Metabolic Pathways Involved in Regulatory T Cell Functionality

Rosalie W M Kempkes, Irma Joosten, Hans J P M Koenen, Xuehui He, Rosalie W M Kempkes, Irma Joosten, Hans J P M Koenen, Xuehui He

Abstract

Regulatory T cells (Treg) are well-known for their immune regulatory potential and are essential for maintaining immune homeostasis. The rationale of Treg-based immunotherapy for treating autoimmunity and transplant rejection is to tip the immune balance of effector T cell-mediated immune activation and Treg-mediated immune inhibition in favor of Treg cells, either through endogenous Treg expansion strategies or adoptive transfer of ex vivo expanded Treg. Compelling evidence indicates that Treg show properties of phenotypic heterogeneity and instability, which has caused considerable debate in the field regarding their correct use. Consequently, for further optimization of Treg-based immunotherapy, it is vital to further our understanding of Treg proliferative, migratory, and suppressive behavior. It is increasingly appreciated that the functional profile of immune cells is highly dependent on their metabolic state. Although the metabolic profiles of effector T cells are progressively understood, little is known on Treg in this respect. The objective of this review is to outline the current knowledge of human Treg metabolic profiles associated with the regulation of Treg functionality. As such information on human Treg is still limited, where information was lacking, we included insightful findings from mouse studies. To assess the available evidence on metabolic pathways involved in Treg functionality, PubMed, and Embase were searched for articles in English indexed before April 28th, 2019 using "regulatory T lymphocyte," "cell metabolism," "cell proliferation," "migration," "suppressor function," and related search terms. Removal of duplicates and search of the references was performed manually. We discerned that while glycolysis fuels the biosynthetic and bioenergetic needs necessary for proliferation and migration of human Treg, suppressive capacity is mainly maintained by oxidative metabolism. Based on the knowledge of metabolic differences between Treg and non-Treg cells, we additionally discuss and propose ways of how human Treg metabolism could be exploited for the betterment of tolerance-inducing therapies.

Keywords: FOXP3; human Treg cells; metabolism; migration; proliferation; suppressive function; tolerance-inducing therapies.

Copyright © 2019 Kempkes, Joosten, Koenen and He.

Figures

Figure 1
Figure 1
Immunosuppressive mechanisms underlying Treg-mediated immune suppression. Treg are characterized by expression of the cell surface markers CD4+, CD25high and CD127low/−, and transcription factor FOXP3. Treg modulate the immune system using their suppressive molecules PD-1, CTLA-4, CD39, and various surface receptors through inhibition of dendritic cell (DC) function and maturation, through the secretion of anti-inflammatory cytokines such as IL-10, TGF-β and IL-35, and/or through direct inhibition of Teff via induction of cytolysis using granzyme and metabolic disruption. Moreover, Treg can reduce Teff activation by limiting TCR-ligand binding.
Figure 2
Figure 2
Overview of cellular metabolism in T cells. The first step of glycolysis is the conversion of glucose into glucose-6-phosphate by hexokinases (HK) or glucokinase (GCK). During glycolysis, glucose can be shuttled away toward the pentose phosphate pathway (PPP), or stay in glycolysis resulting in its conversion into pyruvate. Pyruvate can either be converted to lactate by lactate dehydrogenase (LDH), using the nicotinamide adenine dinucleotide (NADH) produced during glycolysis, or it can be converted into acetyl-CoA and enter the tricarboxylic acid (TCA) cycle. The TCA cycle can also be fueled by acetyl-CoA produced during FAO and α-ketoglutarate following glutaminolysis. Subsequently, the TCA cycle fuels OXPHOS and FAS. NADH and reduced nicotinamide adenine dinucleotide phosphate (NADPH) are produced during the conversion of various metabolites and play an important role in proton homeostasis and ROS production via the NADP+ complex and glutathione metabolism. Although all these metabolic pathways are interlinked, cells can regulate their isolated activity and compensate for alterations in the different pathways. The PI3K-Akt-mTOR pathway plays a role in metabolic regulation through various routes. Glycolysis is stimulated via Akt and HIF-1α while fatty acid synthesis and amino acid synthesis are impacted by and impact mTORC1. The PI3K-Akt pathway also modulates FOXP3 expression, which itself impact metabolism via Myc and Akt signaling. OXPHOS, oxidative phosphorylation; FAO, fatty acid oxidation; FAS, fatty acid synthesis.
Figure 3
Figure 3
Summary of metabolic pathways involved in distinct Treg functionalities. Treg have distinct metabolic phenotypes throughout their different phases, although many pathways remain to be elucidated. (A) During proliferation, Treg have increased glycolysis and FAO. Deficiency of glutamine and tryptophan steers T cells toward Treg differentiation (–31). (B) To support the increased need for energy during migration, Treg increase their glycolytic flux. Metabolic shifts in other pathways have not been described for Treg migration (, , –39). (C) Treg show decreased glycolysis and increased OXPHOS, FAO, FAS and tryptophan metabolism during their phase of suppressive function. No relevance for the TCA cycle has been reported (, , –, , , , , , –53). The blue and red arrows are indicative for increased or decreased activity of the specific pathway in the functional phenotype of Treg, respectively. FAO, fatty acid oxidation; FAS, fatty acid synthesis; TCA, tricarboxylic acid; OXPHOS, oxidative phosphorylation.

References

    1. Fishman JA. Infection in organ transplantation. Am J Transpl. (2017) 17:856–79. 10.1111/ajt.14208
    1. Net JB, Bushell A, Wood KJ, Harden PN. Regulatory T cells: first steps of clinical application in solid organ transplantation. Transpl Int. (2015) 29:3–11. 10.1111/tri.12608
    1. Schmidt A, Oberle N, Krammer PH. Molecular mechanisms of treg-mediated T cell suppression. Front Immunol. (2012) 3:51. 10.3389/fimmu.2012.00051
    1. He X, Koenen HJPM, Slaats JHR, Joosten I. Stabilizing human regulatory T cells for tolerance inducing immunotherapy. Immunotherapy. (2017) 9:735–51. 10.2217/imt-2017-0017
    1. Huynh A, DuPage M, Priyadharshini B, Sage PT, Quiros J, Borges CM, et al. . Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability. Nat Immunol. (2015) 16:188–96. 10.1038/ni.3077
    1. Patsoukis N, Bardhan K, Chatterjee P, Sari D, Liu B, Bell LN, et al. . PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun. (2015) 6:6692. 10.1038/ncomms7692
    1. Kawai K, Uchiyama M, Hester J, Wood K, Issa F. Regulatory T cells for tolerance. Hum Immunol. (2018) 79:294–303. 10.1016/j.humimm.2017.12.013
    1. O'Neill LA, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat Rev Immunol. (2016) 16:553–65. 10.1038/nri.2016.70
    1. Van der Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. (2009) 324:1029–33. 10.1126/science.1160809
    1. Chang CH, Chang CH, Curtis JD, Maggi LB, Faubert B, Villarino AV, et al. . Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell. (2013) 153:1239–51. 10.1016/j.cell.2013.05.016
    1. Gerriets VA, Kishton RJ, Johnson MO, Cohen S, Siska PJ, Nichols AG, et al. . FOXP3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat. Immunol. (2016) 17:1459–66. 10.1038/ni.3577
    1. Koenen HJPM, Smeets RL, Vink PM, Van Rijssen E, Boots AMH, Joosten I. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17 producing cells. Blood. (2008) 112:2340–52. 10.1182/blood-2008-01-133967
    1. Beier UH, Angelin A, Akimova T, Wang L, Liu Y, Xiao H, et al. . Essential role of mitochondrial energy metabolism in FOXP3+ T-regulatory cell function and allograft survival. FASEB J. (2015) 29:2315–26. 10.1096/fj.14-268409
    1. Kishore M, Cheung KCP, Fu H, Bonacina F, Wang G, Coe D, et al. . Marelli-Berg. Regulatory T cell migration is dependent on glucokinase-mediated glycolysis. Immunity. (2017) 47:875–89.e10. 10.1016/j.immuni.2017.10.017
    1. Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, et al. . HIF1α-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. (2011) 208:1367–76. 10.1084/jem.20110278
    1. Clambey ET, McNamee EN, Westrich JA, Glover LE, Campbell EL, Jedlicka P, et al. . Hypoxia-inducible factor-1 alpha-dependent induction of FOXP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci USA. (2012) 109:2784–93. 10.1073/pnas.1202366109
    1. Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. (2006) 3:177–85. 10.1016/j.cmet.2006.02.002
    1. Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. (2006) 3:187–97. 10.1016/j.cmet.2006.01.012
    1. Gerriets VA, Kishton RJ, Nichols AG, MacIntyre AN, Inoue M, Ilkayeva O, et al. . Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J Clin Invest. (2015) 125:194–207. 10.1172/JCI76012
    1. Tarasenko TN, Gomez-Rodriguez J, Sudderth J, DeBerardinis RJ, McGuire PJ. Pyruvate dehydrogenase deficiency reveals metabolic flexibility in T-cells. Mol Genet Metab. (2018) 123:81–2. 10.1016/j.ymgme.2017.12.430
    1. Procaccini C, De Rosa V, Galgani M, Abanni L, Cali G, Porcellini A, et al. . An oscillatory switch in mTOR kinase activity sets regulatory T cell responsiveness. Immunity. (2010) 33:929–41. 10.1016/j.immuni.2010.11.024
    1. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. . Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. (2011) 186:3299–303. 10.4049/jimmunol.1003613
    1. Lee SY, Lee SH, Yang EJ, Kim EK, Kim JK, Shin DY, et al. . Metformin ameliorates inflammatory bowel disease by suppression of the STAT3 signaling pathway and regulation of the between Th17/Treg balance. PLoS ONE. (2015) 10:e0135858. 10.1371/journal.pone.0135858
    1. De Rosa V, Procaccini C, Cali G, Pirozzi G, Fontana S, Zappacosta S, et al. . A key role of leptin in the control of regulatory T cell proliferation. Immunity. (2007) 26:241–55. 10.1016/j.immuni.2007.01.011
    1. Procaccini C, Carbone F, Di Silvestre D, Brambilla F, De Rosa V, Galgani M, et al. The proteomic landscape of human ex vivo regulatory and conventional T cells reveals specific metabolic requirements. Immunity. (2016) 44:406–21. 10.1016/j.immuni.2016.01.028
    1. Cipolletta D, Feuerer M, Li A, Kamei N, Lee J, Shoelson SE, et al. . PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature. (2012) 486:549–53. 10.1038/nature11132
    1. Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. (2009) 15:930–9. 10.1038/nm.2002
    1. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. . Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. (2013) 504:446–50. 10.1038/nature12721
    1. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, et al. . Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. (2014) 40:128–39. 10.1016/j.immuni.2013.12.007
    1. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, de Roos P, et al. . Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. (2013) 504:451–5. 10.1038/nature12726
    1. Klysz D, Tai X, Robert PA, Craveiro M, Cretenet G, Oburoglu L, et al. . Glutamine-dependent alpha-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Sci Signal. (2015) 8:ra97. 10.1126/scisignal.aab2610
    1. Cobbold SP, Adams E, Farquhar CA, Nolan KF, Howie D, Lui KO, et al. . Infectious tolerance via the consumption of essential amino acids and mTOR signaling. Proc Natl Acad Sci USA. (2009) 106:12055–60. 10.1073/pnas.0903919106
    1. Yan Y, Zhang G-X, Gran B, Fallarino F, Yu S, Li M, et al. . IDO upregulates regulatory T cells via tryptophan catabolite and suppresses encephalitogenic T cell responses in experimental autoimmune encephalomyelitis. J Immunol. (2010) 185:5953–61. 10.4049/jimmunol.1001628
    1. Chow Z, Banerjee A, Hickey MJ. Controlling the fire — tissue-specific mechanisms of effector regulatory T-cell homing. Immunol Cell Biol. (2015) 93:355–63. 10.1038/icb.2014.117
    1. Fischer HJ, Sie C, Schumann E, Witte AK, Dressel R, Van Den Brandt J, et al. . The insulin receptor plays a critical role in t cell function and adaptive immunity. J Immunol. (2017) 198:1910–20. 10.4049/jimmunol.1601011
    1. Pompura SL, Dominguez-Villar M. The PI3K/AKT signaling pathway in regulatory T-cell development, stability, and function. J Leukoc. Biol. (2018) 103:1065–76. 10.1002/jlb.2mir0817-349r
    1. Finlay D, Cantrell D. Phosphoinositide 3-kinase and the mammalian target of rapamycin pathways control T cell migration. Ann N Y Acad Sci. (2010) 1183:149–57. 10.1111/j.1749-6632.2009.05134.x
    1. Carlson CM, Endrizzi BT, Wu J, Ding X, Weinreich MA, Walsh ER, et al. . Kruppel-like factor 2 regulates thymocyte and T-cell migration. Nature. (2006) 442:299–302. 10.1038/nature04882
    1. Chen LC, Nicholson YT, Rosborough BR, Thomson AW, Raimondi G. A novel mTORC1-dependent, Akt-independent pathway differentiates the gut tropism of regulatory and conventional CD4 T cells. J Immunol. (2016) 197:1137–47. 10.4049/jimmunol.1600696
    1. Liu G, Burns S, Huang G, Boyd K, Proia RL, Flavell RA, et al. . The receptor S1P1 overrides regulatory T cell-mediated immune suppression through Akt-mTOR. Nat Immunol. (2009) 10:769–77. 10.1038/ni.1743
    1. Priceman SJ, Shen S, Wang L, Deng J, Yue C, Kujawski M, et al. . S1PR1 is crucial for accumulation of regulatory T cells in tumors via STAT3. Cell Rep. (2014) 6:992–9. 10.1016/j.celrep.2014.02.016
    1. Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, et al. . An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. (2011) 478, 197–203. 10.1038/nature10491
    1. Miska J, Lee-Chang C, Rashidi A, Muroski ME, Chang AL, Lopez-Rosas A, et al. . HIF-1α is a metabolic switch between glycolytic-driven migration and oxidative phosphorylation-driven immunosuppression of tregs in glioblastoma. Cell Rep. (2019) 27:226–37.e4. 10.1016/j.celrep.2019.03.029
    1. Walsh PT, Buckler JL, Zhang J, Gelman AE, Dalton NM, Taylor DK, et al. . PTEN inhibits IL-2 receptor-mediated expansion of CD4+ CD25+ Tregs. J Clin Invest. (2006) 116:2521–31. 10.1172/jci28057
    1. Layman AAK, Deng G, O'Leary CE, Tadros S, Thomas RM, Dybas JM, et al. . NDFIP1 restricts mTORC1 signalling and glycolysis in regulatory T cells to prevent autoinflammatory disease. Nat Commun. (2017) 8:15677. 10.1038/ncomms15677
    1. Priyadharshini B, Loschi M, Newton RH, Zhang JW, Finn KK, Gerriets VA, et al. . Cutting edge. TGF-beta and phosphatidylinositol 3-kinase signals modulate distinct metabolism of regulatory T cell subsets. J Immunol. (2018) 201:2215–9. 10.4049/jimmunol.1800311
    1. De Rosa V, Galgani M, Porcellini A, Colamatteo A, Santopaolo M, Zuchegna C, et al. . Glycolysis controls the induction of human regulatory T cells by modulating the expression of FOXP3 exon 2 splicing variants. Nat Immunol. (2015) 16:1174–84. 10.1038/ni.3269
    1. DuPage M, Chopra G, Quiros J, Rosenthal WL, Morar MM, Holohan D, et al. . The chromatin-modifying enzyme Ezh2 is critical for the maintenance of regulatory T cell identity after activation. Immunity. (2015) 42:227–38. 10.1016/j.immuni.2015.01.007
    1. Urbano PCM, Koenen HJPM, Joosten I, He X. An autocrine TNFalpha-tumor necrosis factor receptor 2 loop promotes epigenetic effects inducing human treg stability in vitro. Front Immunol. (2018) 9:573. 10.3389/fimmu.2018.00573
    1. Li X, Liang Y, LeBlanc M, Benner C, Zheng Y. Function of a FOXP3 cis-element in protecting regulatory T cell identity. Cell. (2014) 158:734–48. 10.1016/j.cell.2014.07.030
    1. Sena LA, Li S, Jairaman A, Prakriya M, Ezponda T, Hildeman DA, et al. . Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity. (2013) 38:225–36. 10.1016/j.immuni.2012.10.020
    1. Angelin A, Gil-de-Gomez L, Dahiya S, Jiao J, Guo L, Levine MH, et al. . FOXP3 reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. (2017) 25:1282. 10.1016/j.cmet.2016.12.018
    1. Wedel J, Bruneau S, Liu K, Kong SW, Sage PT, Sabatini DM, et al. . DEPTOR modulates activation responses in CD4+ T cells and enhances immunoregulation following transplantation. Am J Transpl. (2018) 19:77–88. 10.1111/ajt.14995
    1. Finlay DK, Sinclair LV, Feijoo C, Waugh CM, Hagenbeek TJ, Spits H, et al. Phosphoinositide-dependent kinase 1 controls migration and malignant transformation but not cell growth and proliferation in PTEN-null lymphocytes. J Exp Med. (2009) 206:2441–54. 10.1084/jem.20090219
    1. Haas R, Smith J, Rocher-Ros V, Nadkarni S, Montero-Melendez T, D'Acquisto F, et al. . Lactate regulates metabolic and pro-inflammatory circuits in control of T cell migration and effector functions. PLoS Biol. (2015) 13:e1002202. 10.1371/journal.pbio.1002202
    1. Chapman NM, Zeng H, Nguyen TLM, Wang Y, Vogel P, Dhungana Y, et al. . mTOR coordinates transcriptional programs and mitochondrial metabolism of activated Treg subsets to protect tissue homeostasis. Nat Commun. (2018) 9:2095. 10.1038/s41467-018-04392-5
    1. Buck MD, O'Sullivan D, Pearce EL. T cell metabolism drives immunity. J Exp Med. (2015) 212:1345–60. 10.1084/jem.20151159
    1. Hansmann L, Schmidl C, Kett J, Steger L, Andreesen R, Hoffmann P, et al. . Dominant Th2 differentiation of human regulatory T cells upon loss of FOXP3 expression. J Immunol. (2012) 188:1275–82. 10.4049/jimmunol.1102288
    1. Van Gool F, Nguyen MLT, Mumbach MR, Satpathy AT, Rosenthal WL, Giacometti S, et al. . A mutation in the transcription factor Foxp3 drives T helper 2 effector function in regulatory T cells. Immunity. (2019) 50:362–377.e6. 10.1016/j.immuni.2018.12.016
    1. Sawant DV, Wu H, Yao W, Sehra S, Kaplan MH, Dent AL. The transcriptional repressor Bcl6 controls the stability of regulatory T cells by intrinsic and extrinsic pathways. Immunology. (2015) 145:11–23. 10.1111/imm.12393
    1. Howie D, Cobbold SP, Adams E, Ten Bokum A, Necula AS, Zhang W, et al. . FOXP3 drives oxidative phosphorylation and protection from lipotoxicity. JCI Insight. (2017) 2:e89160. 10.1172/jci.insight.89160
    1. Cluxton D, Petrasca A, Moran B, Fletcher JM. Differential regulation of human Treg and Th17 cells by fatty acid synthesis and glycolysis. Front Immunol. (2019) 10:115. 10.3389/fimmu.2019.00115
    1. Zeng H, Yang K, Cloer C, Neale G, Vogel P, Chi H. mTORC1 couples immune signals and metabolic programming to establish T(reg)-cell function. Nature. (2013) 499:485–90. 10.1038/nature12297
    1. Thurnher M, Gruenbacher G. T lymphocyte regulation by mevalonate metabolism. Sci Signal. (2015) 8:re4. 10.1126/scisignal.2005970
    1. Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, Ren J, et al. . Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells. J Biol Chem. (2010) 285:7176–86. 10.1074/jbc.M109.047423
    1. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB. ATP-citrate lyase links cellular metabolism to histone acetylation. Science. (2009) 324:1076–80. 10.1126/science.1164097
    1. Yang WY, Shao Y, Lopez-Pastrana J, Mai J, Wang H, Yang X-F. (2015). Pathological conditions re-shape physiological Tregs into pathological Tregs. Burns Trauma. 3:1. 10.1186/s41038-015-0001-0
    1. Zhang C, Xiao C, Wang P, Xu W, Zhang A, Li Q, et al. . The alteration of Th1/Th2/Th17/Treg paradigm in patients with type 2 diabetes mellitus. Relationship with diabetic nephropathy. Hum Immunol. (2014) 75:289–96. 10.1016/j.humimm.2014.02.007
    1. Wagner NM, Brandhorst G, Czepluch F, Lankeit M, Eberle C, Herzberg S, et al. . Circulating regulatory T cells are reduced in obesity and may identify subjects at increased metabolic and cardiovascular risk. Obesity. (2013) 21:461–8. 10.1002/oby.20087
    1. Wang H, Franco F, Ho PC. Metabolic regulation of Tregs in cancer. Opportunities for immunotherapy Trends Cancer. (2017) 3:583–92. 10.1016/j.trecan.2017.06.005
    1. Negrotto L, Correale J. Amino acid catabolism in multiple sclerosis affects immune homeostasis. J Immunol. (2017) 198:1900–9. 10.4049/jimmunol.1601139
    1. Perl A. Activation of mTOR (mechanistic target of rapamycin) in rheumatic diseases. Nat Rev Rheumatol. (2016) 12:169–82. 10.1038/nrrheum.2015.172
    1. Choi SC, Hutchinson TE, Titov AA, Seay HR, Li S, Brusko TM, et al. . The lupus susceptibility gene Pbx1 regulates the balance between follicular helper T cell and regulatory T cell differentiation. J Immunol. (2016) 197:458–69. 10.4049/jimmunol.1502283
    1. Issa F, Wood KJ. Translating tolerogenic therapies to the clinic - where do we stand? Front Immunol. (2012) 3:254. 10.3389/fimmu.2012.00254
    1. Romano M, Tung SL, Smyth LA, Lombardi G. Treg therapy in transplantation: a general overview. Transpl Int. (2017) 30:745–53. 10.1111/tri.12909
    1. Zhang D, Chia C, Jiao X, Jin W, Kasagi S, Wu R, et al. . D-mannose induces regulatory T cells and suppresses immunopathology. Nat Med. (2017) 23:1036–45. 10.1038/nm.4375
    1. Makita N, Ishiguro J, Suzuki K, Nara F. Dichloroacetate induces regulatory T-cell differentiation and suppresses Th17-cell differentiation by pyruvate dehydrogenase kinase-independent mechanism. J Pharm Pharmaco. (2017) 69:43–51. 10.1111/jphp.12655
    1. Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity. (2014) 41:518–28. 10.1016/j.immuni.2014.09.008
    1. He X, Smeets RL, Koenen HJ, Vink PM, Wagenaars J, Boots AM, et al. . Mycophenolic acid-mediated suppression of human CD4+ T cells: more than mere guanine nucleotide deprivation. Am J Transpl. (2011) 11:439–49. 10.1111/j.1600-6143.2010.03413.x
    1. Kornberg MD, Bhargava P, Calabresi P, Snyder SH. Dimethyl fumarate mediates immune modulation by inhibition of GAPDH and aerobic glycolysis. Mult Scler. (2017) 23:28–9. 10.1177/1352458517693959
    1. Gualdoni GA, Mayer KA, Goschl L, Boucheron N, Ellmeier W, Zlabinger GJ. The AMP analog AICAR modulates the Treg/Th17 axis through enhancement of fatty acid oxidation. FASEB J. (2016) 30:3800–9. 10.1096/fj.201600522R
    1. Zhang J, Shan J, Chen X, Li S, Long D, Li Y. Celastrol mediates Th17 and Treg cell generation via metabolic signaling. Biochem Biophys Res Commun. (2018) 497:883–9. 10.1016/j.bbrc.2018.02.163
    1. Schneider-Schaulies J, Beyersdorf N. CD4+ FOXP3+ regulatory T cell-mediated immunomodulation by anti-depressants inhibiting acid sphingomyelinase. Biol Chem. (2018) 399:1175–82. 10.1515/hsz-2018-0159
    1. Hester J, Schiopu A, Nadig SN, Wood KJ. Low-dose rapamycin treatment increases the ability of human regulatory T cells to inhibit transplant arteriosclerosis in vivo. Am J Transpl. (2012) 12:2008–16. 10.1111/j.1600-6143.2012.04065.x
    1. Sinclair LV, Finlay D, Feijoo C, Cornish GH, Gray A, Ager A, et al. . Phosphatidylinositol-3-OH kinase and nutrient-sensing mTOR pathways control T lymphocyte trafficking. Nat Immunol. (2008) 9:513–21. 10.1038/ni.1603
    1. Freitag J, Berod L, Kamradt T, Sparwasser T. Immunometabolism and autoimmunity. Immunol Cell Biol. (2016) 94:925–34. 10.1038/icb.2016.77
    1. Lee CF, Lo YC, Cheng CH, Furtmuller GJ, Oh B, Andrade-Oliveira V, et al. . Preventing allograft rejection by targeting immune metabolism. Cell Rep. (2015) 13:760–70. 10.1016/j.celrep.2015.09.036
    1. Wawman RE, Bartlett H, Oo YH. Regulatory T cell metabolism in the hepatic microenvironment. Front Immunol. (2018) 8:1889. 10.3389/fimmu.2017.01889
    1. Braza MS, van Leent MM, Lameijer M, Sanchez-Gaytan BL, Arts RJ, Pérez-Medina C, et al. . Inhibiting inflammation with myeloid cell-specific nanobiologics promotes organ transplant acceptance. Immunity. (2018) 49:819–28. e6. 10.1016/j.immuni.2018.09.008

Source: PubMed

3
Abonner