mTOR and lymphocyte metabolism

https://doi.org/10.1016/j.coi.2013.05.002Get rights and content

Highlights

  • Different T cell activation states have distinct metabolic profiles and mTOR activity.

  • mTOR orchestrates T cell quiescence, functional activation, and fate decisions.

  • mTOR is activated by and impinge upon antigen receptor and other immune signaling.

  • mTOR senses metabolic cues and coordinates T cell metabolic reprogramming.

Upon antigen engagement and proper co-stimulation, naïve lymphocytes exit quiescence and undergo clonal expansion and differentiate into functional effector cells, after which they either die through apoptosis or survive as memory cells. Lymphocytes at different activation stages exhibit distinct metabolic signatures. Emerging evidence highlights a central role for the mechanistic target of rapamycin (mTOR) in bridging immune signals and metabolic cues to direct lymphocyte proliferation, differentiation and survival. Here we review recent advances in understanding the functional significance and signal transduction of mTOR in T cell biology, and the interplay between mTOR signaling and metabolic programs.

Introduction

Mammalian cells are usually exposed to a constant supply of nutrients, but they do not normally take up nutrients and proliferate until they are stimulated by extrinsic factors [1]. Naïve lymphocytes, like most cells in normal tissues, have a quiescent status, in which they primarily rely on catabolic metabolism and derive most of their ATP from oxidative phosphorylation, particularly fatty acid β-oxidation [2, 3, 4••]. Quiescent lymphocytes also break down intracellular components through autophagy to supply molecules for oxidative phosphorylation [5]. Upon antigen recognition and co-stimulation, lymphocytes downregulate fatty acid β-oxidation and rapidly increase glycolytic, glutaminolytic and pentose phosphate pathways to provide biosynthetic materials and energy for cell growth and proliferation [3, 4••, 6]. Activated and effector T cells preferentially utilize aerobic glycolysis to meet their energy demands, a phenomenon known as the Warburg effect, which is also a metabolic feature of many cancer cells [1]. After clonal expansion and clearance of invading foreign pathogens, most effector T cells undergo apoptosis while some differentiate into long-lived memory cells. Memory T cells, like naïve T cells, are quiescent and have a catabolic metabolism [7, 8]. A separate T cell subset, FOXP3+ regulatory T cells (Treg), also exhibits relatively high fatty acid β-oxidation but low glycolysis [9••, 10]. Thus, during immune responses, T cells experience two major metabolic switches, from catabolic naïve T cells to anabolic activated/effector T cells and then again transition into catabolic memory T cells (Figure 1). Emerging evidence indicates that metabolism is closely coupled with the differentiation and function of T cells at different stages of their life span [11].

The serine/threonine kinase mTOR consists of two distinct complexes: mTOR complex 1 (mTORC1) and 2 (mTORC2). Two scaffold proteins, regulatory associated protein of mTOR (RAPTOR) and rapamycin-insensitive companion of mTOR (RICTOR), are the defining components of mTORC1 and mTORC2, respectively [12]. While mTORC1 is sensitive to rapamycin, mTORC2 can be inhibited by prolonged or high dose of rapamycin treatment in CD4+ T cells [13••, 14••], but not in effector CD8+ T cells [15]. Many upstream signals activate mTORC1 pathway through the small GTPase RHEB (RAS homologue enriched in brain). The tuberous sclerosis 1 (TSC1) and TSC2 form a complex that inactivates RHEB through its GAP (GTPase-activating protein) activity, thereby suppressing mTORC1 activity. Further upstream, the PI3K-AKT pathway inactivates TSC1/TSC2 complex while AMP-activated protein kinase (AMPK) enhances its activity. Therefore, TSC1/TSC2 complex functions as a molecular switch that controls mTORC1 activity. S6K1 and 4E-BP1 are two best-characterized downstream targets of mTORC1 that regulates protein translation. Moreover, mTORC1 pathway also promotes glycolysis and lipid biosynthesis while inhibiting autophagy. mTORC2 is activated by PI3K signaling, but detailed mechanism is lacking. mTORC2 controls several AGC family kinases, including AKT, SGK1 and PKC-α and is involved in regulating metabolism, apoptosis and cytoskeletal organization [12]. In particular, phosphorylation of AKT-Ser473 by mTORC2 promotes FOXO1/3a phosphorylation and subsequent cytoplasm translocation and degradation [16, 17].

In lymphocytes, diverse environmental signals, including antigens, growth factors, cytokines and nutrients regulate mTOR to direct immune responses and fate decisions [18, 19]. Since the roles of mTOR and metabolic pathways have been extensively studied in mature T cells in the periphery, we will mainly focus on these cells. First, we will briefly describe the roles of mTOR in T cell homeostasis under steady state and antigen-triggered activation and differentiation. Second, we will discuss the functional effects and mechanistic basis of mTOR in sensing and propagating diverse immune signals, especially those mediated by TCR, co-stimulation and cytokine receptors. Third, we will present the emerging evidence on mTOR-dependent metabolic reprogramming of T cell responses, by focusing on the interaction between mTOR and transcription factors associated with cell metabolism such as MYC and HIF1, and the potential interplay between mTOR-controlled metabolites and immune signaling. As our discussion focuses on T cells, we refer the readers to an excellent recent review describing the PI3K-AKT-mTOR pathway in B cells [20].

Section snippets

mTOR in T cell quiescence

The quiescent status of naïve T cells is not a default state determined by the lack of mitogenic stimuli, but is an actively maintained process [7]. Uncontrolled mTORC1 activation by TSC1 deletion in T cells leads to loss of quiescence and predisposes T cells to apoptotic death [21, 22, 23]. Consequently, Tsc1−/− mice have markedly reduced peripheral T cell numbers. TSC1-deficient T cells exhibit semi-activated phenotypes, with a larger cell size, increased metabolic gene expression and cell

mTOR and regulation of immune signals

mTOR activity can be positively or negatively regulated by multiple inputs (Figure 2). The classic model posits that three major signals are required for proper T cell activation and differentiation: TCR engagement by antigen-MHC complex, costimulatory signals, and inflammatory cytokines. These signals, together with additional immune-modulatory receptors including Toll-like receptors (TLRs) and G protein-coupled receptors (GPCRs), are sensed and integrated by mTOR; once activated, mTOR can in

mTOR is activated by hormones and nutrients

mTOR signaling is intimately linked with cellular metabolism. Hormones and nutrients are important factors that modulate systemic and cellular metabolism and they feed into the mTOR pathway. Leptin is an adipocyte-derived hormone that controls food intake and metabolism. Recent studies have demonstrated that leptin-induced mTOR activation is critical for effector T cell proliferation [52], whereas it maintains the in vitro anergic status of Treg and negatively controls Treg proliferation [69, 70

Conclusion

T cells are at the center of adaptive immunity that protects the body from pathogen infections, or mediates self-destructive autoimmune diseases. mTOR integrates immune signals and metabolic cues to direct T cell homeostatic and functional fates, and this is shaped by the extensive interplay between mTOR signaling and cell metabolism. Despite the recent remarkable advances, a number of questions remain to be answered. We have yet to fully understand mTOR-associated upstream signal inputs and

Competing interest statement

The authors declare no competing financial interests.

References and recommended reading

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

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We acknowledge the large number of researchers who have contributed to this field whose work was not cited owing to space limitations. The authors’ research is supported by US National Institutes of Health (R01 NS064599 and R21 AI094089), National Multiple Sclerosis Society, Lupus Research Institute, and the American Lebanese Syrian Associated Charities (HC).

References (87)

  • R.R. Rao et al.

    The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin

    Immunity

    (2010)
  • Y.M. Kerdiles et al.

    Foxo1 links homing and survival of naive T cells by regulating L-selectin, CCR7 and interleukin 7 receptor

    Nat Immunol

    (2009)
  • J.D. Powell et al.

    The mammalian target of rapamycin: linking T cell differentiation, function, and metabolism

    Immunity

    (2010)
  • L.M. Francisco et al.

    PD-L1 regulates the development, maintenance, and function of induced regulatory T cells

    J Exp Med

    (2009)
  • J.E. Smith-Garvin et al.

    T cell activation

    Annu Rev Immunol

    (2009)
  • C. Procaccini et al.

    Leptin-induced mTOR activation defines a specific molecular and transcriptional signature controlling CD4+ effector T cell responses

    J Immunol

    (2012)
  • R. Tomasoni et al.

    Rapamycin-sensitive signals control TCR/CD28-driven Ifng, Il4 and Foxp3 transcription and promoter region methylation

    Eur J Immunol

    (2011)
  • S. Spiegel et al.

    The outs and the ins of sphingosine-1-phosphate in immunity

    Nat Rev Immunol

    (2011)
  • A.W. Thomson et al.

    Immunoregulatory functions of mTOR inhibition

    Nat Rev Immunol

    (2009)
  • C. Procaccini et al.

    An oscillatory switch in mTOR kinase activity sets regulatory T cell responsiveness

    Immunity

    (2010)
  • E.V. Schmidt et al.

    Growth controls connect: interactions between c-myc and the tuberous sclerosis complex-mTOR pathway

    Cell Cycle

    (2009)
  • H. Nakamura et al.

    TCR engagement increases hypoxia-inducible factor-1 alpha protein synthesis via rapamycin-sensitive pathway under hypoxic conditions in human peripheral T cells

    J Immunol

    (2005)
  • E.V. Dang et al.

    Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1

    Cell

    (2011)
  • K.E. Wellen et al.

    A two-way street: reciprocal regulation of metabolism and signalling

    Nat Rev Mol Cell Biol

    (2012)
  • S. Kagami et al.

    Protein geranylgeranylation regulates the balance between Th17 cells and Foxp3+ regulatory T cells

    Int Immunol

    (2009)
  • N. Ladygina et al.

    Dynamic palmitoylation and the role of DHHC proteins in T cell activation and anergy

    Adv Immunol

    (2011)
  • M.G. Vander Heiden et al.

    Understanding the Warburg effect: the metabolic requirements of cell proliferation

    Science

    (2009)
  • J.J. Lum et al.

    Autophagy in metazoans: cell survival in the land of plenty

    Nat Rev Mol Cell Biol

    (2005)
  • C.A. Doughty et al.

    Antigen receptor-mediated changes in glucose metabolism in B lymphocytes: role of phosphatidylinositol 3-kinase signaling in the glycolytic control of growth

    Blood

    (2006)
  • E.L. Pearce

    Metabolism in T cell activation and differentiation

    Curr Opin Immunol

    (2010)
  • R.D. Michalek et al.

    Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets

    J Immunol

    (2011)
  • R. Wang et al.

    Metabolic checkpoints in activated T cells

    Nat Immunol

    (2012)
  • M. Laplante et al.

    mTOR signaling in growth control and disease

    Cell

    (2012)
  • K. Lee et al.

    Mammalian target of rapamycin protein complex 2 regulates differentiation of Th1 and Th2 cell subsets via distinct signaling pathways

    Immunity

    (2010)
  • W. Ouyang et al.

    Foxo: in command of T lymphocyte homeostasis and tolerance

    Trends Immunol

    (2011)
  • S.M. Hedrick et al.

    FOXO transcription factors throughout T cell biology

    Nat Rev Immunol

    (2012)
  • H. Chi

    Regulation and function of mTOR signalling in T cell fate decisions

    Nat Rev Immunol

    (2012)
  • J.D. Powell et al.

    Regulation of immune responses by mTOR

    Annu Rev Immunol

    (2012)
  • T.F. O’Brien et al.

    Regulation of T-cell survival and mitochondrial homeostasis by TSC1

    Eur J Immunol

    (2011)
  • Q. Wu et al.

    The tuberous sclerosis complex-mammalian target of rapamycin pathway maintains the quiescence and survival of naive T cells

    J Immunol

    (2011)
  • K. Yang et al.

    The tumor suppressor Tsc1 enforces quiescence of naive T cells to promote immune homeostasis and function

    Nat Immunol

    (2011)
  • R. Zoncu et al.

    mTOR: from growth signal integration to cancer, diabetes and ageing

    Nat Rev Mol Cell Biol

    (2011)
  • J.J. O'Shea et al.

    Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells

    Science

    (2010)
  • Cited by (79)

    • Innate and adaptive immunity in cancer

      2021, Cancer Immunology and Immunotherapy: Volume 1 of Delivery Strategies and Engineering Technologies in Cancer Immunotherapy
    • Exercise immunology: Future directions

      2020, Journal of Sport and Health Science
    View all citing articles on Scopus
    View full text