Elsevier

Human Immunology

Volume 74, Issue 12, December 2013, Pages 1501-1509
Human Immunology

Inhibition of indoleamine 2,3-dioxygenase in mixed lymphocyte reaction affects glucose influx and enzymes involved in aerobic glycolysis and glutaminolysis in alloreactive T-cells

https://doi.org/10.1016/j.humimm.2013.08.268Get rights and content

Abstract

Indoleamine 2,3-dioxygenase (IDO) suppresses adaptive immunity. T-cell proliferation and differentiation to effector cells require increased glucose consumption, aerobic glycolysis and glutaminolysis. The effect of IDO on the above metabolic pathways was evaluated in alloreactive T-cells. Mixed lymphocyte reaction (MLR) in the presence or not of the IDO inhibitor, 1-methyl-dl-tryptophan (1-MT), was used. In MLRs, 1-MT decreased tryptophan consumption, increased cell proliferation, glucose influx and lactate production, whereas it decreased tricarboxylic acid cycle activity. In T-cells, from the two pathways that could sense tryptophan depletion, i.e. general control nonrepressed 2 (GCN2) kinase and mammalian target of rapamycin complex 1, 1-MT reduced only the activity of the GCN2 kinase. Additionally 1-MT treatment of MLRs altered the expression and/or the phosphorylation state of glucose transporter-1 and of key enzymes involved in glucose metabolism and glutaminolysis in alloreactive T-cells in a way that favors glucose influx, aerobic glycolysis and glutaminolysis. Thus in alloreactive T-cells, IDO through activation of the GCN2 kinase, decreases glucose influx and alters key enzymes involved in metabolism, decreasing aerobic glycolysis and glutaminolysis. Acting in such a way, IDO could be considered as a constraining factor for alloreactive T-cell proliferation and differentiation to effector T-cell subtypes.

Introduction

Indoleamine 2,3-dioxygenase (IDO) catalyzes the initial rate-limiting step of tryptophan degradation along the kynurenine pathway. IDO is inducible by various inflammatory stimuli and it is expressed in antigen presenting cells (APCs), such as monocytes, macrophages and dendritic cells. In the local microenvironment of inflammation, IDO expression in APCs depletes tryptophan and suppresses adaptive immunity [1], [2]. Amino acid depletion is sensed by T-cells through two pathways. Tryptophan depletion by IDO activates the general control nonrepressed 2 (GCN2) kinase, which in turn phosphorylates the eukaryotic translation initiation factor 2α (eIF2α) ultimately altering the translation program of the T-cells [3]. Another pathway able to sense tryptophan depletion is the mammalian target of rapamycin complex 1 (mTORC1) pathway. T-cells fail to proliferate in response to antigen once tryptophan or other essential amino acid becomes sparse, associated with a reduced mTORC1 signaling [4].

Experimental studies showed that IDO mediated immunosuppression ameliorates the clinical course of autoimmune diseases [5], [6], [7] and reduces graft rejection [8], [9], [10]. Inhibition of T-cell function via IDO is also mediated by non-APC cell types. Expression of IDO in tumor cells contributes to escape of tumor by immunosurveillance [11], while its expression in paternally derived placental trophoblast contributes to a successful semi-allogenic pregnancy [12], [13]. Hemodialysis patients are characterized by impaired adaptive immunity and exhibit increased IDO expression, further enhanced in the non-responders to hepatitis B virus vaccination [14]. Interestingly in the above population plasma IDO concentration is inversely related to blood T-cell count [15]. IDO exerts its immunosuppressive action in both CD4+ and CD8+ T-cells [16]. Among others, IDO suppresses adaptive immunity by affecting T-cell differentiation, for instance the differentiation of latter to regulatory T-cells (Treg) [17], [18], [19], [20], [21].

Activated T-cells have increased metabolic requirements in order to support proliferation and effector function. Increased glucose uptake and aerobic glycolysis fuel this demand [22], [23]. Effector CD4+ T-cells require distinct metabolic programs to support their function in opposition to Treg. More precisely, effector CD4+ T-cells express high levels of the glucose transporter-1 (GLUT1) and are reliant on glucose metabolism, whereas Treg express low levels of GLUT1 and are reliant on lipid oxidation. Direct manipulation of cell metabolism determines the fate of CD4+ T-cells [24], [25]. An elegant study showed that in order to fulfill the bioenergetic and biosynthetic demand of proliferation, activated T-cells reprogram their metabolic pathways from pyruvate oxidation and fatty acid β-oxidation via the tricarboxylic acid (TCA) cycle to the glycolytic, pentose-phosphate, and glutaminolytic pathways [26]. Interestingly, in a previous study the IDO inhibitor 1-methyl-dl-tryptophan(1-MT) suppresses mitochondrial function and induces aerobic glycolysis in peripheral blood mononuclear cells (PBMCs) during an immune response against tetanus toxoid [27].

In this study we evaluated the effect of IDO on T-cell metabolism, since IDO affects T-cell activation and differentiation [16], [17], [18], [19], [20], and direct manipulation of glucose metabolism modulates lymphocyte survival, proliferation, differentiation and function [24], [25]. For this purpose the model of alloreactivity, two-way mixed lymphocyte reaction (MLR) [28], and the specific IDO inhibitor 1-MT were used. 1-MT is a competitive, non-toxic IDO inhibitor [29] that has been successfully used to break immune privilege of placenta and tolerance against grafts [8], [12].

Section snippets

Subjects

Blood samples from 10 healthy volunteers (7 men, 37 ± 9 years old) were collected for the evaluation of 1-MT cytotoxicity in resting PMBCs. Blood samples from five non-relative healthy volunteers (all men, 34 ± 8 years old) were also collected in order to perform 10 couples of MLRs. An informed consent was obtained from each individual enrolled into the study and the hospital ethics committee gave its approval to the study protocol.

Peripheral blood mononuclear cell isolation and culture

PBMCs were isolated from whole blood by Ficoll–Hypaque density

1-MT was almost non-toxic in resting peripheral blood mononuclear cells

In resting untreated PBMCs, LDH release assay revealed a cytotoxicity of 11.92 ± 0.73%. In PBMCs treated with 100 μM 1-MT, cytotoxicity was slightly higher 14.12 ± 0.49% (p < 0.001) (Fig. 1A). Despite the statistical significance, the 2% increase in cytotoxicity due to 1-MT treatment compared to spontaneous cell death indicates that 1-MT at the used concentration is almost non-toxic for PMBCs.

1-MT decreased tryptophan consumption, increased proliferation, glucose consumption, aerobic glycolysis and decreased TCA cycle activity in two-way mixed lymphocyte reaction

1-MT decreased l-tryptophan consumption significantly. l-tryptophan concentration in the supernatants of MLRs

Discussion

Indoleamine 2,3-dioxygenase expression in APCs, and more specifically in monocytes in the case of MLR, depletes tryptophan and suppresses adaptive immunity [1], [2]. It is also known that activated T cells in order to fulfill the bioenergetic and biosynthetic demand of proliferation, reprogram their metabolic pathways from pyruvate oxidation and fatty acid β-oxidation via the TCA cycle to the glycolytic, pentose-phosphate, and glutaminolytic pathways [26]. In addition the various T-cell

Declaration of interest

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

Contribution of authors

The project and the experimental design were conceptualized by T.E. after significant discussions with I.S. Almost all the experiments were conducted by T.E. and G.P. Flow cytometry analysis was performed by T.E., G.P., E.Y. and D.M. T.E., S.A., G.A. and V.L. analyzed the data. All authors contributed towards writing the manuscript.

References (43)

  • R.J. Deberardinis et al.

    Brick by brick: metabolism and tumor cell growth

    Curr Opin Genet Dev

    (2008)
  • G. Kroemer et al.

    Tumor cell metabolism: cancer’s Achilles’ heel

    Cancer Cell

    (2008)
  • S.P. Cobbold et al.

    Infectious tolerance via the consumption of essential amino acids and mTOR signaling

    Proc Natl Acad Sci USA

    (2009)
  • S.K. Seo et al.

    4-1BB-mediated immunotherapy of rheumatoid arthritis

    Nat Med

    (2004)
  • E. Kwidzinski et al.

    Indoleamine 2,3-dioxygenase is expressed in the CNS and down-regulates autoimmune inflammation

    FASEB J

    (2005)
  • A.M. Alexander et al.

    Indoleamine 2,3-dioxygenase expression in transplanted NOD islets prolongs graft survival after adoptive transfer of diabetogenic splenocytes

    Diabetes

    (2002)
  • S.C. Beutelspacher et al.

    Function of indoleamine 2,3-dioxygenase in corneal allograft rejection and prolongation of allograft survival by over-expression

    Eur J Immunol

    (2006)
  • D.H. Munn et al.

    Indoleamine 2,3-dioxygenase and tumor-induced tolerance

    J Clin Invest

    (2007)
  • D.H. Munn et al.

    Prevention of allogeneic fetal rejection by tryptophan catabolism

    Science

    (1998)
  • A.L. Mellor et al.

    Prevention of T cell-driven complement activation and inflammation by tryptophan catabolism during pregnancy

    Nat Immunol

    (2001)
  • T. Eleftheriadis et al.

    Plasma indoleamine 2,3-dioxygenase and arginase type I may contribute to decreased blood T-cell count in hemodialysis patients

    Ren Fail

    (2012)
  • Cited by (24)

    • GC-TOF/MS-based metabolomics approach to study the cellular immunotoxicity of deoxynivalenol on murine macrophage ANA-1 cells

      2016, Chemico-Biological Interactions
      Citation Excerpt :

      Although initial observations in this area were made almost a century ago, studies over the past decade have elucidated the molecular basis for how extracellular signals control the uptake and catabolism of nutrients in quiescent and activated immune cells. Collectively, these studies have revealed that the metabolic pathways of oxidative metabolism [20], glycolysis [21], and glutaminolysis [22] preferentially fuel the cell fate decisions and effector functions of immune cells [23]. DON is rapidly metabolized for urinary excretion as glucuronide conjugates within 24 h of dietary exposure in human body [24].

    • Sugar, fat, and protein: New insights into what T cells crave

      2015, Current Opinion in Immunology
      Citation Excerpt :

      In addition, Akt may not be the only way T cells control glucose uptake. Other studies have expanded on T cell regulation of glucose transport, including GCN2, a target of the indoleamine 2,3-dioxygenase (IDO) pathway, and leptin, an adipokine that plays a central role in systemic metabolism [18,19]. These two signaling pathways can modulate Glut1 and Glut3 expression in an Akt-independent manner, suggesting that many distinct pathways may be able to modulate the glycolytic machinery.

    • Multiple Roles of the Stress Sensor GCN2 in Immune Cells

      2023, International Journal of Molecular Sciences
    View all citing articles on Scopus
    View full text