Skip to main content
Advances in Nutrition logoLink to Advances in Nutrition
. 2016 Jul 11;7(4):798S–805S. doi: 10.3945/an.115.011221

Leucine Metabolism in T Cell Activation: mTOR Signaling and Beyond1,2,3

Elitsa A Ananieva 4,*, Jonathan D Powell 5, Susan M Hutson 6
PMCID: PMC4942864  PMID: 27422517

Abstract

In connection with the increasing interest in metabolic regulation of the immune response, this review discusses current advances in understanding the role of leucine and leucine metabolism in T lymphocyte (T cell) activation. T cell activation during the development of an immune response depends on metabolic reprogramming to ensure that sufficient nutrients and energy are taken up by the highly proliferating T cells. Leucine has been described as an important essential amino acid and a nutrient signal that activates complex 1 of the mammalian target of rapamycin (mTORC1), which is a critical regulator of T cell proliferation, differentiation, and function. The role of leucine in these processes is further discussed in relation to amino acid transporters, leucine-degrading enzymes, and other metabolites of leucine metabolism. A new model of T cell regulation by leucine is proposed and outlines a chain of events that leads to the activation of mTORC1 in T cells.

Keywords: immune function, leucine, BCATc, T cells, mTORC1, KIC

Introduction

T cells play a central role in the cell-mediated immunity against pathogens, autoimmune disorders, and cancer. For the initiation of an immune response, T cells clonally expand, acquire effector functions, and reach a state of full activation known as signal 1 plus signal 2 activation (1, 2). These processes are high energy demanding and are accompanied by distinct changes in nutrient uptake and cellular metabolism (metabolic reprogramming) (3). During metabolic reprogramming, T cells enter a highly glycolytic state (4), show a marked increase in protein synthesis (4), and have an enhanced uptake of amino acids (5, 6). Not surprisingly, a deficiency in either dietary protein or amino acids has long been known to impair immune function and to increase human susceptibility to infections (7). Current advances in cellular metabolism have shown that amino acids are not simply building blocks in the polypeptide chain of proteins but are also important regulators of cellular processes including metabolism, protein translation, and cell growth and proliferation (811). The expansion of actively proliferating T cells is dependent on arginine; arginine is an important precursor of polyamines (via ornithine), creatine, agmatine, and protein synthesis (12, 13). Myeloid suppressor cells are able to suppress activated T cells by manipulating the metabolism of arginine through enzymes such as NO synthetase and arginase (13). Similarly, T cells require tryptophan during expansion, and the lack of tryptophan blocks their proliferation. Enzymes, such as indoleamine 2,3-dioxygenase (IDO)7, inhibit T cell proliferation by depleting tryptophan and play roles in autoimmunity and anti-inflammatory responses (14, 15).

Leucine is an essential amino acid that also plays a role in muscle atrophy, obesity, and metabolic disorders; liver disease; and even cancer (1620). Evidence suggests that leucine is important for the adaptive immune response in which leucine plays a role in T cell activation. This is associated with the long-known role of leucine as an activator of mammalian target of rapamycin complex 1 (mTORC). More recently, research on the mTORC in association with the immune response identified the mTOR pathway as a critical regulator of immune function (2124). This review provides an overview of leucine in health and disease and then summarizes the most current understanding of the effects that leucine and leucine metabolism have on T cell activation.

Current Status of Knowledge

Leucine is the most common proteinogenic amino acid with major metabolic roles.

Leucine, together with the other BCAAs, isoleucine and valine, comprise ∼40% of the free essential amino acids in blood plasma (25). The first catabolic step is limited in liver; and leucine is available to skeletal muscle where it functions as a nutrient signal, is used for protein synthesis, and serves as a metabolic fuel and/or a nitrogen donor for the synthesis of glutamine and alanine (26, 27). Leucine is not limited to acting as a substrate for protein synthesis. It is actually a well-described regulator of protein turnover that stimulates protein synthesis and inhibits protein degradation (11, 28, 29). Many leucine-supplementation studies linked leucine to body-weight control, whole-body energy expenditure, and/or postexercise recovery of muscle protein (2830). For example, recovery of rat muscle protein synthesis after a strenuous 2-h treadmill run was stimulated by oral leucine supplementation in combination with glucose and sucrose. The efficient restoration of muscle glycogen was accomplished by the supplementation of glucose and sucrose, whereas leucine had a pronounced stimulatory effect on muscle protein synthesis (19). Apart from leucine’s role in high-performance physical activity and postexercise muscle recovery, leucine intake was linked to reduced adiposity and the prevention of age- or diet-induced obesity (3133). Adult male Wistar rats maintained on a food-restricted and low-dose leucine supplementation showed increased body fat loss and increased liver protein concentrations (33). However, a leucine-rich (4%) diet fed to aged rats decreased body fat but did not have an effect on metabolic indicators of chronic diseases, such as total cholesterol, TGs, and glycemia (30). Nevertheless, several other studies highlighted the therapeutic potential of leucine supplementation for the prevention or treatment of diabetes and obesity (31, 32). Leucine supplementation via drinking water in mice fed a high-fat diet led to significantly reduced weight gain and improved hyperglycemia and hypercholesterolemia and prevented diet-induced obesity (31). These changes were associated with a leucine-induced increase in resting energy expenditure (31). Consistent with this study, mice deficient in the mitochondrial branched-chain aminotransferase (BCATm), which catalyzes the first step in leucine degradation (see below), remained lean after being fed a high-fat diet (32). The lean phenotype of global BCATm knockout (BCATm−/−) mice was attributed to increased protein turnover and increased energy expenditure. These mice showed elevated plasma leucine concentrations that correlated with improved insulin sensitivity and glucose tolerance with high-fat diet feeding, indicating that disruption in leucine metabolism may be a potential therapeutic target for obesity (32). Thus, it is evident that leucine is an important regulator of a variety of cellular functions with important effects on metabolic health and disease.

Leucine transport and metabolism in health and disease.

As an essential amino acid, leucine cannot be synthesized in the body but must be obtained from the diet in humans. Soon after consumption of a protein-containing meal, the concentration of leucine increases and is transported across the cell membrane by a family of l-type amino acid transporters (LATs). This family consists of 4 Na+-independent neutral amino acid transporters: LAT1–LAT4. LAT1 and LAT2 are also known as solute carrier (Slc) 7 (Slc7a5 and Slc7a8, respectively), whereas LAT3 and LAT4 are known as Slc43 (Slc43a1 and Slc43a2, respectively) (34). LAT1 and LAT2 require a binding partner and deliver a wider range of neutral amino acids compared with LAT3 and LAT4; the latter are facilitated diffusers and more specific to the leucine, isoleucine, valine, phenylalanine, and methionine (34, 35). The 4 transporters have different expression patterns and tissue localization, although they overlap to some extent (36). LAT1 is mainly associated with spleen, activated lymphocytes, and brain, and binding of leucine to LAT1 (Slc7a5) has been the most studied (6, 3739). Leucine transport is dependent on glutamine and proceeds via a 2-step transport mechanism (38). First, glutamine is transported inside the cell via the glutamine transporter (Slc1a5) that regulates glutamine intracellular concentrations. Next, a complex of Slc7a5 and Slc3a2 (another glutamine transporter) uses intracellular glutamine as an efflux substrate to regulate the uptake of extracellular leucine into the cells (38). Once inside the cell, leucine can either regulate cellular processes, be incorporated into protein, or undergo degradation starting with transfer of its amino group to α-ketoglutarate (transamination). The initiation of leucine degradation occurs primarily in the mitochondria of skeletal muscle and other tissues, because leucine as well as isoleucine and valine transamination is limited in the liver (26). Leucine transamination is catalyzed by the BCATm enzyme (40). This is a reversible transfer of the α-amino group of leucine to α-ketoglutarate to form glutamate and α-ketoisocaproate (KIC) (Figure 1). Approximately 20% of leucine is converted into KIC, whereas the rest of leucine is used for protein synthesis in skeletal muscle (42). Glutamate, on the other hand, undergoes either amidation to glutamine or transamination to α-ketoglutarate to generate alanine in multiple tissues (42, 43).

FIGURE 1.

FIGURE 1

Overview of leucine metabolism. BCATc and BCATm catalyze the first step in leucine degradation by transferring nitrogen from leucine to α-ketoglutarate to produce glutamate and KIC. After leucine is metabolized to KIC, KIC is either metabolized into isovaleryl-CoA by BCKDC or into HMB by an enzyme referred as to KIC dioxygenase (41). Alternatively, one of the derivatives of isovaleryl-CoA, MC-CoA, can be converted into HMB. Ultimately, these metabolites yield acetoacetyl-CoA and acetoacetate and are used for energy production. Apart from being metabolized, leucine stimulates protein synthesis by activating the mTORC1 signaling pathway. BCATc, cytosolic branched-chain aminotransferase; BCATm, mitochondrial branched-chain aminotransferase; BCKDC, branched-chain α-keto acid dehydrogenase complex; HMB, β-hydroxy-β-methylbutyrate; KIC, α-ketoisocaproate; MC-CoA, β-methyl-crotonyl-CoA; mTORC1, complex 1 of the mammalian target of rapamycin; S6K, p70 ribosomal S6 kinase; 4E-BP1, eukaryotic translation initiation factor 4E–binding protein 1.

BCATm is expressed in most human tissues except for liver hepatocytes (26). Likewise, BCATm is constitutively expressed in T cells (44). Another enzyme that transaminates leucine is the cytosolic branched-chain aminotransferase (BCATc). In contrast to BCATm, BCATc is expressed in the nervous system but has limited expression in other adult human tissues (26). However, many cancer types as well as activated T cells express BCATc, and BCATc is implicated as an important prognostic marker for cancer and a potential candidate for an immunosuppressive enzyme (4447).

Once BCATm converts leucine to KIC in muscle, a substantial amount of KIC is released into the bloodstream and further metabolized in the liver by the branched-chain α-keto acid dehydrogenase complex (BCKDC), a large multienzyme complex that contains multiple copies of 3 enzymes: a branched-chain α-keto acid decarboxylase (E1), a dihydrolipoyl transacylase (E2), and a dihydrolipoyl dehydrogenase (E3) (26). BCKDC activity is regulated by phosphorylation/dephosphorylation. It is inhibited by the branched-chain α-keto acid dehydrogenase kinase (BDK), which phosphorylates the E1α subunit of the E1 enzyme. This process is reversed by protein phosphatase (PPM1K), which activates BCKDC (48, 49). Leucine transamination by BCATm and oxidative decarboxylation by BCKDC regulate the supply of leucine for tissue protein synthesis and other leucine functions. They are also important for the prevention of buildup of toxic metabolites or excessive leucine concentrations. A congenital deficiency in BCKDC leading to maple syrup urine disease (MSUD) is associated with elevated plasma leucine concentrations and the presence of branched-chain α-keto acids (BCKAs) in the urine (50). One mechanism that explains leucine toxicity in MSUD is leucine interference with neurotransmitter synthesis. Leucine competes for amino acid transporters with other amino acids such as tyrosine and phenylalanine, which are precursors of neurotransmitters (51). However, toxic leucine concentrations may also contribute to disruption in the energy metabolism of the brain where leucine can inhibit pyruvate dehydrogenase and α-ketoglutarate dehydrogenase (52, 53). Apart from leucine, the leucine metabolite KIC also accumulates in patients with MSUD and can affect the brain bioenergetic homeostasis (54). The mechanism includes uncoupling of oxidative phosphorylation and inhibition of α-ketoglutarate dehydrogenase activity by KIC (54). In addition, in cancer patients with cachexia, leucine supplementation enhanced tumor progression, although it was intended to protect against cancer-induced cachexia. This is evident from a recent study that showed that leucine supplementation enhanced tumor growth in both lean and overweight mice with pancreatic cancer (18). Thus, although leucine has important functions in stimulating protein synthesis, excess leucine and KIC concentrations may have a negative impact on biological processes. Another key metabolite of leucine metabolism is β-hydroxy-β-methylbutyrate (HMB) (Figure 1). Approximately 5% of leucine is irreversibly converted to HMB, and HMB has been used as a dietary substitute of leucine with no adverse effects (42). HMB can stimulate protein synthesis (55) and attenuate protein degradation (56) in a much smaller dosage than leucine and, as such, HMB has the potential to substitute for leucine as a nutrient signal when leucine supplementation is not practical or desirable (42).

Leucine and leucine metabolism in immune impairment.

A number of studies from the 1970s to the present have shown that inadequate uptake of leucine or the other BCAAs leads to immune impairment (5760). Jose and Good (57) showed that dietary restriction of leucine caused a significant decrease in the lysis of tumor cells by lymphocytes. Likewise, decreased concentrations of BCAAs, commonly seen in patients with advanced liver cirrhosis, were associated with impairment of the function and maturation of dendritic cells (58). Oral administration of BCAAs had stimulatory effects on peripheral blood mononuclear cells (PBMCs) in these patients and led to increased IFN-γ production (58). Patients with advanced chronic hepatitis C suffered from malnutrition, and plasma BCAAs were decreased to similar concentrations seen in patients with liver cirrhosis (58, 59). Malnutrition impaired IFN-γ signaling in these patients; however, an increase in the plasma concentrations of BCAAs upregulated IFN-γ signaling and was proposed as a therapeutic approach for chronic hepatitis C (59). The mechanism of BCAA function in chronic hepatitis C was further explored in a human hemochromatotic cell line (Huh-7.5) grown in low–amino acid media and infected with hepatitis C virus followed by supplementation with BCAAs. This mechanism involved the activation of the mTORC1 signaling pathway, restoration of IFN-γ signaling, and repression of the replication of hepatitis C virus by BCAAs (59).

Immunomodulatory effects of leucine metabolites (HMB, KIC) were studied in human PBMCs and in sheep (6163). Treatment with HMB reduced TNF-α concentrations in the PBMC culture medium, modified T helper (Th) 1/Th2 cytokine production toward a Th2 profile, and impaired lymphocyte proliferation and progression through the cell cycle (61). Similarly, when KIC was tested in activated PMBCs, it was found that KIC suppressed lymphocyte DNA synthesis (62). On the contrary, KIC was shown to stimulate lymphocyte blastogenesis and antibody responses in sheep (63). Moreover, feeding lambs with KIC prevented adrenocorticotropin-induced suppression of lymphocyte function, and this effect was not achieved by feeding the lambs with leucine (63). Considering that KIC transaminates with glutamate to form leucine (64), the effect of KIC could also reflect changes in intracellular leucine or glutamate and its metabolites or a direct action of high concentrations of intracellular KIC. Elucidating the underlying mechanism or mechanisms may reveal therapeutic uses for leucine metabolites.

Molecular mechanisms of leucine function.

There is evidence that shows a role for leucine in regulating the mTOR signaling pathway (16, 17, 6568). As elegantly described by Laplante and Sabatini (69, 70), mTOR signaling integrates extracellular and intracellular signals to regulate protein translation, and cell metabolism, growth, proliferation, and survival. Thus, mTOR is activated during cellular processes that use energy and nutrients, such as tumor formation, angiogenesis, insulin resistance, adipogenesis, and T cell activation (22, 7072). The mTOR signaling pathway contains 2 multiprotein complexes, mTORC1 and mTORC2. The 2 complexes have different sensitivity to the drug rapamycin, with mTORC1 being the primary target of rapamycin. The 2 complexes also differ in their upstream regulators, downstream outputs, and protein composition, all of which are described in detail by Laplante and Sabatini (69). Here, attention is given to one of the upstream proteins of mTORC1 called Rag GTPase, because leucine is known to activate mTORC1 in a Rag GTPase–dependent manner (73, 74). Mammals have 4 Rag GTPases (A–D), which can form heterodimers. Leucine promotes the loading of Rag A and Rag B with GTP, thus enabling this heterodimer to interact with mTORC1 (via Raptor) leading to mTORC1 activation (73). The exact mechanism of the regulatory role of leucine is dependent on the enzyme leucyl–transfer RNA synthetase that catalyzes the ligation of leucine to its transfer RNA. This enzyme senses leucine cellular concentrations and activates the Rag complex (74). Rag GTPase activates mTORC1 and triggers the translocation of mTORC1 to the lysosomal surface. There, mTORC1 interacts with another protein, Ras homolog enriched in brain (Rheb), a small GTPase that activates mTORC1 (75). The interaction between mTORC1, Rag GTPases, and Rheb on the lysosomal surface is possible only if amino acids such as leucine are available, signifying the important role of the lysosome in amino acid sensing by the mTORC1 signaling pathway (70, 75). The stimulatory effects of leucine on protein synthesis during exercise, protein-energy malnutrition, and adipogenesis are associated with the leucine-dependent activation of mTORC1 as well as activation of downstream targets of mTORC1 such as the p70 ribosomal S6 kinase 1 (S6K1) and the eukaryotic translation initiation factor 4E–binding protein 1 (4E-BP1) (Figure 1) (16, 76, 77). On the other hand, withdrawal of leucine was shown to be as effective in inhibiting mTORC1 signaling as was withdrawal of all amino acids (65). These findings strongly suggest a central role for leucine in regulating the mTORC1 signaling pathway in a variety of cellular processes.

Leucine is indispensable for mTORC1 regulation of T cell activation.

The mTOR signaling pathway is a vital link between the development of immune response, the surrounding environment, and cellular metabolism (22). In T cells, a primary role of mTOR signaling pathway is to sense and integrate environmental cues that dictate the fate of naive T cells upon T cell receptor engagement and is essential for Th1 and Th17 differentiation (78). Thus, it is not surprising that knocking out mTORC1 in T cells affects their lineage commitment (21, 22, 24). Delgoffe et al. (21) showed that the absence of mTORC1 impaired the ability of T cells to differentiate into Th1 or Th17 cells. Similarly, T cells lacking the mTORC1 activator Rheb failed to differentiate to Th1 and Th17 cells (79). On the other hand, deletion of tuberous sclerosis complex 1 (TSC1), an upstream inhibitor of mTORC1, caused multiorgan inflammation in mice not expressing TSC1 as a consequence of hyperactivation of T cells by greater activity of mTORC1 and elevated Th1 and Th17 responses (80). Interestingly, a deficiency in the amino acid transporters Slc7a5 (LAT1) and Slc1a5 (ASCT2) in mice also impaired the differentiation of Th1 and Th17 cells in an mTORC1-dependent manner (5, 39). As discussed above, Slc1a5 is a glutamine transporter that controls glutamine uptake and glutamine intracellular concentrations (38). Although glutamine has been studied extensively in T cells and immunity (8186), the role of leucine in T cell activation is only now emerging. Leucine availability was shown to be essential for T cell activation and proliferation (87, 88). In Jurkat T cells and activated primary mouse T cells, the leucine structural antagonist N-acetyl–leucine amide (NALA) exerted similar effects on cell cycle progression, cell proliferation, cytokine production, and downstream targets of mTORC1 such as rapamycin, suggesting the restriction of leucine availability (87, 88). Rapamycin inhibits mTORC1 and renders T cells hyporesponsive (anergic) even when they are given full signal 1 and signal 2 activation (87). The stimulation of T cells in the presence of rapamycin makes them tolerant, such that they fail to produce substantial IL-2 or IFN-γ upon rechallenge (even in the absence of rapamycin) (89). By promoting tolerance, rapamycin as well as other inhibitors of mTOR have become attractive agents for preventing transplant rejection (78, 90, 91). Other means to inhibit mTOR signaling in T cells and reduce graft rejection is by limiting essential amino acids, including leucine. Skin grafts in T cell–deficient mice were shown to express transcripts of 4 amino acid–consuming enzymes (BCATc being one of them) (92). Transplanted tissues are enriched in regulatory T cells (Tregs), which are known to maintain peripheral tolerance to both self- and nonself-antigens (78, 93). Cobbold et al. (92) postulated that tolerant Tregs could establish “infectious tolerance” by inducing amino acid–consuming enzymes. This resulted in localized depletion of essential amino acids including leucine depletion and promoted the induction of anergy and more Tregs (92). Our preliminary analysis of CD4+CD25+ Tregs infiltrating prostate cancer revealed that BCATc is markedly upregulated in these cells along with Foxp3. These data support a potential role of BCATc in tumor-induced Tregs (J Powell, unpublished data, 2010).

During activation, the T cell receptor (TCR) engagement, coupled with CD28 signaling, upregulates the mTOR signaling pathway, which, in turn, stimulates glycolysis for energy and metabolites necessary for the increased biosynthetic demands of the proliferating T cells (94). This metabolic reprogramming increases the nutrient demands of T cells and leads to an increased expression of glucose and amino acid transporters (39). Sinclair et al. (39) found that the leucine transporter Slc7a5 was induced by the TCR signaling in a nuclear factor of activated T cells (NFAT)–dependent manner. Cyclosporine A, an immunosuppressive drug that inhibits this pathway, suppressed the expression of Slc7a5 in TCR-activated T cells. Slc7a5 mediated the intracellular transport of leucine, which was essential for the activity of mTORC1 during T cell activation. The role of Slc7a5 in intracellular leucine uptake during T cell activation was further confirmed in human T cells (6). Thus, leucine transport was established as an important factor controlling mTORC1 activity in T cells.

The significance of Slc7a5 involvement in T cell activation was further shown in Slc7a5 null T cells, which failed to properly differentiate into CD4+ and CD8+ T cells (39). However, the severe phenotype of Slc7a5 null T cells was not solely due to the loss of mTORC1 activity caused by failed leucine uptake but also reflected the inability of these cells to express c-myc (39). The transcriptional factor c-MYC was identified as a critical regulator of T cell activation–induced metabolic reprogramming associated with global changes in genes important for glucose catabolism, glutaminolysis, ornithine and polyamine biosynthesis, and glucose and amino acid transport (GLUT1 and SLC7A5 among them) (82). The c-MYC–mediated induction of glycolysis and glutaminolysis in activated T cells is reminiscent of the same processes in cancer cells (95). Although a direct connection between c-MYC, leucine metabolism, and T cells has not been found, BCATc has been described as one of the c-MYC target genes in cancer cells (47, 95, 96).

To better understand the mechanism of c-MYC leucine regulatory role during T cell activation, our group explored the impact of leucine metabolic enzymes in this process. We found that BCATc, which is not normally expressed in naive resting T cells, was induced by the TCR in a manner similar to Slc7a5 (39, 44). TCR alone was sufficient to trigger BCATc expression, whereas the mitochondrial enzyme, BCATm, was expressed constitutively. Furthermore, activated T cells from global BCATc−/− mice showed increased phosphorylation of mTORC1 downstream targets ribosomal protein S6 and 4E-BP1. These cells had higher intracellular leucine concentrations and also showed higher rates of glycolysis, glycolytic capacity, and glycolytic reserve when compared with activated wild-type cells (44). Our results, along with the role of Slc7a5 in T cell activation, are consistent with a model in which TCR triggers the expression of Slc7a5 and BCATc to regulate the uptake and the cytosolic concentrations of leucine, respectively (Figure 2). In this model, BCATc is a part of a negative feedback loop controlling the input of leucine toward the mTORC1 signaling pathway (Figure 2A). Excess leucine is transaminated to KIC, which exits the cells (44). Loss of BCATc expression eliminates cytosolic leucine catabolism and, as a result, the availability of leucine to activate mTORC1 is not limited, potentially resulting in T cell hyperactivation (Figure 2B). Future studies in animal models of human diseases will be important to verify this paradigm in vivo. Nevertheless, these studies show that leucine uptake and leucine metabolism in T cells are critical for the regulation of the mTORC1 signaling pathway during T cell activation, and manipulating the immune response by targeting leucine could prove useful in treating infections, autoimmunity, and/or cancer.

FIGURE 2.

FIGURE 2

Model of Slc7a5 and BCATc regulation of mTORC1 activity in T cells. (A) Slc7a5 expression is induced by TCR in T cells leading to increased leucine uptake. Leucine activates mTORC1, which stimulates protein translation and glycolysis that ultimately results in T cell activation. BCATc is also induced by TCR and can initiate cytosolic transamination of leucine, providing a negative feedback regulation of mTORC1 activity. The BCKA product of leucine transamination (KIC) is transported outside the cells. (B) When BCATc is lost, the supply of leucine to mTORC1 is increased, which could result in T cell hyperactivation. BCATc, cytosolic branched-chain aminotransferase; BCKA, branched-chain α-ketoacid; KIC, α-ketoisocaproate; mTORC1, complex 1 of the mammalian target of rapamycin; TCR, T cell receptor; Slc7a5, solute carrier family 7a5 [l-type amino acid transporter (LAT1)]; α-KG, α-ketoglutarate.

Conclusions

Substantial progress has been made in our understanding of leucine and its role as a nutrient signal activating the mTORC1 pathway in processes as diverse as muscle function, insulin resistance, and the activation of the immune response. Moreover, new and exciting studies have connected leucine metabolism via BCATc with cancer progression; thus, BCATc has emerged as a new prognostic marker for cancer (45, 97). Amino acid metabolic enzymes in tryptophan and arginine metabolism, such as IDO, argininosuccinate synthase 1 (ASS1), and arginase, have been well established in cancer and cancer immunotherapy and are subjects of preclinical and clinical trials (98, 99). Further studies targeting BCATc and leucine metabolism in cancer and immune disorders will expand our knowledge and opportunities for development of novel therapeutic approaches.

Acknowledgments

We thank Adele Addington for critical evaluation of the manuscript. All authors read and approved the final manuscript.

Footnotes

7

Abbreviations used: ASS1, argininosuccinate synthase 1; BCATc, cytosolic branched-chain aminotransferase; BCATm, mitochondrial branched-chain aminotransferase; BCKA, branched-chain α-keto acid; BCKDC, branched-chain α-keto acid dehydrogenase complex; BDK, branched-chain α-keto acid dehydrogenase kinase; HMB, β-hydroxy-β-methylbutyrate; IDO, indoleamine 2,3-dioxygenase; KIC, α-ketoisocaproate; LAT, l-type amino acid transporter; MSUD, maple syrup urine disease; mTOR, mammalian target of rapamycin; mTORC, mammalian target of rapamycin complex; NALA, N-acetyl–leucine amide; NFAT, nuclear factor of activated T cells; PBMC, peripheral blood mononuclear cell; PPM1K, protein phosphatase, Mg2+/Mn2+ dependent 1K; Rheb, Ras homolog enriched in brain; S6K1, p70 ribosomal S6 kinase 1; Slc, solute carrier; TCR, T cell receptor; Th, T helper; Treg, regulatory T cell; TSC1, tuberous sclerosis complex 1; 4E-BP1, eukaryotic translation initiation factor 4E-binding protein 1.

References

  • 1.Schwartz RH. Costimulation of T lymphocytes: the role of CD28, CTLA-4, and B7/BB1 in interleukin-2 production and immunotherapy. Cell 1992;71:1065–8. [DOI] [PubMed] [Google Scholar]
  • 2.Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol 2009;27:591–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Buck MD, O’Sullivan D, Pearce EL. T cell metabolism drives immunity. J Exp Med 2015;212:1345–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Jones RG, Thompson CB. Revving the engine: signal transduction fuels T cell activation. Immunity 2007;27:173–8. [DOI] [PubMed] [Google Scholar]
  • 5.Nakaya M, Xiao Y, Zhou X, Chang JH, Chang M, Cheng X, Blonska M, Lin X, Sun SC. Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity 2014;40:692–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hayashi K, Jutabha P, Endou H, Sagara H, Anzai N. LAT1 is a critical transporter of essential amino acids for immune reactions in activated human T cells. J Immunol 2013;191:4080–5. [DOI] [PubMed] [Google Scholar]
  • 7.Li P, Yin YL, Li D, Kim SW, Wu G. Amino acids and immune function. Br J Nutr 2007;98:237–52. [DOI] [PubMed] [Google Scholar]
  • 8.Barbul A. Arginine: biochemistry, physiology, and therapeutic implications. JPEN J Parenter Enteral Nutr 1986;10:227–38. [DOI] [PubMed] [Google Scholar]
  • 9.Brand K. Glutamine and glucose metabolism during thymocyte proliferation. pathways of glutamine and glutamate metabolism. Biochem J 1985;228:353–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Grohmann U, Bronte V. Control of immune response by amino acid metabolism. Immunol Rev 2010;236:243–64. [DOI] [PubMed] [Google Scholar]
  • 11.Garlick PJ. The role of leucine in the regulation of protein metabolism. J Nutr 2005;135(6, Suppl):1553S–6S. [DOI] [PubMed] [Google Scholar]
  • 12.Wu G, Morris SM Jr. Arginine metabolism: nitric oxide and beyond. Biochem J 1998;336(Pt 1):1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P. L-arginine metabolism in myeloid cells controls T-lymphocyte functions. Trends Immunol 2003;24:302–6. [DOI] [PubMed] [Google Scholar]
  • 14.Mellor AL, Munn DH. Tryptophan catabolism and T-cell tolerance: immunosuppression by starvation? Immunol Today 1999;20:469–73. [DOI] [PubMed] [Google Scholar]
  • 15.Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol 2004;4:762–74. [DOI] [PubMed] [Google Scholar]
  • 16.Columbus DA, Steinhoff-Wagner J, Suryawan A, Nguyen HV, Hernandez-Garcia A, Fiorotto ML, Davis TA. Impact of prolonged leucine supplementation on protein synthesis and lean growth in neonatal pigs. Am J Physiol Endocrinol Metab 2015;309:E601–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gao X, Tian F, Wang X, Zhao J, Wan X, Zhang L, Wu C, Li N, Li J. Leucine supplementation improves acquired growth hormone resistance in rats with protein-energy malnutrition. PLoS One 2015;10:e0125023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Liu KA, Lashinger LM, Rasmussen AJ, Hursting SD. Leucine supplementation differentially enhances pancreatic cancer growth in lean and overweight mice. Cancer Metab 2014;2(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Anthony JC, Anthony TG, Layman DK. Leucine supplementation enhances skeletal muscle recovery in rats following exercise. J Nutr 1999;129:1102–6. [DOI] [PubMed] [Google Scholar]
  • 20.Churchward-Venne TA, Breen L, Di Donato DM, Hector AJ, Mitchell CJ, Moore DR, Stellingwerff T, Breuille D, Offord EA, Baker SK, et al. Leucine supplementation of a low-protein mixed macronutrient beverage enhances myofibrillar protein synthesis in young men: a double-blind, randomized trial. Am J Clin Nutr 2014;99:276–86. [DOI] [PubMed] [Google Scholar]
  • 21.Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B, Worley PF, Kozma SC, Powell JD. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 2009;30:832–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Powell JD, Pollizzi KN, Heikamp EB, Horton MR. Regulation of immune responses by mTOR. Annu Rev Immunol 2012;30:39–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liu C, Chapman NM, Karmaus PW, Zeng H, Chi H. mTOR and metabolic regulation of conventional and regulatory T cells. J Leukoc Biol 2015 Feb 24 (Epub ahead of print; pii: jlb.2RI0814-408R). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Delgoffe GM, Powell JD. mTOR: taking cues from the immune microenvironment. Immunology 2009;127:459–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hutson SM, Sweatt AJ, Lanoue KF. Branched-chain [corrected] amino acid metabolism: implications for establishing safe intakes. J Nutr 2005;135(6, Suppl):1557S–64S. Erratum in J Nutr. 2005;135:2009. [DOI] [PubMed] [Google Scholar]
  • 26.Sweatt AJ, Wood M, Suryawan A, Wallin R, Willingham MC, Hutson SM. Branched-chain amino acid catabolism: unique segregation of pathway enzymes in organ systems and peripheral nerves. Am J Physiol Endocrinol Metab 2004;286:E64–76. [DOI] [PubMed] [Google Scholar]
  • 27.Garber AJ, Karl IE, Kipnis DM. Alanine and glutamine synthesis and release from skeletal muscle. II. The precursor role of amino acids in alanine and glutamine synthesis. J Biol Chem 1976;251:836–43. [PubMed] [Google Scholar]
  • 28.Norton LE, Layman DK. Leucine regulates translation initiation of protein synthesis in skeletal muscle after exercise. J Nutr 2006;136(Suppl):533S–7S. [DOI] [PubMed] [Google Scholar]
  • 29.Sugawara T, Ito Y, Nishizawa N, Nagasawa T. Regulation of muscle protein degradation, not synthesis, by dietary leucine in rats fed a protein-deficient diet. Amino Acids 2009;37:609–16. [DOI] [PubMed] [Google Scholar]
  • 30.Vianna D, Resende GF, Torres-Leal FL, Pantaleao LC, Donato J Jr, Tirapegui J. Long-term leucine supplementation reduces fat mass gain without changing body protein status of aging rats. Nutrition 2012;28:182–9. [DOI] [PubMed] [Google Scholar]
  • 31.Zhang Y, Guo K, LeBlanc RE, Loh D, Schwartz GJ, Yu YH. Increasing dietary leucine intake reduces diet-induced obesity and improves glucose and cholesterol metabolism in mice via multimechanisms. Diabetes 2007;56:1647–54. [DOI] [PubMed] [Google Scholar]
  • 32.She P, Reid TM, Bronson SK, Vary TC, Hajnal A, Lynch CJ, Hutson SM. Disruption of BCATm in mice leads to increased energy expenditure associated with the activation of a futile protein turnover cycle. Cell Metab 2007;6:181–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Donato J Jr, Pedrosa RG, Cruzat VF, Pires IS, Tirapegui J. Effects of leucine supplementation on the body composition and protein status of rats submitted to food restriction. Nutrition 2006;22:520–7. [DOI] [PubMed] [Google Scholar]
  • 34.Wang Q, Holst J. L-type amino acid transport and cancer: targeting the mTORC1 pathway to inhibit neoplasia. Am J Cancer Res 2015;5:1281–94. [PMC free article] [PubMed] [Google Scholar]
  • 35.Kanai Y, Segawa H, Miyamoto K, Uchino H, Takeda E, Endou H. Expression cloning and characterization of a transporter for large neutral amino acids activated by the heavy chain of 4F2 antigen (CD98). J Biol Chem 1998;273:23629–32. [DOI] [PubMed] [Google Scholar]
  • 36.Segawa H, Fukasawa Y, Miyamoto K, Takeda E, Endou H, Kanai Y. Identification and functional characterization of a Na+-independent neutral amino acid transporter with broad substrate selectivity. J Biol Chem 1999;274:19745–51. [DOI] [PubMed] [Google Scholar]
  • 37.Mastroberardino L, Spindler B, Pfeiffer R, Skelly PJ, Loffing J, Shoemaker CB, Verrey F. Amino-acid transport by heterodimers of 4F2hc/CD98 and members of a permease family. Nature 1998;395:288–91. [DOI] [PubMed] [Google Scholar]
  • 38.Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, Yang H, Hild M, Kung C, Wilson C, et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 2009;136:521–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sinclair LV, Rolf J, Emslie E, Shi YB, Taylor PM, Cantrell DA. Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat Immunol 2013;14:500–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hutson SM, Fenstermacher D, Mahar C. Role of mitochondrial transamination in branched chain amino acid metabolism. J Biol Chem 1988;263:3618–25. [PubMed] [Google Scholar]
  • 41.Zanchi NE, Gerlinger-Romero F, Guimaraes-Ferreira L, de Siqueira Filho MA, Felitti V, Lira FS, Seelaender M, Lancha AH Jr. HMB supplementation: clinical and athletic performance-related effects and mechanisms of action. Amino Acids 2011;40:1015–25. [DOI] [PubMed] [Google Scholar]
  • 42.Duan Y, Li F, Li Y, Tang Y, Kong X, Feng Z, Anthony TG, Watford M, Hou Y, Wu G, et al. The role of leucine and its metabolites in protein and energy metabolism. Amino Acids 2015;48:41–51. [DOI] [PubMed] [Google Scholar]
  • 43.Wu G, Thompson JR, Sedgwick GW, Drury M. Formation of alanine and glutamine in chick (Gallus domesticus) skeletal muscle. Comp Biochem Physiol B 1989;93:609–13. [DOI] [PubMed] [Google Scholar]
  • 44.Ananieva EA, Patel CH, Drake CH, Powell JD, Hutson SM. Cytosolic branched chain aminotransferase (BCATc) regulates mTORC1 signaling and glycolytic metabolism in CD4+ T cells. J Biol Chem 2014;289:18793–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tönjes M, Barbus S, Park YJ, Wang W, Schlotter M, Lindroth AM, Pleier SV, Bai AH, Karra D, Piro RM, et al. BCAT1 promotes cell proliferation through amino acid catabolism in gliomas carrying wild-type IDH1. Nat Med 2013;19:901–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yoshikawa R, Yanagi H, Shen CS, Fujiwara Y, Noda M, Yagyu T, Gega M, Oshima T, Yamamura T, Okamura H, et al. ECA39 is a novel distant metastasis-related biomarker in colorectal cancer. World J Gastroenterol 2006;12:5884–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhou W, Feng X, Ren C, Jiang X, Liu W, Huang W, Liu Z, Li Z, Zeng L, Wang L, et al. Over-expression of BCAT1, a c-myc target gene, induces cell proliferation, migration and invasion in nasopharyngeal carcinoma. Mol Cancer 2013;12:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lynch CJ, Adams SH. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat Rev Endocrinol 2014;10:723–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Harris RA, Popov KM, Zhao Y, Kedishvili NY, Shimomura Y, Crabb DW. A new family of protein kinases—the mitochondrial protein kinases. Adv Enzyme Regul 1995;35:147–62. [DOI] [PubMed] [Google Scholar]
  • 50.Burrage LC, Nagamani SC, Campeau PM, Lee BH. Branched-chain amino acid metabolism: from rare Mendelian diseases to more common disorders. Hum Mol Genet 2014;23 R1:R1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Oldendorf WH, Szabo J. Amino acid assignment to one of three blood-brain barrier amino acid carriers. Am J Physiol 1976;230:94–8. [DOI] [PubMed] [Google Scholar]
  • 52.Patel MS, Auerbach VH, Grover WD, Wilbur DO. Effect of the branched-chain alpha-keto acids on pyruvate metabolism by homogenates of human brain. J Neurochem 1973;20:1793–6. [DOI] [PubMed] [Google Scholar]
  • 53.Patel MS. Inhibition by the branched-chain 2-oxo acids of the 2-oxoglutarate dehydrogenase complex in developing rat and human brain. Biochem J 1974;144:91–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Amaral AU, Leipnitz G, Fernandes CG, Seminotti B, Schuck PF, Wajner M. Alpha-ketoisocaproic acid and leucine provoke mitochondrial bioenergetic dysfunction in rat brain. Brain Res 2010;1324:75–84. [DOI] [PubMed] [Google Scholar]
  • 55.Wheatley SM, El-Kadi SW, Suryawan A, Boutry C, Orellana RA, Nguyen HV, Davis SR, Davis TA. Protein synthesis in skeletal muscle of neonatal pigs is enhanced by administration of beta-hydroxy-beta-methylbutyrate. Am J Physiol Endocrinol Metab 2014;306:E91–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Noh KK, Chung KW, Choi YJ, Park MH, Jang EJ, Park CH, Yoon C, Kim ND, Kim MK, Chung HY. Beta-hydroxy beta-methylbutyrate improves dexamethasone-induced muscle atrophy by modulating the muscle degradation pathway in SD rat. PLoS One 2014;9:e102947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jose DG, Good RA. Quantitative effects of nutritional protein and calorie deficiency upon immune responses to tumors in mice. Cancer Res 1973;33:807–12. [PubMed] [Google Scholar]
  • 58.Kakazu E, Ueno Y, Kondo Y, Fukushima K, Shiina M, Inoue J, Tamai K, Ninomiya M, Shimosegawa T. Branched chain amino acids enhance the maturation and function of myeloid dendritic cells ex vivo in patients with advanced cirrhosis. Hepatology 2009;50:1936–45. [DOI] [PubMed] [Google Scholar]
  • 59.Honda M, Takehana K, Sakai A, Tagata Y, Shirasaki T, Nishitani S, Muramatsu T, Yamashita T, Nakamoto Y, Mizukoshi E, et al. Malnutrition impairs interferon signaling through mTOR and FoxO pathways in patients with chronic hepatitis C. Gastroenterology 2011;141:128,40, 140.e1–2. [DOI] [PubMed] [Google Scholar]
  • 60.Kobayashi M, Ikeda K, Arase Y, Suzuki Y, Suzuki F, Akuta N, Hosaka T, Murashima N, Saitoh S, Someya T, et al. Inhibitory effect of branched-chain amino acid granules on progression of compensated liver cirrhosis due to hepatitis C virus. J Gastroenterol 2008;43:63–70. [DOI] [PubMed] [Google Scholar]
  • 61.Nunes EA, Lomax AR, Noakes PS, Miles EA, Fernandes LC, Calder PC. Beta-hydroxy-beta-methylbutyrate modifies human peripheral blood mononuclear cell proliferation and cytokine production in vitro. Nutrition 2011;27:92–9. [DOI] [PubMed] [Google Scholar]
  • 62.Santos KD, Rocha M, Wannmacher CM, Wajner M. The influence of organic acids on the proliferation of human peripheral lymphocytes activated by concanavalin A and pokeweed mitogen. Int J Immunopharmacol 1996;18:761–9. [DOI] [PubMed] [Google Scholar]
  • 63.Kuhlman G, Roth JA, Nissen S. Effects of alpha-ketoisocaproate on adrenocorticotropin-induced suppression of lymphocyte function in sheep. Am J Vet Res 1991;52:388–92. [PubMed] [Google Scholar]
  • 64.Hutson SM, Zapalowski C, Cree TC, Harper AE. Regulation of leucine and alpha-ketoisocaproic acid metabolism in skeletal muscle. effects of starvation and insulin. J Biol Chem 1980;255:2418–26. [PubMed] [Google Scholar]
  • 65.Hara K, Yonezawa K, Weng QP, Kozlowski MT, Belham C, Avruch J. Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF-4E BP1 through a common effector mechanism. J Biol Chem 1998;273:14484–94. [DOI] [PubMed] [Google Scholar]
  • 66.Melnik BC. Leucine signaling in the pathogenesis of type 2 diabetes and obesity. World J Diabetes 2012;3:38–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pedroso JA, Zampieri TT, Donato J Jr. Reviewing the effects of L-leucine supplementation in the regulation of food intake, energy balance, and glucose homeostasis. Nutrients 2015;7:3914–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gil JH, Kim CK. Effects of different doses of leucine ingestion following eight weeks of resistance exercise on protein synthesis and hypertrophy of skeletal muscle in rats. J Exerc Nutrition Biochem 2015;19:31–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Laplante M, Sabatini DM. mTOR signaling at a glance. J Cell Sci 2009;122:3589–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 2012;149:274–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Waickman AT, Powell JD. mTOR, metabolism, and the regulation of T-cell differentiation and function. Immunol Rev 2012;249:43–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Waickman AT, Powell JD. Mammalian target of rapamycin integrates diverse inputs to guide the outcome of antigen recognition in T cells. J Immunol 2012;188:4721–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, Sabatini DM. The rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 2008;320:1496–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Han JM, Jeong SJ, Park MC, Kim G, Kwon NH, Kim HK, Ha SH, Ryu SH, Kim S. Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell 2012;149:410–24. [DOI] [PubMed] [Google Scholar]
  • 75.Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini DM. Ragulator-rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 2010;141:290–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gao HN, Hu H, Zheng N, Wang JQ. Leucine and histidine independently regulate milk protein synthesis in bovine mammary epithelial cells via mTOR signaling pathway. J Zhejiang Univ Sci B 2015;16:560–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lynch CJ, Fox HL, Vary TC, Jefferson LS, Kimball SR. Regulation of amino acid-sensitive TOR signaling by leucine analogues in adipocytes. J Cell Biochem 2000;77:234–51. [DOI] [PubMed] [Google Scholar]
  • 78.Lo YC, Lee CF, Powell JD. Insight into the role of mTOR and metabolism in T cells reveals new potential approaches to preventing graft rejection. Curr Opin Organ Transplant 2014;19:363–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell JD. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol 2011;12:295–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Park Y, Jin HS, Lopez J, Elly C, Kim G, Murai M, Kronenberg M, Liu YC. TSC1 regulates the balance between effector and regulatory T cells. J Clin Invest 2013;123:5165–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Klysz D, Tai X, Robert PA, Craveiro M, Cretenet G, Oburoglu L, Mongellaz C, Floess S, Fritz V, Matias MI, 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. [DOI] [PubMed] [Google Scholar]
  • 82.Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, McCormick LL, Fitzgerald P, Chi H, Munger J, Green DR. The transcription factor myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 2011;35:871–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Blagih J, Coulombe F, Vincent EE, Dupuy F, Galicia-Vazquez G, Yurchenko E, Raissi TC, van der Windt GJ, Viollet B, Pearce EL, et al. The energy sensor AMPK regulates T cell metabolic adaptation and effector responses in vivo. Immunity 2015;42:41–54. [DOI] [PubMed] [Google Scholar]
  • 84.Sikalidis AK. Amino acids and immune response: a role for cysteine, glutamine, phenylalanine, tryptophan and arginine in T-cell function and cancer? Pathol Oncol Res 2015;21:9–17. [DOI] [PubMed] [Google Scholar]
  • 85.Hsiung YC, Liu JJ, Hou YC, Yeh CL, Yeh SL. Effects of dietary glutamine on the homeostasis of CD4+ T cells in mice with dextran sulfate sodium-induced acute colitis. PLoS One 2014;9:e84410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hou YC, Wu JM, Wang MY, Wu MH, Chen KY, Yeh SL, Lin MT. Glutamine supplementation attenuates expressions of adhesion molecules and chemokine receptors on T cells in a murine model of acute colitis. Mediators Inflamm 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Zheng Y, Delgoffe GM, Meyer CF, Chan W, Powell JD. Anergic T cells are metabolically anergic. J Immunol 2009;183:6095–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hidayat S, Yoshino K, Tokunaga C, Hara K, Matsuo M, Yonezawa K. Inhibition of amino acid-mTOR signaling by a leucine derivative induces G1 arrest in Jurkat cells. Biochem Biophys Res Commun 2003;301:417–23. [DOI] [PubMed] [Google Scholar]
  • 89.Powell JD, Lerner CG, Schwartz RH. Inhibition of cell cycle progression by rapamycin induces T cell clonal anergy even in the presence of costimulation. J Immunol 1999;162:2775–84. [PubMed] [Google Scholar]
  • 90.Gao W, Lu Y, El Essawy B, Oukka M, Kuchroo VK, Strom TB. Contrasting effects of cyclosporine and rapamycin in de novo generation of alloantigen-specific regulatory T cells. Am J Transplant 2007;7:1722–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Hsieh MM, Kang EM, Fitzhugh CD, Link MB, Bolan CD, Kurlander R, Childs RW, Rodgers GP, Powell JD, Tisdale JF. Allogeneic hematopoietic stem-cell transplantation for sickle cell disease. N Engl J Med 2009;361:2309–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cobbold SP, Adams E, Farquhar CA, Nolan KF, Howie D, Lui KO, Fairchild PJ, Mellor AL, Ron D, Waldmann H. Infectious tolerance via the consumption of essential amino acids and mTOR signaling. Proc Natl Acad Sci USA 2009;106:12055–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012;30:531–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR, Elstrom RL, June CH, Thompson CB. The CD28 signaling pathway regulates glucose metabolism. Immunity 2002;16:769–77. [DOI] [PubMed] [Google Scholar]
  • 95.Yeung SJ, Pan J, Lee MH. Roles of p53, MYC and HIF-1 in regulating glycolysis— the seventh hallmark of cancer. Cell Mol Life Sci 2008;65:3981–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Wang ZQ, Faddaoui A, Bachvarova M, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Guillemette C, Gobeil S, Macdonald E, et al. BCAT1 expression associates with ovarian cancer progression: possible implications in altered disease metabolism. Oncotarget 2015;6:31522–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chang IW, Wu WJ, Wang YH, Wu TF, Liang PI, He HL, Yeh BW, Li CF. BCAT1 overexpression is an indicator of poor prognosis in patients with urothelial carcinomas of the upper urinary tract and urinary bladder. Histopathology 2015 Jul 14(Epub ahead of print; DOI: 10.1111/his.12778). [DOI] [PubMed] [Google Scholar]
  • 98.Phillips MM, Sheaff MT, Szlosarek PW. Targeting arginine-dependent cancers with arginine-degrading enzymes: opportunities and challenges. Cancer Res Treat 2013;45:251–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Platten M, von Knebel Doeberitz N, Oezen I, Wick W, Ochs K. Cancer immunotherapy by targeting IDO1/TDO and their downstream effectors. Front Immunol 2014;5:673. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Advances in Nutrition are provided here courtesy of American Society for Nutrition

RESOURCES