Metabolism in T cell activation and differentiation

https://doi.org/10.1016/j.coi.2010.01.018Get rights and content

When naïve or memory T cells encounter foreign antigen along with proper co-stimulation they undergo rapid and extensive clonal expansion. In mammals, this type of proliferation is fairly unique to cells of the adaptive immune system and requires a considerable expenditure of energy and cellular resources. While research has often focused on the roles of cytokines, antigenic signals, and co-stimulation in guiding T cell responses, data indicate that, at a fundamental level, it is cellular metabolism that regulates T cell function and differentiation and therefore influences the final outcome of the adaptive immune response. This review will focus on some earlier fundamental observations regarding T cell bioenergetics and its role in regulating cellular function, as well as recent work that suggests that manipulating the immune response by targeting lymphocyte metabolism could prove useful in treatments against infection and cancer.

Introduction

Unlike the case for unicellular organisms where dramatic changes in nutrient availability affect proliferation, mammalian cells reside in nutrient-rich environments where cellular proliferation is controlled by extrinsic signals, such as growth factors, which regulate nutrient utilization [1]. One of the most striking changes to affect T cells after initial antigenic stimulation is an increase in cell size accompanied by a metabolic switch to glycolysis, which is required to support their growth, proliferation, and effector functions [2, 3, 4] (Figure 1). During TCR stimulation, signals from growth factor cytokines like IL-2, and the ligation of co-stimulatory CD28, lead to an increase in glycolysis by inducing the PI3K-dependent activation of Akt [5, 6]. Activated Akt can promote the mTOR (mammalian target of rapamycin) pathway, a key regulator of translation [7], as well as stimulate glycolysis by increasing glycolytic enzyme activity and enhancing the expression of nutrient transporters, enabling increased utilization of glucose and amino acids [8, 9, 10••, 11, 12, 13]. Together these changes lead to the increase in nutrient utilization and glucose metabolism that facilitates activation and proliferation.

As T cells undergo clonal expansion they preferentially ferment glucose to meet their energy demands, even though there is sufficient oxygen present to support mitochondrial oxidative phosphorylation [14, 15, 16]. This phenomenon is known as the Warburg effect [17], and is an unusual metabolic aspect of proliferating T cells and cancer cells. Since ATP production by aerobic glycolysis is much less efficient than by oxidative phosphorylation, a question remains as to why proliferating T cells favor this form of metabolism. One explanation, largely based on observations from Craig Thompson's laboratory, is that glycolysis is an essentially anabolic form of metabolism that leaves cellular building blocks, such as amino acids and fatty acids untouched, as well as produces lactate, all of which can be incorporated into cellular components [18]. A cell that converts building blocks into biomass most efficiently will proliferate the fastest and in a host fighting an infection, rapid expansion of antigen-specific T cells could offer a decisive advantage [19].

In contrast to proliferating T cells, quiescent T cells (i.e. naïve and memory cells), like most cells in normal tissues, interchangeably breakdown glucose, amino acids, and lipids to catabolically fuel ATP generation [2, 18] (Figure 1). The posited effects of growth factors on resting T cell survival are related to their ability to modulate the surface expression of nutrient transporters [20]. Quiescent cells can also use autophagy (the break down of intracellular components) to supply the molecules to fuel oxidative phosphorylation [21]. There is growing evidence that quiescence is under active transcriptional control [22]. TOB1 (transducer of ERBB2 1) [23], LKLF (lung Krüppel-like factor) [24], and FOXO (Forkhead box class O) transcription factors all have been suggested to promote quiescence in lymphocytes by actively maintaining the expression of inhibitors of cellular activation [25, 26]. Furthermore, FOXO transcription factors have been shown to modulate metabolic functions [27, 28] and the family of Krüppel-like factors (KLFs) has been shown to regulate adipocyte differentiation and glucose homeostasis in mammals [29], which may suggest a degree of metabolic control in maintaining quiescence.

Implicit in the striking divergence of metabolic phenotypes between proliferating and quiescent T cells is the idea that the conversion, or switching, between differing metabolic states is required to effectively generate a given T cell fate [10••, 18]. This not only applies to the switch from quiescence to glycolysis that accompanies naïve T cell activation, but also to the promotion of catabolism that appears to be important for the generation of quiescent memory T cells after infection [30••] (Figure 2). Each of these metabolic states represents a unique target of intervention for manipulating the T cell response and ameliorating disease.

Section snippets

Metabolic regulation of T cell responses

It is well documented that proliferating T cells require glucose to survive, and in the absence of glucose they cannot support their bioenergetic demands and undergo apoptosis [4, 31, 32]. Much less is known about how lipid metabolism regulates T cell responses. However, in a recent study of great interest, Bensinger et al. examined the influence of cellular lipid metabolism on the immune response and showed that LXR (Liver X receptor), a member of the nuclear receptor family of transcription

Harnessing immunity through metabolism

The fact that metabolism underlies the functional capacity of a T cell suggests that altering cellular metabolism may influence the final outcome of the adaptive response. In addition, lymphocytes may be particularly amenable to metabolic manipulation since their development is marked by striking changes in metabolism. Since clearance or control of pathogens or tumors usually requires T cell mediated immunity, the ability to deliberately manipulate T cell responses by regulating metabolism

T cell life span: parallels with organismal life span

Immunological memory is the basis of vaccination, and promoting the generation of long-lived memory T cells is a focus of vaccine development. The greatest insights into the molecular pathways that control longevity have come from studies in worms, yeast, and fruit flies. This work has shown that insulin/IGF signaling, DAF-16/FOXO transcription factors, and TOR all play crucial roles in regulating metabolism as well as life span [65, 66, 67, 68, 69, 70]. The cellular response to environmental

Metabolic contributions

The question as to precisely what fluctuations in metabolism provide to T cells during different phases of an immune response remains incompletely answered. One assumption is that switching to catabolic forms of metabolism in the face of metabolic stress, that is, after infection is cleared and associated signals dissipate, is a survival mechanism that provides energy in the form of ATP, and that this allows T cells to assume quiescence and become long-lived. However, it may be more complicated

Conclusion

Successful disease prevention or therapy often depends on T cells performing to their expected capacity. The ability to alter T cell performance could prove useful in settings of prophylactic vaccination where the induction of a population of long-lived memory T cells is the goal, or in cases of therapeutic cancer vaccines, where effective tumor therapy depends on the reliable ex vivo expansion of a patient's T cells. If we can manipulate T cell metabolism, and many drugs that do this are

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

I would like to thank Edward Pearce, Matt Walsh, Pedro Cejas, Rusty Jones, and Yongwon Choi who contributed to the ideas and discussions that are the basis of this review.

The author's laboratory is supported by a grant from the Emerald Foundation.

References (81)

  • L. Klotz et al.

    The nuclear receptor PPAR gamma selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity

    J Exp Med

    (2009)
  • M. Prlic et al.

    Exploring regulatory mechanisms of CD8+ T cell contraction

    Proc Natl Acad Sci U S A

    (2008)
  • S.H. Um et al.

    Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity

    Nature

    (2004)
  • T. Peng et al.

    The immunosuppressant rapamycin mimics a starvation-like signal distinct from amino acid and glucose deprivation

    Mol Cell Biol

    (2002)
  • R.J. Shaw et al.

    The LKB1 tumor suppressor negatively regulates mTOR signaling

    Cancer Cell

    (2004)
  • J. Kerner et al.

    Fatty acid import into mitochondria

    Biochim Biophys Acta

    (2000)
  • K. Inoki et al.

    TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth

    Cell

    (2006)
  • R.L. Rutishauser et al.

    Transcriptional repressor Blimp-1 promotes CD8(+) T cell terminal differentiation and represses the acquisition of central memory T cell properties

    Immunity

    (2009)
  • A. Kallies et al.

    Blimp-1 transcription factor is required for the differentiation of effector CD8(+) T cells and memory responses

    Immunity

    (2009)
  • H. Shin et al.

    A role for the transcriptional repressor Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection

    Immunity

    (2009)
  • M. Heffner et al.

    Loss of T cell receptor-induced Bmi-1 in the KLRG1(+) senescent CD8(+) T lymphocyte

    Proc Natl Acad Sci U S A

    (2007)
  • M. Yamashita et al.

    Bmi1 regulates memory CD4 T cell survival via repression of the Noxa gene

    J Exp Med

    (2008)
  • J. Liu et al.

    Bmi1 regulates mitochondrial function and the DNA damage response pathway

    Nature

    (2009)
  • P. Kapahi et al.

    Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway

    Curr Biol

    (2004)
  • P. Kapahi et al.

    TOR pathway: linking nutrient sensing to life span

    Sci Aging Knowledge Environ

    (2004)
  • M. Kaeberlein et al.

    Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients

    Science

    (2005)
  • P. Kapahi et al.

    Positive correlation between mammalian life span and cellular resistance to stress

    Free Radic Biol Med

    (1999)
  • V.N. Anisimov et al.

    Insulin and longevity: antidiabetic biguanides as geroprotectors

    Biogerontology

    (2003)
  • M.C. Wang et al.

    Fat metabolism links germline stem cells and longevity in C. elegans

    Science

    (2008)
  • M. Kaeberlein et al.

    Cell signaling. Aging is RSKy business

    Science

    (2009)
  • H.L. Wieman et al.

    Cytokine stimulation promotes glucose uptake via phosphatidylinositol-3 kinase/Akt regulation of Glut1 activity and trafficking

    Mol Biol Cell

    (2007)
  • A.C. Gingras et al.

    Regulation of translation initiation by FRAP/mTOR

    Genes Dev

    (2001)
  • J.T. Barata et al.

    Activation of PI3K is indispensable for interleukin 7-mediated viability, proliferation, glucose use, and growth of T cell acute lymphoblastic leukemia cells

    J Exp Med

    (2004)
  • R.L. Elstrom et al.

    Akt stimulates aerobic glycolysis in cancer cells

    Cancer Res

    (2004)
  • J.C. Rathmell et al.

    Activated Akt promotes increased resting T cell size. CD28-independent T cell growth, and development of autoimmunity and lymphoma

    Eur J Immunol

    (2003)
  • A.L. Edinger et al.

    Akt maintains cell size and survival by increasing mTOR-dependent nutrient uptake

    Mol Biol Cell

    (2002)
  • R.J. DeBerardinis et al.

    The biology of cancer: metabolic reprogramming fuels cell growth and proliferation

    Cell Metab

    (2008)
  • E.F. Greiner et al.

    Glucose is essential for proliferation and the glycolytic enzyme induction that provokes a transition to glycolytic energy production

    J Biol Chem

    (1994)
  • O. Warburg

    On the origin of cancer cells

    Science

    (1956)
  • C.J. Fox et al.

    Fuel feeds function: energy metabolism and the T-cell response

    Nat Rev Immunol

    (2005)
  • Cited by (226)

    • Metabolic dysregulation of lymphocytes in autoimmune diseases

      2024, Trends in Endocrinology and Metabolism
    View all citing articles on Scopus
    View full text