
Beier, U.H., et al. Essential role of mitochondrial energy metabolism in Foxp3+ T-regulatory cell function and allograft survival. FASEB J. 29 (2015), 2315–2326.
Lin, W., et al. Sirtuin4 suppresses the anti-neuroinflammatory activity of infiltrating regulatory T cells in the traumatically injured spinal cord. Immunology 158 (2019), 362–374.
Wang, K., et al. SIRT5 contributes to colorectal cancer growth by regulating T cell activity. J Immunol Res, 2020, 2020, 3792409.
Hamaidi, I., et al. Sirt2 inhibition enhances metabolic fitness and effector functions of tumor-reactive T cells. Cell Metab. 32 (2020), 420–436.
Wei, X., et al. Depot-specific regulation of NAD+/SIRTs metabolism identified in adipose tissue of mice in response to high-fat diet feeding or calorie restriction. J. Nutr. Biochem., 80, 2020, 108377.
Quinn, W.J., et al. Lactate limits T cell proliferation via the NAD(H) redox state. Cell Rep., 33, 2020, 108500.
Angelin, A., et al. Foxp3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. 25 (2017), 1282–1293.
Zhang, D., et al. Metabolic regulation of gene expression by histone lactylation. Nature 574 (2019), 575–580.
Chen, A.-N., et al. Lactylation, a novel metabolic reprogramming code: current status and prospects. Front. Immunol., 12, 2021, 688910.
Wang, Z., et al. Histone posttranslational modifications of CD4+ T cell in autoimmune diseases. Int. J. Mol. Sci., 17, 2016, 1547.
Alaskhar Alhamwe, B., et al. Histone modifications and their role in epigenetics of atopy and allergic diseases. Allergy Asthma Clin. Immunol., 14, 2018, 39.
Tan, M., et al. Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell 146 (2011), 1016–1028.
Ntorla, A., Burgoyne, J.R., The regulation and function of histone crotonylation. Front. Cell Dev. Biol., 9, 2021, 624914.
Jiang, G., et al. Protein lysine crotonylation: past, present, perspective. Cell Death Dis., 12, 2021, 703.
Delaney, K., et al. Histone lysine methacrylation is a dynamic post-translational modification regulated by HAT1 and SIRT2. Cell Discov., 7, 2021, 122.
Zhu, D., et al. Histone citrullination: a new target for tumors. Mol. Cancer, 20, 2021, 90.
Liu, J., et al. Histone succinylation and its function on the nucleosome. J. Cell. Mol. Med. 25 (2021), 7101–7109.
Dehennaut, V., et al. O-GlcNAcylation, an epigenetic mark. Focus on the histone code, TET family proteins, and Polycomb group proteins. Front. Endocrinol., 5, 2014, 155.
Ghoneim, H.E., et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170 (2017), 142–157.
Korman, A.J., et al. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat. Rev. Drug Discov. 21 (2022), 509–528.
Carty, S.A., et al. The loss of TET2 promotes CD8+ T cell memory differentiation. J. Immunol. 200 (2018), 82–91.
Matias, M.I., et al. Regulatory T cell differentiation is controlled by αKG-induced alterations in mitochondrial metabolism and lipid homeostasis. Cell Rep., 37, 2021, 109911.
Xu, T., et al. Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism. Nature 548 (2017), 228–233.
Laukka, T., et al. Fumarate and succinate regulate expression of hypoxia-inducible genes via TET enzymes. J. Biol. Chem. 291 (2016), 4256–4265.
Weinberg, S.E., et al. Mitochondrial complex III is essential for suppressive function of regulatory T cells. Nature 565 (2019), 495–499.
Angiari, S., et al. Pharmacological activation of pyruvate kinase M2 inhibits CD4+ T cell pathogenicity and suppresses autoimmunity. Cell Metab. 31 (2020), 391–405.
Damasceno, L.E.A., et al. PKM2 promotes Th17 cell differentiation and autoimmune inflammation by fine-tuning STAT3 activation. J. Exp. Med., 217, 2020, e20190613.
Almeida, L., et al. Metabolic pathways in T cell activation and lineage differentiation. Semin. Immunol. 28 (2016), 514–524.
Chang, C.-H., et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153 (2013), 1239–1251.
Shyer, J.A., et al. Metabolic signaling in T cells. Cell Res. 30 (2020), 649–659.
Mondragón, L., et al. GAPDH overexpression in the T cell lineage promotes angioimmunoblastic T cell lymphoma through an NF-κB-dependent mechanism. Cancer Cell 36 (2019), 268–287.
Shimazu, T., et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339 (2013), 211–214.
Zhang, H., et al. Ketogenesis-generated β-hydroxybutyrate is an epigenetic regulator of CD8+ T-cell memory development. Nat. Cell Biol. 22 (2020), 18–25.
Apostolova, P., Pearce, E.L., Lactic acid and lactate: revisiting the physiological roles in the tumor microenvironment. Trends Immunol. 43 (2022), 969–977.
Brand, A., et al. LDHA-associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab. 24 (2016), 657–671.
Elia, I., et al. Tumor cells dictate anti-tumor immune responses by altering pyruvate utilization and succinate signaling in CD8+ T cells. Cell Metab. 34 (2022), 1137–1150.
Feng, Q., et al. Lactate increases stemness of CD8+ T cells to augment anti-tumor immunity. Nat. Commun., 13, 2022, 4981.
Geiger, R., et al. L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167 (2016), 829–842.
Carriche, G.M., et al. Regulating T-cell differentiation through the polyamine spermidine. J. Allergy Clin. Immunol. 147 (2021), 335–348.
Tang, K., et al. Ammonia detoxification promotes CD8+ T cell memory development by urea and citrulline cycles. Nat. Immunol. 24 (2023), 162–173.
Leeming, E.R., et al. Effect of diet on the gut microbiota: rethinking intervention duration. Nutrients, 11, 2019, 2862.
Ivanov, I.I., et al. T cell responses to the microbiota. Annu. Rev. Immunol. 40 (2022), 559–587.
Collins, N., Dietary regulation of memory T cells. Int. J. Mol. Sci., 21, 2020, 4363.
Tan, J., et al. Your regulatory T cells are what you eat: how diet and gut microbiota affect regulatory T cell development. Front. Nutr., 9, 2022, 878382.
Arroyo Hornero, R., et al. The impact of dietary components on regulatory T cells and disease. Front. Immunol., 11, 2020, 253.
Kim, C.H., et al. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw. 14 (2014), 277–288.
Schiweck, C., et al. Regulation of CD4+ and CD8+ T cell biology by short-chain fatty acids and its relevance for autoimmune pathology. Int. J. Mol. Sci., 23, 2022, 8272.
Yerinde, C., et al. Metabolic control of epigenetics and its role in CD8+ T cell differentiation and function. Front. Immunol., 10, 2019, 2718.
Wu, Q., et al. Serine and metabolism regulation: a novel mechanism in antitumor immunity and senescence. Aging Dis. 11 (2020), 1640–1653.
Pietrocola, F., et al. Acetyl coenzyme A: a central metabolite and second messenger. Cell Metab. 21 (2015), 805–821.
Wellen, K.E., et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324 (2009), 1076–1080.
Lochner, M., et al. Fatty acid metabolism in the regulation of T cell function. Trends Immunol. 36 (2015), 81–91.
Moffett, J.R., et al. Acetate revisited: a key biomolecule at the nexus of metabolism, epigenetics and oncogenesis – part 1: acetyl-CoA, acetogenesis and acyl-CoA short-chain synthetases. Front. Physiol., 11, 2020, 580167.