GET THE APP

T cell Metabolism - Regulating Energy
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2012) Volume 0, Issue 0

T cell Metabolism - Regulating Energy

Clemens Cammann1*, Burkhart Schraven1,2 and Jonathan A. Lindquist1
1Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University, Magdeburg, Germany
2Department of Immune Control, Helmholtz Centre for Infection Research, Braunschweig, Germany
*Corresponding Author: Clemens Cammann, Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University, Leipziger Strasse 44 39120 Magdeburg, Germany, Tel: +49-391-671-7900, Fax: +49-391-671-5852 Email:

Abstract

Antigenic stimulation of T cells initiates a change from the resting state into an activated one mediated by triggering the T cell receptor (TCR). This change is characterized by rapid proliferation, differentiation and acquisition of effector functions. To maintain the energetic needs accompanied by this processes, T cells are able to adapt their uptake and utilization of extracellular nutrients. Proliferation and differentiation into distinct subsets of T lymphocytes like effector-, regulatory-, and memory T cells is mediated by antigens, various cytokines and growth factors through their respective signaling pathways they trigger. Since these subsets acquire different functions in the immune system, their metabolic profiles also differ. Throughout the last decade the role metabolism was intensively investigated and evolved into one major part in understanding activation and differentiation processes in T cells. Key molecules like AKT and AMPK were described to be major regulators of metabolism. Therefore, we discuss in this review which signaling molecules are known regulate metabolic pathways in T cells and we give an overview over the mechanisms how they accomplish this task.

Introduction

T cells play a central role in the immune system and are important for cell mediated immunity. A functional immune response requires rapid cell growth, proliferation, and the production of effector proteins. In the presence of specific antigens T lymphocytes must rapidly shift from a resting state to an activated one to accomplish these tasks. The activation of T cells is accompanied by a huge demand for ATP; the universal energy carrier in cell metabolism. The main processes to generate ATP are glycolysis and the citric acid cycle followed by oxidative phosphorylation. In resting T cells oxidative phosphorylation was described to be the central energy producing process [1,2]. Furthermore it was described that upon activation the energy produced by oxidative phosphorylation in resting cells is not sufficient. Therefore lymphocytes undergo a metabolic shift to an increased glycolytic rate, which leads in turn to lactate production [3-5]. This reprogramming of cellular metabolism is described in the literature as anaerobic glycolysis for example during intense muscular activity [6] where myocytes switch their metabolism under “working” conditions, in the absence of oxygen, towards elevated levels of glucose transport and high rates of glycolysis. However Otto Warburg first observed these features for cancer cells in the presence of oxygen [7] therefore it was called “aerobic glycolysis”. Aerobic glycolysis was long thought to be a feature unique to cancer cells. However, we now know that the Warburg effect is also observed during the rapid proliferation of primary T cells, and it is viewed as a general feature of anabolic metabolism [5,8]. Anabolic metabolism is a characteristic feature of proliferating cells, which have to synthesize all cellular material in order to form two daughter cells. Therefore they require energy carriers like ATP and macromolecular precursors to generate biomass in form of proteins, ribonucleotides and lipids. By upregulating their glycolytic rate, cells become capable to maintain these anabolic mechanisms.

In the last years the focus shifted from metabolism itself to the question of how T cells regulate this metabolic shift. During the immune response T cells become activated by triggering of the T cell receptor, which initiates specific signaling events. This includes the activation of the phosphoinositide-3-kinase(PI3K)/Proteinkinase B (AKT) pathway, mammalian target of rapamycin (mTOR), and adenosinemonophosphate- activated protein kinase (AMPK), which were shown to play a central role in regulating T cell metabolism [2,4,5,9,10]. In this review we will focus on AKT and AMPK, how these signaling molecules can regulate metabolic pathways in T cells and provide an overview of potential mechanisms used to accomplish this task.

Changes in T cell Metabolism upon Activation

Resting T cells require relatively low amounts of energy for housekeeping functions, i.e. homeostasis. Most of this energy is produced by oxidative phosphorylation (OXPHOS) through the degradation of glucose, fatty acids, and glutamine [1,2]. Upon activation, cellular programs direct T cells towards proliferation, differentiation, and cytokine production. The subsequent need for energy and metabolic precursors was shown to be accomplished by a strong upregulation of glycolysis [4,11], which is characterized by an increased uptake of glucose, increased expression of glycolytic enzymes and the generation of lactate from pyruvate. Although the generation of ATP by glycolysis is inefficient when compared to OXPHOS (2ATP<36ATP), upregulating glycolysis has the advantage of being a fast process and was shown to protect cells against apoptosis [11-13]. Since upregulating glycolysis without a corresponding increase in OXPHOS would lead to an accumulation of the end product pyruvate, it was shown that the excess pyruvate generated is converted to lactate by lactate dehydrogenase [4,14]. This step is essential to regenerate the reducing agent NADH, which is needed to maintain the high glycolytic turnover. Since high concentrations of lactate are toxic to the cells, the lactate produced is also secreted.

These observations lead to the conclusion that glucose is the major energy source of activated lymphocytes. This was also confirmed by showing that removing glucose in activated T cells leads to an inhibition of T cell proliferation and cytokine production [11]. In addition, also other metabolic pathways have been shown to play a role in T cell metabolism, e.g. increased glutamine consumption was shown to be essential for T cell function [8,15,16]. Glutamine is degraded via the TCA cycle, providing a nitrogen source for non-essential amino acids and nucleotides, and refilling the intermediates of the TCA cycle which are also used for biosynthetic processes that are essential for maintaining T cell proliferation [8]. At the end of the TCA-cycle malate dehydrogenase converts the generated malate to pyruvate. This pyruvate together with an upregulated glycolysis can foster the generation of lactate. Beside the generation of ATP, T cells also require NADPH to support lipid and nucleotide biosynthesis. NADPH is generated in two different processes, the pentose-phosphate-pathway dependent on glucose-6-phosphate and the last step of glutamine degradation – the conversion from malate to pyruvate. This suggests that glucose and glutamine are the major nutrients needed for proliferation in T cells.

AKT and AMPK in T cell Metabolism

The most prominent pathway responsible for upregulating glycolysis is the PI3K/AKT pathway. In T cells, coligation of the TCR and CD28 lead to direct phosphorylation of phosphatidylinositol-4,5- bisphosphate (PIP2) by phosphoinositide 3-kinase (PI3K) which leads to increased levels of phosphatidylinositol-3,4,5-trisphosphate (PIP3). AKT translocates to the plasma membrane by binding PIP3 via its PHdomain, where it can be phosphorylated by PDK1 on Thr308. For full activation, AKT requires additional phosphorylation on Ser473 by mTOR complex 2. It was shown for Tcells that sustained activation of AKT upregulates the surface expression of glucose transporter 1 (GLUT1) and increases the activity of the rate-limiting glycolytic enzyme hexokinase [4,17]. A previous study by McIntyre [18] reported that AKT had no effect on glucose uptake in CD8+ T cells. The authors of this study suggested that the upstream kinase PDK1 is responsible for increased glucose uptake and is therefore dispensable for CD8+ T cell metabolism. A study done in our lab came to the same conclusion when analyzing glucose uptake. But there was strong evidence that AKT is needed for upregulation of lactate dehydrogenase (unpublished results). The three major regulating enzymes of glycolysis are hexokinase, phosphofructokinase and pyruvate kinase. Although a link between AKT and pyruvate kinase was not investigated so far, the observations that AKT regulates the activity of hexokinase and phosphofructokinase [17] leads to the hypothesis that AKT is responsible for upregulating enzymes of glycolysis and lactate production. Additionally PDK1 or other members of the AGC kinase family can be responsible for increased glucose uptake in T cells. Since this contradicts the results of previous studies [4,11], the experimental conditions need to be critically discussed (Figure 1). The observations on the activating role of AKT were mostly performed in primary human T cells stimulated with CD3 and CD28, which fully activate AKT. This led to upregulation of GLUT1 expression and glucose uptake which could be inhibited by addition of cytotoxic T-lymphocyte antigen 4 (CTLA4) [4]. The recent studies observing a dispensable role of AKT were done in murine CD8+ T cells under physiological conditions using peptide stimulations without costimulation. Since it was shown before that CD8+ T cells do not require costimulation via CD28 [19] this might also lead to different outcomes in metabolic regulation. It was shown before under physiological conditions that the activation of AKT is sustained, but weak [20]. It might be that under these conditions a weak activation of AKT leads to compensatory mechanisms which also induce glucose uptake and GLUT1 upregulation. Possible targets of this mechanism could be the activation of PDK1 or the activation of the MAP-kinase ERK, which was also shown to be responsible for upregulated glucose uptake [21].

clinical-cellular-immunology-regulatory-functions

Figure 1: Different regulatory functions on AKT and PDK1 on human CD4 and murine CD8 T cells.
GLUT1: Glucose-Transporter 1; HK: Hexokinase; LDH: Lactate dehydrogenase.

Another important regulator of cellular metabolism is AMPK, which promotes ATP conservation and production through the activation of glycolysis, fatty acid oxidation, and the inhibition of ATP-consuming pathways, such as protein synthesis, fatty acid synthesis, gluconeogenesis, and glycogen synthesis [22,23]. AMPK can be activated by an increase in the AMP:ATP ratio followed by phosphorylation through LKB1 (a serine/threonine kinase). In addition it has been shown that Ca2+-calmodulin-dependent kinase kinase 2 (CAMKK2) can activate AMPK independent of AMP levels [24]. Recently, it was found that LKB1 is essential for the survival of thymocytes and development of T-cell progenitors and is required for CD4+ and CD8+ T-cell development [25,26]. LKB1-deficient peripheral T cells were shown to have enhanced glucose uptake and a higher glycolytic rate [25,26]. This suggests that LKB1/AMPK antagonize the PI3K/AKT/mTOR pathway, which promotes anabolism. This could be confirmed by the observations that AMPK inhibits mTOR activity [25] and that activation of AMPK was shown to be transient upon T cell stimulation [24]. Additionally, AMPK was shown to be required for memory T cell differentiation. Addition of the drug metformin caused an sustained activation of AMPK and subsequently led to increased numbers of memory T cells. Recent studies showing that LKB1/AMPK influences assymetric cell division in D. melanogaster [27-29], suggest that there could be a role for AMPK in the assymetric divisionT cells [30]. Since sustained activation of AKT is needed for effector T cell differentiation and AMPK activation appears to be only transient under these conditions, one could hypothesize that the contact of a T cell to an APC could also lead to a polarized distribution of metabolites. In this scenario the half of the T cell containing the immunological synapse would differentiate into an effector T cell, whereas the distal part would lead to memory T cell formation (Figure 2). While this hypothesis is attractive, it requires further investigation.

clinical-cellular-immunology-asymmetric-cell

Figure 2: Different metabolic programs in asymmetric cell division-AKT and AMPK influence the generation of effector and memory T cells.

Recently several studies have further analyzed the connection between the major metabolic regulators and metabolism. It was investigated whether transcription factors like HIF1a (hypoxia inducible factor1a) and MYC play an important role in expression of metabolic enzymes. Hif1a is a transcription factor that regulates the expression of genes that encode for glycolytic enzymes [31] as well as downregulates mitochondrial oxygen consumption by blocking the entrance of pyruvate into the TCA cycle [32]. Hif1a is constitutely active, but under normoxic conditions, it is rapidly degraded. Under low oxygen conditions the degradation of Hif1a is inhibited and it translocates to the nucleus where it upregulates glycolytic genes and drives the cell towards aerobic glycolysis. A recent study showed that the activation of CD4+ and CD8+ T cells leads to a strong, but transient induction of HIF1a. However, this activation was not responsible for metabolic changes in these cells and was not critical for proliferation [33]. In addition, another study observed that HIF1a is strongly induced only in the TH17 subset [34]. Another factor investigated was the proto-oncogenic transcription factor Myc, which was shown to be induced upon T cell stimulation [15]. The deletion of Myc lead to an impaired upregulation of glycolysis and glutaminolysis, and a decreased activation of targets downstream of mTOR. These observations led to the conclusion that Myc is probably the major transcription factor regulating T cell metabolism upon activation. Taking together all studies on T cell metabolism, it becomes clear that different T cell subsets have different metabolic profiles. CD4+ and CD8+ effector cells show strong activation of glycolysis and lactate production, mediated via PI3K/AKT pathway, which correlates with their ability to proliferate and produce cytokines that promote a productive immune response. In contrast, both regulatory T cells (Tregs) and memory T cells fail to upregulate glucose metabolism [35]. Their demand for glucose is much lower and is replaced by using lipids as an energy source through ß-oxidation. This is also compatible with their function, as Tregs and memory T cells are long-lived cells witha slow rate of replication.

Summary

Primary T cells are able to upregulate metabolism from a quiescent to an activated one in order to maintain their energetic needs for proliferation, differentiation, and cytokine production. The PI3K/ AKT pathway plays a central role in regulating T cell metabolism. Understanding T cell metabolism provides insight into how T cells can deal with their energetic needs and how this may affect their function. The ability of T cells to switch between states of low and high energy consumption, which then in turn drives them towards their specific function, shows the interplay between signalling events and the metabolic program. Improving our understanding as to how these processes are regulated will not only provide insight in to how immune cells function, but it may also reveal targets for suppressing T cell-mediated autoimmune diseases or provide tools to improving the immune response.

Acknowledgments

B.S., and J.A.L. are members of the Magdeburg Center for Systems Biology (MaCS) and B.S. and J.A.L. are members of the SYBILLA consortium [European Union 7th Frame Program]. This work was supported in part by grants from the German Research Society (DFG) [JL 1031/1-3], the German Ministry of Education and Research (BMBF) FOR-SYS program [0313922], the State of Sachsen-Anhalt (Dynamic Systems) [XD3639HP/0306], and the European Union 7th Frame Program (SYBILLA) [HEALTH-F4-2008-201106].

Conflict of Interest

The authors declare no competing financial interest.

References

  1. Krauss S, Brand MD, Buttgereit F (2001) Signaling takes a breath--new quantitative perspectives on bioenergetics and signal transduction. Immunity 15: 497-502.
  2. Fox CJ, Hammerman PS, Thompson CB (2005) Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol 5: 844-852.
  3. Maciver NJ, Jacobs SR, Wieman HL, Wofford JA, Coloff JL, et al. (2008) Glucose metabolism in lymphocytes is a regulated process with significant effects on immune cell function and survival. J Leukoc Biol 84: 949-957.
  4. Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, et al. (2002) The CD28 signaling pathway regulates glucose metabolism. Immunity 16: 769-777.
  5. Jones RG, Thompson CB (2007) Revving the engine: signal transduction fuels T cell activation. Immunity 27: 173-178.
  6. Andres R, cader G, zierler KL (1955) Metabolic exchange of human muscle in situ. Am J Phys Med 34: 286-290.
  7. Warburg O (1956) On respiratory impairment in cancer cells. Science 124: 269-270.
  8. Vander Heiden MG, Cantley LC, Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324: 1029-1033.
  9. Peter C, Waldmann H, Cobbold SP (2010) MTOR signalling and metabolic regulation of T cell differentiation. Curr Opin Immunol 22: 655-661.
  10. Powell JD, Delgoffe GM (2010) The mammalian target of rapamycin: linking T cell differentiation, function, and metabolism. Immunity 33: 301-311.
  11. Jacobs SR, Herman CE, Maciver NJ, Wofford JA, Wieman HL, et al. (2008) Glucose uptake is limiting in T cell activation and requires CD28-mediated Akt-dependent and independent pathways. J Immunol 180: 4476-4486.
  12. Cham CM, Gajewski TF (2005) Glucose availability regulates IFN-gamma production and p70S6 kinase activation in CD8+ effector T cells. J Immunol 174: 4670-4677.
  13. Alves NL, Derks IA, Berk E, Spijker R, van Lier RA, et al. (2006) The Noxa/Mcl-1 axis regulates susceptibility to apoptosis under glucose limitation in dividing T cells. Immunity 24: 703-716.
  14. Brand K, Aichinger S, Forster S, Kupper S, Neumann B, et al. (1988) Cell-cycle-related metabolic and enzymatic events in proliferating rat thymocytes. Eur J Biochem 172: 695-702.
  15. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, et al. (2011) The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35: 871-882.
  16. Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch E, et al. (2010) Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. J Immunol 185: 1037-1044.
  17. Gottlob K, Majewski N, Kennedy S, Kandel E, Robey RB, et al. (2001) Inhibition of early apoptotic events by Akt/PKB is dependent on the first committed step of glycolysis and mitochondrial hexokinase. Genes Dev 15: 1406-1418.
  18. Macintyre AN, Finlay D, Preston G, Sinclair LV, Waugh CM, et al. (2011) Protein kinase B controls transcriptional programs that direct cytotoxic T cell fate but is dispensable for T cell metabolism. Immunity 34: 224-236.
  19. Wang B, Maile R, Greenwood R, Collins EJ, Frelinger JA (2000) Naive CD8+ T cells do not require costimulation for proliferation and differentiation into cytotoxic effector cells. J Immunol 164: 1216-1222.
  20. Wang X, Simeoni L, Lindquist JA, Saez-Rodriguez J, Ambach A, et al. (2008) Dynamics of proximal signaling events after TCR/CD8-mediated induction of proliferation or apoptosis in mature CD8+ T cells. J Immunol 180: 6703-6712.
  21. Marko AJ, Miller RA, Kelman A, Frauwirth KA (2010) Induction of glucose metabolism in stimulated T lymphocytes is regulated by mitogen-activated protein kinase signaling. PLoS One 5: e15425.
  22. Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, et al. (2004) The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A 101: 3329-3335.
  23. Mihaylova MM, Shaw RJ (2011) The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol 13: 1016-1023.
  24. Tamás P, Hawley SA, Clarke RG, Mustard KJ, Green K, et al. (2006) Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes. J Exp Med 203: 1665-1670.
  25. MacIver NJ, Blagih J, Saucillo DC, Tonelli L, Griss T, et al. (2011) The liver kinase B1 is a central regulator of T cell development, activation, and metabolism. J Immunol 187: 4187-4198.
  26. Cao Y, Li H, Liu H, Zheng C, Ji H, et al. (2010) The serine/threonine kinase LKB1 controls thymocyte survival through regulation of AMPK activation and Bcl-XL expression. Cell Res 20: 99-108.
  27. Lee JH, Koh H, Kim M, Kim Y, Lee SY, et al. (2007) Energy-dependent regulation of cell structure by AMP-activated protein kinase. Nature 447: 1017-1020.
  28. Mirouse V, Swick LL, Kazgan N, St Johnston D, Brenman JE (2007) LKB1 and AMPK maintain epithelial cell polarity under energetic stress. J Cell Biol 177: 387-392.
  29. Martin SG, St Johnston D (2003) A role for Drosophila LKB1 in anterior-posterior axis formation and epithelial polarity. Nature 421: 379-384.
  30. Chang JT, Palanivel VR, Kinjyo I, Schambach F, Intlekofer AM, et al. (2007) Asymmetric T lymphocyte division in the initiation of adaptive immune responses. Science 315: 1687-1691.
  31. Semenza GL (2009) Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology (Bethesda) 24: 97-106.
  32. Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC (2006) HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab 3: 187-197.
  33. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, et al. (2011) Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186: 3299-3303.
  34. Shi LZ, Wang R, Huang G, Vogel P, Neale G, et al. (2011) HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med 208: 1367-1376.
  35. Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, et al. (2009) Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460: 103-107.
Citation: Cammann C, Schraven B, Lindquist JA (2012) T cell Metabolism– Regulating Energy. J Clin Cell Immunol S12:012

Copyright: © 2012 Cammann C, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top