Lucine Effect On Immunity

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

SUPPLEMENT

Leucine Metabolism in T Cell Activation: mTOR


Signaling and Beyond1–3
Elitsa A Ananieva,4* Jonathan D Powell,5 and Susan M Hutson6
4
Department of Biochemistry and Nutrition, Des Moines University, Des Moines, IA; 5Department of Oncology, Johns Hopkins University School of
Medicine, Baltimore, MD; and 6Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA

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. Adv Nutr 2016;7(Suppl):798S–805S.

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

Introduction reprogramming, T cells enter a highly glycolytic state (4),


T cells play a central role in the cell-mediated immunity show a marked increase in protein synthesis (4), and have
against pathogens, autoimmune disorders, and cancer. For an enhanced uptake of amino acids (5, 6). Not surprisingly,
the initiation of an immune response, T cells clonally ex- a deficiency in either dietary protein or amino acids has long
pand, acquire effector functions, and reach a state of full been known to impair immune function and to increase hu-
activation known as signal 1 plus signal 2 activation (1, 2). man susceptibility to infections (7). Current advances in cel-
These processes are high energy demanding and are accom- lular metabolism have shown that amino acids are not simply
panied by distinct changes in nutrient uptake and cellular me- building blocks in the polypeptide chain of proteins but are
tabolism (metabolic reprogramming) (3). During metabolic also important regulators of cellular processes including
metabolism, protein translation, and cell growth and prolif-
1
Published in a supplement to Advances in Nutrition. Some of the reviews in this supplement eration (8–11). The expansion of actively proliferating
were presented at the symposium "Translational and Transformational Concepts in Amino T cells is dependent on arginine; arginine is an important
Acid Sensing” held 29 March 2015 at the ASN Scientific Sessions and Annual Meeting at precursor of polyamines (via ornithine), creatine, agmatine,
Experimental Biology 2015 in Boston, MA. The symposium was sponsored by the American
Society for Nutrition (ASN), the ASN Energy and Macronutrient Metabolism Research and protein synthesis (12, 13). Myeloid suppressor cells are
Interest Section (RIS), and the Nutrient-Gene Interaction RIS and was supported by able to suppress activated T cells by manipulating the metabo-
Ajinimoto, Co., Inc. Other articles in this supplement are selected reviews by grant-funded
researchers from the Ajinimoto Acid Research Program (3ARP). The Supplement
lism of arginine through enzymes such as NO synthetase and
Coordinators for this supplement were Susan M Hutson and Tracy G Anthony. Supplement arginase (13). Similarly, T cells require tryptophan during expan-
Coordinator disclosures: Susan M Hutson received travel and registration expenses for the sion, and the lack of tryptophan blocks their proliferation. En-
ASN Scientific Sessions and Annual Meeting at Experimental Biology 2015. Tracy G Anthony
received travel and registration expenses for the ASN Scientific Sessions and Annual
zymes, such as indoleamine 2,3-dioxygenase (IDO)7, inhibit
Meeting at Experimental Biology 2015. Publication costs for this supplement were defrayed T cell proliferation by depleting tryptophan and play roles
in part by the payment of page charges. This publication must therefore be hereby marked in autoimmunity and anti-inflammatory responses (14, 15).
“advertisement” in accordance with 18 USC section 1734 solely to indicate this fact. The
opinions expressed in this publication are those of the author(s) and are not attributable to
Leucine is an essential amino acid that also plays a role in
the sponsors or the publisher, Editor, or Editorial Board of Advances in Nutrition. muscle atrophy, obesity, and metabolic disorders; liver disease;
2
Supported by NIH grants DK34738 (SMH) and R01CA098109 (JDP). and even cancer (16–20). Evidence suggests that leucine is
3
Author disclosures: EA Ananieva, JD Powell, and SM Hutson, no conflicts of interest.
*To whom correspondence should be addressed. E-mail: elitsa.ananieva-stoyanova@dmu. important for the adaptive immune response in which leu-
edu. cine plays a role in T cell activation. This is associated with the

798S ã2016 American Society for Nutrition. Adv Nutr 2016;7(Suppl):798S–805S; doi:10.3945/an.115.011221.
long-known role of leucine as an activator of mammalian tar- induced obesity (31). These changes were associated with a
get of rapamycin complex 1 (mTORC). More recently, research leucine-induced increase in resting energy expenditure
on the mTORC in association with the immune response iden- (31). Consistent with this study, mice deficient in the mito-
tified the mTOR pathway as a critical regulator of immune chondrial branched-chain aminotransferase (BCATm), which
function (21–24). This review provides an overview of leucine catalyzes the first step in leucine degradation (see below), re-
in health and disease and then summarizes the most current mained lean after being fed a high-fat diet (32). The lean phe-
understanding of the effects that leucine and leucine metab- notype of global BCATm knockout (BCATm2/2) mice was
olism have on T cell activation. attributed to increased protein turnover and increased energy
expenditure. These mice showed elevated plasma leucine con-
centrations that correlated with improved insulin sensitivity
Current Status of Knowledge and glucose tolerance with high-fat diet feeding, indicating
Leucine is the most common proteinogenic amino acid that disruption in leucine metabolism may be a potential ther-
with major metabolic roles. Leucine, together with the other apeutic target for obesity (32). Thus, it is evident that leucine is
BCAAs, isoleucine and valine, comprise ;40% of the free es-
an important regulator of a variety of cellular functions with
sential amino acids in blood plasma (25). The first catabolic
important effects on metabolic health and disease.
step is limited in liver; and leucine is available to skeletal muscle
where it functions as a nutrient signal, is used for protein syn-
Leucine transport and metabolism in health and disease.
thesis, and serves as a metabolic fuel and/or a nitrogen donor
As an essential amino acid, leucine cannot be synthesized
for the synthesis of glutamine and alanine (26, 27). Leucine is
in the body but must be obtained from the diet in humans.
not limited to acting as a substrate for protein synthesis. It is
Soon after consumption of a protein-containing meal, the
actually a well-described regulator of protein turnover that
concentration of leucine increases and is transported
stimulates protein synthesis and inhibits protein degradation
across the cell membrane by a family of L-type amino acid
(11, 28, 29). Many leucine-supplementation studies linked leu-
transporters (LATs). This family consists of 4 Na+-independent
cine to body-weight control, whole-body energy expenditure,
neutral amino acid transporters: LAT1–LAT4. LAT1 and
and/or postexercise recovery of muscle protein (28–30). For
LAT2 are also known as solute carrier (Slc) 7 (Slc7a5 and
example, recovery of rat muscle protein synthesis after a
Slc7a8, respectively), whereas LAT3 and LAT4 are known
strenuous 2-h treadmill run was stimulated by oral leucine
as Slc43 (Slc43a1 and Slc43a2, respectively) (34). LAT1
supplementation in combination with glucose and sucrose.
and LAT2 require a binding partner and deliver a wider
The efficient restoration of muscle glycogen was accom-
range of neutral amino acids compared with LAT3 and
plished by the supplementation of glucose and sucrose,
LAT4; the latter are facilitated diffusers and more specific
whereas leucine had a pronounced stimulatory effect
to the leucine, isoleucine, valine, phenylalanine, and me-
on muscle protein synthesis (19). Apart from leucine’s role
thionine (34, 35). The 4 transporters have different expres-
in high-performance physical activity and postexercise mus-
sion patterns and tissue localization, although they overlap
cle recovery, leucine intake was linked to reduced adiposity
to some extent (36). LAT1 is mainly associated with spleen,
and the prevention of age- or diet-induced obesity (31–33).
activated lymphocytes, and brain, and binding of leucine
Adult male Wistar rats maintained on a food-restricted and
to LAT1 (Slc7a5) has been the most studied (6, 37–39).
low-dose leucine supplementation showed increased body
Leucine transport is dependent on glutamine and proceeds
fat loss and increased liver protein concentrations (33). How-
via a 2-step transport mechanism (38). First, glutamine is
ever, a leucine-rich (4%) diet fed to aged rats decreased body
transported inside the cell via the glutamine transporter
fat but did not have an effect on metabolic indicators of
(Slc1a5) that regulates glutamine intracellular concentra-
chronic diseases, such as total cholesterol, TGs, and glycemia
tions. Next, a complex of Slc7a5 and Slc3a2 (another glu-
(30). Nevertheless, several other studies highlighted the ther-
tamine transporter) uses intracellular glutamine as an
apeutic potential of leucine supplementation for the preven-
efflux substrate to regulate the uptake of extracellular leucine
tion or treatment of diabetes and obesity (31, 32). Leucine
into the cells (38). Once inside the cell, leucine can either
supplementation via drinking water in mice fed a high-fat diet
regulate cellular processes, be incorporated into protein,
led to significantly reduced weight gain and improved hy-
or undergo degradation starting with transfer of its amino
perglycemia and hypercholesterolemia and prevented diet-
group to a-ketoglutarate (transamination). The initia-
tion of leucine degradation occurs primarily in the mito-
7
Abbreviations used: ASS1, argininosuccinate synthase 1; BCATc, cytosolic branched-chain chondria of skeletal muscle and other tissues, because
aminotransferase; BCATm, mitochondrial branched-chain aminotransferase; BCKA,
branched-chain a-keto acid; BCKDC, branched-chain a-keto acid dehydrogenase complex; leucine as well as isoleucine and valine transamination is
BDK, branched-chain a-keto acid dehydrogenase kinase; HMB, limited in the liver (26). Leucine transamination is cata-
b-hydroxy-b-methylbutyrate; IDO, indoleamine 2,3-dioxygenase; KIC, a-ketoisocaproate; lyzed by the BCATm enzyme (40). This is a reversible trans-
LAT, L-type amino acid transporter; MSUD, maple syrup urine disease; mTOR, mammalian
target of rapamycin; mTORC, mammalian target of rapamycin complex; NALA, fer of the a-amino group of leucine to a-ketoglutarate to
N-acetyl–leucine amide; NFAT, nuclear factor of activated T cells; PBMC, peripheral blood form glutamate and a-ketoisocaproate (KIC) (Figure 1).
mononuclear cell; PPM1K, protein phosphatase, Mg2+/Mn2+ dependent 1K; Rheb, Ras Approximately 20% of leucine is converted into KIC, whereas
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, the rest of leucine is used for protein synthesis in skeletal
eukaryotic translation initiation factor 4E-binding protein 1. muscle (42). Glutamate, on the other hand, undergoes either

Leucine and immunity 799S


FIGURE 1 Overview of leucine metabolism.
BCATc and BCATm catalyze the first step in
leucine degradation by transferring nitrogen
from leucine to a-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 a-keto acid dehydrogenase
complex; HMB, b-hydroxy-b-methylbutyrate; KIC,
a-ketoisocaproate; MC-CoA, b-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.

amidation to glutamine or transamination to a-ketoglutarate the presence of branched-chain a-keto acids (BCKAs) in
to generate alanine in multiple tissues (42, 43). the urine (50). One mechanism that explains leucine toxic-
BCATm is expressed in most human tissues except for ity in MSUD is leucine interference with neurotransmitter
liver hepatocytes (26). Likewise, BCATm is constitutively ex- synthesis. Leucine competes for amino acid transporters
pressed in T cells (44). Another enzyme that transaminates with other amino acids such as tyrosine and phenylalanine,
leucine is the cytosolic branched-chain aminotransferase which are precursors of neurotransmitters (51). However,
(BCATc). In contrast to BCATm, BCATc is expressed in the toxic leucine concentrations may also contribute to disrup-
nervous system but has limited expression in other adult hu- tion in the energy metabolism of the brain where leucine
man tissues (26). However, many cancer types as well as ac- can inhibit pyruvate dehydrogenase and a-ketoglutarate de-
tivated T cells express BCATc, and BCATc is implicated as hydrogenase (52, 53). Apart from leucine, the leucine me-
an important prognostic marker for cancer and a potential tabolite KIC also accumulates in patients with MSUD and
candidate for an immunosuppressive enzyme (44–47). can affect the brain bioenergetic homeostasis (54). The
Once BCATm converts leucine to KIC in muscle, a sub- mechanism includes uncoupling of oxidative phosphoryla-
stantial amount of KIC is released into the bloodstream and tion and inhibition of a-ketoglutarate dehydrogenase activ-
further metabolized in the liver by the branched-chain ity by KIC (54). In addition, in cancer patients with
a-keto acid dehydrogenase complex (BCKDC), a large mul- cachexia, leucine supplementation enhanced tumor pro-
tienzyme complex that contains multiple copies of 3 en- gression, although it was intended to protect against cancer-
zymes: a branched-chain a-keto acid decarboxylase (E1), induced cachexia. This is evident from a recent study
a dihydrolipoyl transacylase (E2), and a dihydrolipoyl de- that showed that leucine supplementation enhanced tumor
hydrogenase (E3) (26). BCKDC activity is regulated by growth in both lean and overweight mice with pancreatic
phosphorylation/dephosphorylation. It is inhibited by cancer (18). Thus, although leucine has important functions
the branched-chain a-keto acid dehydrogenase kinase in stimulating protein synthesis, excess leucine and KIC con-
(BDK), which phosphorylates the E1a subunit of the E1 en- centrations may have a negative impact on biological processes.
zyme. This process is reversed by protein phosphatase Another key metabolite of leucine metabolism is b-hydroxy-
(PPM1K), which activates BCKDC (48, 49). Leucine trans- b-methylbutyrate (HMB) (Figure 1). Approximately 5%
amination by BCATm and oxidative decarboxylation by of leucine is irreversibly converted to HMB, and HMB has
BCKDC regulate the supply of leucine for tissue protein been used as a dietary substitute of leucine with no adverse
synthesis and other leucine functions. They are also impor- effects (42). HMB can stimulate protein synthesis (55) and
tant for the prevention of buildup of toxic metabolites or attenuate protein degradation (56) in a much smaller dos-
excessive leucine concentrations. A congenital deficiency age than leucine and, as such, HMB has the potential to
in BCKDC leading to maple syrup urine disease (MSUD) is substitute for leucine as a nutrient signal when leucine sup-
associated with elevated plasma leucine concentrations and plementation is not practical or desirable (42).

800S Supplement
Leucine and leucine metabolism in immune impairment. T cell activation (22, 70–72). The mTOR signaling path-
A number of studies from the 1970s to the present have way contains 2 multiprotein complexes, mTORC1 and
shown that inadequate uptake of leucine or the other BCAAs mTORC2. The 2 complexes have different sensitivity to
leads to immune impairment (57–60). Jose and Good (57) the drug rapamycin, with mTORC1 being the primary target
showed that dietary restriction of leucine caused a signifi- of rapamycin. The 2 complexes also differ in their upstream
cant decrease in the lysis of tumor cells by lymphocytes. regulators, downstream outputs, and protein composition,
Likewise, decreased concentrations of BCAAs, commonly all of which are described in detail by Laplante and Sabatini
seen in patients with advanced liver cirrhosis, were associ- (69). Here, attention is given to one of the upstream proteins
ated with impairment of the function and maturation of of mTORC1 called Rag GTPase, because leucine is known to
dendritic cells (58). Oral administration of BCAAs had stim- activate mTORC1 in a Rag GTPase–dependent manner (73,
ulatory effects on peripheral blood mononuclear cells 74). Mammals have 4 Rag GTPases (A–D), which can form
(PBMCs) in these patients and led to increased IFN-g pro- heterodimers. Leucine promotes the loading of Rag A and
duction (58). Patients with advanced chronic hepatitis C Rag B with GTP, thus enabling this heterodimer to interact
suffered from malnutrition, and plasma BCAAs were de- with mTORC1 (via Raptor) leading to mTORC1 activation
creased to similar concentrations seen in patients with liver (73). The exact mechanism of the regulatory role of leucine
cirrhosis (58, 59). Malnutrition impaired IFN-g signaling in is dependent on the enzyme leucyl–transfer RNA synthetase
these patients; however, an increase in the plasma concen- that catalyzes the ligation of leucine to its transfer RNA. This
trations of BCAAs upregulated IFN-g signaling and was pro- enzyme senses leucine cellular concentrations and activates
posed as a therapeutic approach for chronic hepatitis C (59). the Rag complex (74). Rag GTPase activates mTORC1 and
The mechanism of BCAA function in chronic hepatitis C triggers the translocation of mTORC1 to the lysosomal sur-
was further explored in a human hemochromatotic cell face. There, mTORC1 interacts with another protein, Ras
line (Huh-7.5) grown in low–amino acid media and infected homolog enriched in brain (Rheb), a small GTPase that ac-
with hepatitis C virus followed by supplementation with tivates mTORC1 (75). The interaction between mTORC1,
BCAAs. This mechanism involved the activation of the Rag GTPases, and Rheb on the lysosomal surface is possible
mTORC1 signaling pathway, restoration of IFN-g signaling, only if amino acids such as leucine are available, signifying
and repression of the replication of hepatitis C virus by the important role of the lysosome in amino acid sensing
BCAAs (59). by the mTORC1 signaling pathway (70, 75). The stimula-
Immunomodulatory effects of leucine metabolites tory effects of leucine on protein synthesis during exercise,
(HMB, KIC) were studied in human PBMCs and in sheep protein-energy malnutrition, and adipogenesis are associ-
(61–63). Treatment with HMB reduced TNF-a concentra- ated with the leucine-dependent activation of mTORC1 as
tions in the PBMC culture medium, modified T helper well as activation of downstream targets of mTORC1 such
(Th) 1/Th2 cytokine production toward a Th2 profile, and as the p70 ribosomal S6 kinase 1 (S6K1) and the eukaryotic
impaired lymphocyte proliferation and progression through translation initiation factor 4E–binding protein 1 (4E-BP1)
the cell cycle (61). Similarly, when KIC was tested in acti- (Figure 1) (16, 76, 77). On the other hand, withdrawal of
vated PMBCs, it was found that KIC suppressed lymphocyte leucine was shown to be as effective in inhibiting mTORC1
DNA synthesis (62). On the contrary, KIC was shown to signaling as was withdrawal of all amino acids (65). These
stimulate lymphocyte blastogenesis and antibody responses findings strongly suggest a central role for leucine in regulat-
in sheep (63). Moreover, feeding lambs with KIC prevented ing the mTORC1 signaling pathway in a variety of cellular
adrenocorticotropin-induced suppression of lymphocyte processes.
function, and this effect was not achieved by feeding the
lambs with leucine (63). Considering that KIC transami- Leucine is indispensable for mTORC1 regulation of
nates with glutamate to form leucine (64), the effect of T cell activation. The mTOR signaling pathway is a vital
KIC could also reflect changes in intracellular leucine or glu- link between the development of immune response, the sur-
tamate and its metabolites or a direct action of high concen- rounding environment, and cellular metabolism (22). In T
trations of intracellular KIC. Elucidating the underlying cells, a primary role of mTOR signaling pathway is to sense
mechanism or mechanisms may reveal therapeutic uses and integrate environmental cues that dictate the fate of na-
for leucine metabolites. ive T cells upon T cell receptor engagement and is essential
for Th1 and Th17 differentiation (78). Thus, it is not sur-
Molecular mechanisms of leucine function. There is evi- prising that knocking out mTORC1 in T cells affects their
dence that shows a role for leucine in regulating the lineage commitment (21, 22, 24). Delgoffe et al. (21) showed
mTOR signaling pathway (16, 17, 65–68). As elegantly de- that the absence of mTORC1 impaired the ability of T cells
scribed by Laplante and Sabatini (69, 70), mTOR signaling to differentiate into Th1 or Th17 cells. Similarly, T cells lack-
integrates extracellular and intracellular signals to regulate ing the mTORC1 activator Rheb failed to differentiate to
protein translation, and cell metabolism, growth, prolifera- Th1 and Th17 cells (79). On the other hand, deletion of tu-
tion, and survival. Thus, mTOR is activated during cellular berous sclerosis complex 1 (TSC1), an upstream inhibitor of
processes that use energy and nutrients, such as tumor for- mTORC1, caused multiorgan inflammation in mice not ex-
mation, angiogenesis, insulin resistance, adipogenesis, and pressing TSC1 as a consequence of hyperactivation of T cells

Leucine and immunity 801S


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 a-ketoacid; KIC, a-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)]; a-KG, a-ketoglutarate.

by greater activity of mTORC1 and elevated Th1 and Th17 re- Foxp3. These data support a potential role of BCATc in tu-
sponses (80). Interestingly, a deficiency in the amino acid mor-induced Tregs (J Powell, unpublished data, 2010).
transporters Slc7a5 (LAT1) and Slc1a5 (ASCT2) in mice also During activation, the T cell receptor (TCR) engagement,
impaired the differentiation of Th1 and Th17 cells in an coupled with CD28 signaling, upregulates the mTOR signal-
mTORC1-dependent manner (5, 39). As discussed above, ing pathway, which, in turn, stimulates glycolysis for energy
Slc1a5 is a glutamine transporter that controls glutamine up- and metabolites necessary for the increased biosynthetic de-
take and glutamine intracellular concentrations (38). Although mands of the proliferating T cells (94). This metabolic reprog-
glutamine has been studied extensively in T cells and immunity ramming increases the nutrient demands of T cells and leads
(81–86), the role of leucine in T cell activation is only now to an increased expression of glucose and amino acid trans-
emerging. Leucine availability was shown to be essential for porters (39). Sinclair et al. (39) found that the leucine trans-
T cell activation and proliferation (87, 88). In Jurkat T cells porter Slc7a5 was induced by the TCR signaling in a nuclear
and activated primary mouse T cells, the leucine structural an- factor of activated T cells (NFAT)–dependent manner. Cyclo-
tagonist N-acetyl–leucine amide (NALA) exerted similar effects sporine A, an immunosuppressive drug that inhibits this
on cell cycle progression, cell proliferation, cytokine produc- pathway, suppressed the expression of Slc7a5 in TCR-acti-
tion, and downstream targets of mTORC1 such as rapamycin, vated T cells. Slc7a5 mediated the intracellular transport of
suggesting the restriction of leucine availability (87, 88). Rapa- leucine, which was essential for the activity of mTORC1 dur-
mycin inhibits mTORC1 and renders T cells hyporesponsive ing T cell activation. The role of Slc7a5 in intracellular leucine
(anergic) even when they are given full signal 1 and signal 2 ac- uptake during T cell activation was further confirmed in hu-
tivation (87). The stimulation of T cells in the presence of ra- man T cells (6). Thus, leucine transport was established as an
pamycin makes them tolerant, such that they fail to produce important factor controlling mTORC1 activity in T cells.
substantial IL-2 or IFN-g upon rechallenge (even in the ab- The significance of Slc7a5 involvement in T cell activa-
sence of rapamycin) (89). By promoting tolerance, rapamycin tion was further shown in Slc7a5 null T cells, which failed
as well as other inhibitors of mTOR have become attractive to properly differentiate into CD4+ and CD8+ T cells (39).
agents for preventing transplant rejection (78, 90, 91). Other However, the severe phenotype of Slc7a5 null T cells was
means to inhibit mTOR signaling in T cells and reduce graft not solely due to the loss of mTORC1 activity caused by
rejection is by limiting essential amino acids, including leucine. failed leucine uptake but also reflected the inability of these
Skin grafts in T cell–deficient mice were shown to express tran- cells to express c-myc (39). The transcriptional factor c-MYC
scripts of 4 amino acid–consuming enzymes (BCATc being was identified as a critical regulator of T cell activation–induced
one of them) (92). Transplanted tissues are enriched in regula- metabolic reprogramming associated with global changes in
tory T cells (Tregs), which are known to maintain peripheral genes important for glucose catabolism, glutaminolysis, or-
tolerance to both self- and nonself-antigens (78, 93). Cobbold nithine and polyamine biosynthesis, and glucose and amino
et al. (92) postulated that tolerant Tregs could establish “infec- acid transport (GLUT1 and SLC7A5 among them) (82). The
tious tolerance” by inducing amino acid–consuming enzymes. c-MYC–mediated induction of glycolysis and glutaminolysis
This resulted in localized depletion of essential amino acids in activated T cells is reminiscent of the same processes in
including leucine depletion and promoted the induction of cancer cells (95). Although a direct connection between
anergy and more Tregs (92). Our preliminary analysis of c-MYC, leucine metabolism, and T cells has not been found,
CD4+CD25+ Tregs infiltrating prostate cancer revealed that BCATc has been described as one of the c-MYC target genes
BCATc is markedly upregulated in these cells along with in cancer cells (47, 95, 96).

802S Supplement
To better understand the mechanism of c-MYC leucine References
regulatory role during T cell activation, our group ex- 1. Schwartz RH. Costimulation of T lymphocytes: the role of CD28,
plored the impact of leucine metabolic enzymes in this CTLA-4, and B7/BB1 in interleukin-2 production and immunotherapy.
Cell 1992;71:1065–8.
process. We found that BCATc, which is not normally ex-
2. Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev
pressed in naive resting T cells, was induced by the TCR in Immunol 2009;27:591–619.
a manner similar to Slc7a5 (39, 44). TCR alone was suffi- 3. Buck MD, O’Sullivan D, Pearce EL. T cell metabolism drives immunity.
cient to trigger BCATc expression, whereas the mitochon- J Exp Med 2015;212:1345–60.
drial enzyme, BCATm, was expressed constitutively. 4. Jones RG, Thompson CB. Revving the engine: signal transduction fuels
Furthermore, activated T cells from global BCATc 2/2
T cell activation. Immunity 2007;27:173–8.
5. Nakaya M, Xiao Y, Zhou X, Chang JH, Chang M, Cheng X, Blonska M,
mice showed increased phosphorylation of mTORC1 Lin X, Sun SC. Inflammatory T cell responses rely on amino acid trans-
downstream targets ribosomal protein S6 and 4E-BP1. porter ASCT2 facilitation of glutamine uptake and mTORC1 kinase ac-
These cells had higher intracellular leucine concentrations tivation. Immunity 2014;40:692–705.
and also showed higher rates of glycolysis, glycolytic ca- 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
pacity, and glycolytic reserve when compared with acti-
human T cells. J Immunol 2013;191:4080–5.
vated wild-type cells (44). Our results, along with the 7. Li P, Yin YL, Li D, Kim SW, Wu G. Amino acids and immune function.
role of Slc7a5 in T cell activation, are consistent with a Br J Nutr 2007;98:237–52.
model in which TCR triggers the expression of Slc7a5 8. Barbul A. Arginine: biochemistry, physiology, and therapeutic implica-
and BCATc to regulate the uptake and the cytosolic con- tions. JPEN J Parenter Enteral Nutr 1986;10:227–38.
9. Brand K. Glutamine and glucose metabolism during thymocyte prolif-
centrations of leucine, respectively (Figure 2). In this
eration. pathways of glutamine and glutamate metabolism. Biochem J
model, BCATc is a part of a negative feedback loop con- 1985;228:353–61.
trolling the input of leucine toward the mTORC1 signaling 10. Grohmann U, Bronte V. Control of immune response by amino acid
pathway (Figure 2A). Excess leucine is transaminated to metabolism. Immunol Rev 2010;236:243–64.
KIC, which exits the cells (44). Loss of BCATc expression 11. Garlick PJ. The role of leucine in the regulation of protein metabolism.
J Nutr 2005;135(6, Suppl):1553S–6S.
eliminates cytosolic leucine catabolism and, as a result, the 12. Wu G, Morris SM Jr. Arginine metabolism: nitric oxide and beyond.
availability of leucine to activate mTORC1 is not limited, Biochem J 1998;336(Pt 1):1–17.
potentially resulting in T cell hyperactivation (Figure 2B). 13. Bronte V, Serafini P, Mazzoni A, Segal DM, Zanovello P. L-arginine me-
Future studies in animal models of human diseases will tabolism in myeloid cells controls T-lymphocyte functions. Trends Im-
be important to verify this paradigm in vivo. Nevertheless, munol 2003;24:302–6.
14. Mellor AL, Munn DH. Tryptophan catabolism and T-cell tolerance: im-
these studies show that leucine uptake and leucine metab-
munosuppression by starvation? Immunol Today 1999;20:469–73.
olism in T cells are critical for the regulation of the 15. Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and
mTORC1 signaling pathway during T cell activation, and tryptophan catabolism. Nat Rev Immunol 2004;4:762–74.
manipulating the immune response by targeting leucine 16. Columbus DA, Steinhoff-Wagner J, Suryawan A, Nguyen HV,
could prove useful in treating infections, autoimmunity, Hernandez-Garcia A, Fiorotto ML, Davis TA. Impact of pro-
longed leucine supplementation on protein synthesis and lean growth
and/or cancer. in neonatal pigs. Am J Physiol Endocrinol Metab 2015;309:E601–10.
17. Gao X, Tian F, Wang X, Zhao J, Wan X, Zhang L, Wu C, Li N, Li J.
Conclusions Leucine supplementation improves acquired growth hormone resis-
Substantial progress has been made in our understanding tance in rats with protein-energy malnutrition. PLoS One 2015;10:
of leucine and its role as a nutrient signal activating the e0125023.
mTORC1 pathway in processes as diverse as muscle func- 18. Liu KA, Lashinger LM, Rasmussen AJ, Hursting SD. Leucine supple-
mentation differentially enhances pancreatic cancer growth in lean
tion, insulin resistance, and the activation of the immune and overweight mice. Cancer Metab 2014;2(1):6.
response. Moreover, new and exciting studies have con- 19. Anthony JC, Anthony TG, Layman DK. Leucine supplementation en-
nected leucine metabolism via BCATc with cancer pro- hances skeletal muscle recovery in rats following exercise. J Nutr
gression; thus, BCATc has emerged as a new prognostic 1999;129:1102–6.
20. Churchward-Venne TA, Breen L, Di Donato DM, Hector AJ, Mitchell
marker for cancer (45, 97). Amino acid metabolic enzymes
CJ, Moore DR, Stellingwerff T, Breuille D, Offord EA, Baker SK,
in tryptophan and arginine metabolism, such as IDO, ar- et al. Leucine supplementation of a low-protein mixed macronutrient
gininosuccinate synthase 1 (ASS1), and arginase, have beverage enhances myofibrillar protein synthesis in young men: a dou-
been well established in cancer and cancer immunother- ble-blind, randomized trial. Am J Clin Nutr 2014;99:276–86.
apy and are subjects of preclinical and clinical trials (98, 21. Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B,
99). Further studies targeting BCATc and leucine metabo- Worley PF, Kozma SC, Powell JD. The mTOR kinase differentially reg-
ulates effector and regulatory T cell lineage commitment. Immunity
lism in cancer and immune disorders will expand our 2009;30:832–44.
knowledge and opportunities for development of novel 22. Powell JD, Pollizzi KN, Heikamp EB, Horton MR. Regulation of im-
therapeutic approaches. mune responses by mTOR. Annu Rev Immunol 2012;30:39–68.
23. Liu C, Chapman NM, Karmaus PW, Zeng H, Chi H. mTOR and
metabolic regulation of conventional and regulatory T cells. J
Acknowledgments
Leukoc Biol 2015 Feb 24 (Epub ahead of print; pii: jlb.2RI0814-
We thank Adele Addington for critical evaluation of the 408R).
manuscript. All authors read and approved the final 24. Delgoffe GM, Powell JD. mTOR: taking cues from the immune micro-
manuscript. environment. Immunology 2009;127:459–65.

Leucine and immunity 803S


25. Hutson SM, Sweatt AJ, Lanoue KF. Branched-chain [corrected] amino 45. Tönjes M, Barbus S, Park YJ, Wang W, Schlotter M, Lindroth AM, Ple-
acid metabolism: implications for establishing safe intakes. J Nutr 2005; ier SV, Bai AH, Karra D, Piro RM, et al. BCAT1 promotes cell prolifer-
135(6, Suppl):1557S–64S. Erratum in J Nutr. 2005;135:2009. ation through amino acid catabolism in gliomas carrying wild-type
26. Sweatt AJ, Wood M, Suryawan A, Wallin R, Willingham MC, Hutson IDH1. Nat Med 2013;19:901–8.
SM. Branched-chain amino acid catabolism: unique segregation of 46. Yoshikawa R, Yanagi H, Shen CS, Fujiwara Y, Noda M, Yagyu T, Gega
pathway enzymes in organ systems and peripheral nerves. Am J Physiol M, Oshima T, Yamamura T, Okamura H, et al. ECA39 is a novel distant
Endocrinol Metab 2004;286:E64–76. metastasis-related biomarker in colorectal cancer. World J Gastroenter-
27. Garber AJ, Karl IE, Kipnis DM. Alanine and glutamine synthesis and ol 2006;12:5884–9.
release from skeletal muscle. II. The precursor role of amino acids in 47. Zhou W, Feng X, Ren C, Jiang X, Liu W, Huang W, Liu Z, Li Z, Zeng L,
alanine and glutamine synthesis. J Biol Chem 1976;251:836–43. Wang L et al. Over-expression of BCAT1, a c-myc target gene, induces
28. Norton LE, Layman DK. Leucine regulates translation initiation of protein cell proliferation, migration and invasion in nasopharyngeal carcinoma.
synthesis in skeletal muscle after exercise. J Nutr 2006;136(Suppl):533S–7S. Mol Cancer 2013;12:53.
29. Sugawara T, Ito Y, Nishizawa N, Nagasawa T. Regulation of muscle pro- 48. Lynch CJ, Adams SH. Branched-chain amino acids in metabolic signal-
tein degradation, not synthesis, by dietary leucine in rats fed a protein- ling and insulin resistance. Nat Rev Endocrinol 2014;10:723–36.
deficient diet. Amino Acids 2009;37:609–16. 49. Harris RA, Popov KM, Zhao Y, Kedishvili NY, Shimomura Y, Crabb
30. Vianna D, Resende GF, Torres-Leal FL, Pantaleao LC, Donato J Jr., Ti- DW. A new family of protein kinases—the mitochondrial protein ki-
rapegui J. Long-term leucine supplementation reduces fat mass gain nases. Adv Enzyme Regul 1995;35:147–62.
without changing body protein status of aging rats. Nutrition 2012; 50. Burrage LC, Nagamani SC, Campeau PM, Lee BH. Branched-chain
28:182–9. amino acid metabolism: from rare Mendelian diseases to more com-
31. Zhang Y, Guo K, LeBlanc RE, Loh D, Schwartz GJ, Yu YH. Increasing mon disorders. Hum Mol Genet 2014;23 R1:R1–8.
dietary leucine intake reduces diet-induced obesity and improves glu- 51. Oldendorf WH, Szabo J. Amino acid assignment to one of three blood-
cose and cholesterol metabolism in mice via multimechanisms. Diabe- brain barrier amino acid carriers. Am J Physiol 1976;230:94–8.
tes 2007;56:1647–54. 52. Patel MS, Auerbach VH, Grover WD, Wilbur DO. Effect of the
32. She P, Reid TM, Bronson SK, Vary TC, Hajnal A, Lynch CJ, Hutson SM. branched-chain alpha-keto acids on pyruvate metabolism by homoge-
Disruption of BCATm in mice leads to increased energy expenditure as- nates of human brain. J Neurochem 1973;20:1793–6.
sociated with the activation of a futile protein turnover cycle. Cell 53. Patel MS. Inhibition by the branched-chain 2-oxo acids of the 2-oxo-
Metab 2007;6:181–94. glutarate dehydrogenase complex in developing rat and human brain.
33. Donato J Jr., Pedrosa RG, Cruzat VF, Pires IS, Tirapegui J. Effects of Biochem J 1974;144:91–7.
leucine supplementation on the body composition and protein status 54. Amaral AU, Leipnitz G, Fernandes CG, Seminotti B, Schuck PF, Wajner
of rats submitted to food restriction. Nutrition 2006;22:520–7. M. Alpha-ketoisocaproic acid and leucine provoke mitochondrial bio-
34. Wang Q, Holst J. L-type amino acid transport and cancer: targeting the energetic dysfunction in rat brain. Brain Res 2010;1324:75–84.
mTORC1 pathway to inhibit neoplasia. Am J Cancer Res 2015;5:1281–94. 55. Wheatley SM, El-Kadi SW, Suryawan A, Boutry C, Orellana RA,
35. Kanai Y, Segawa H, Miyamoto K, Uchino H, Takeda E, Endou H. Ex- Nguyen HV, Davis SR, Davis TA. Protein synthesis in skeletal mus-
pression cloning and characterization of a transporter for large neutral cle of neonatal pigs is enhanced by administration of beta-hydroxy-
amino acids activated by the heavy chain of 4F2 antigen (CD98). J Biol beta-methylbutyrate. Am J Physiol Endocrinol Metab 2014;306:
Chem 1998;273:23629–32. E91–9.
36. Segawa H, Fukasawa Y, Miyamoto K, Takeda E, Endou H, Kanai Y. 56. Noh KK, Chung KW, Choi YJ, Park MH, Jang EJ, Park CH, Yoon C,
Identification and functional characterization of a Na+-independent Kim ND, Kim MK, Chung HY. Beta-hydroxy beta-methylbutyrate im-
neutral amino acid transporter with broad substrate selectivity. J Biol proves dexamethasone-induced muscle atrophy by modulating the
Chem 1999;274:19745–51. muscle degradation pathway in SD rat. PLoS One 2014;9:e102947.
37. Mastroberardino L, Spindler B, Pfeiffer R, Skelly PJ, Loffing J, Shoe- 57. Jose DG, Good RA. Quantitative effects of nutritional protein and cal-
maker CB, Verrey F. Amino-acid transport by heterodimers of orie deficiency upon immune responses to tumors in mice. Cancer Res
4F2hc/CD98 and members of a permease family. Nature 1998;395: 1973;33:807–12.
288–91. 58. Kakazu E, Ueno Y, Kondo Y, Fukushima K, Shiina M, Inoue J, Tamai K,
38. Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, Ninomiya M, Shimosegawa T. Branched chain amino acids enhance the
Yang H, Hild M, Kung C, Wilson C, et al. Bidirectional transport of maturation and function of myeloid dendritic cells ex vivo in patients
amino acids regulates mTOR and autophagy. Cell 2009;136:521–34. with advanced cirrhosis. Hepatology 2009;50:1936–45.
39. Sinclair LV, Rolf J, Emslie E, Shi YB, Taylor PM, Cantrell DA. Control of 59. Honda M, Takehana K, Sakai A, Tagata Y, Shirasaki T, Nishitani S,
amino-acid transport by antigen receptors coordinates the metabolic Muramatsu T, Yamashita T, Nakamoto Y, Mizukoshi E, et al. Malnu-
reprogramming essential for T cell differentiation. Nat Immunol trition impairs interferon signaling through mTOR and FoxO path-
2013;14:500–8. ways in patients with chronic hepatitis C. Gastroenterology 2011;141:
40. Hutson SM, Fenstermacher D, Mahar C. Role of mitochondrial trans- 128,40, 140.e1–2.
amination in branched chain amino acid metabolism. J Biol Chem 60. Kobayashi M, Ikeda K, Arase Y, Suzuki Y, Suzuki F, Akuta N, Hosaka
1988;263:3618–25. T, Murashima N, Saitoh S, Someya T, et al. Inhibitory effect of
41. Zanchi NE, Gerlinger-Romero F, Guimaraes-Ferreira L, de Siqueira Fil- branched-chain amino acid granules on progression of compensated
ho MA, Felitti V, Lira FS, Seelaender M, Lancha AH Jr. HMB supple- liver cirrhosis due to hepatitis C virus. J Gastroenterol 2008;43:63–
mentation: clinical and athletic performance-related effects and 70.
mechanisms of action. Amino Acids 2011;40:1015–25. 61. Nunes EA, Lomax AR, Noakes PS, Miles EA, Fernandes LC, Calder PC.
42. Duan Y, Li F, Li Y, Tang Y, Kong X, Feng Z, Anthony TG, Watford M, Beta-hydroxy-beta-methylbutyrate modifies human peripheral blood
Hou Y, Wu G, et al. The role of leucine and its metabolites in protein mononuclear cell proliferation and cytokine production in vitro. Nu-
and energy metabolism. Amino Acids 2015;48:41–51. trition 2011;27:92–9.
43. Wu G, Thompson JR, Sedgwick GW, Drury M. Formation of alanine 62. Santos KD, Rocha M, Wannmacher CM, Wajner M. The influence of
and glutamine in chick (Gallus domesticus) skeletal muscle. Comp Bi- organic acids on the proliferation of human peripheral lymphocytes ac-
ochem Physiol B 1989;93:609–13. tivated by concanavalin A and pokeweed mitogen. Int J Immunophar-
44. Ananieva EA, Patel CH, Drake CH, Powell JD, Hutson SM. Cytosolic macol 1996;18:761–9.
branched chain aminotransferase (BCATc) regulates mTORC1 signal- 63. Kuhlman G, Roth JA, Nissen S. Effects of alpha-ketoisocaproate on
ing and glycolytic metabolism in CD4+ T cells. J Biol Chem 2014; adrenocorticotropin-induced suppression of lymphocyte function in
289:18793–804. sheep. Am J Vet Res 1991;52:388–92.

804S Supplement
64. Hutson SM, Zapalowski C, Cree TC, Harper AE. Regulation of leucine 83. Blagih J, Coulombe F, Vincent EE, Dupuy F, Galicia-Vazquez G, Yurchenko
and alpha-ketoisocaproic acid metabolism in skeletal muscle. effects of E, Raissi TC, van der Windt GJ, Viollet B, Pearce EL, et al. The energy sen-
starvation and insulin. J Biol Chem 1980;255:2418–26. sor AMPK regulates T cell metabolic adaptation and effector responses in
65. Hara K, Yonezawa K, Weng QP, Kozlowski MT, Belham C, Avruch J. vivo. Immunity 2015;42:41–54.
Amino acid sufficiency and mTOR regulate p70 S6 kinase and eIF-4E 84. Sikalidis AK. Amino acids and immune response: a role for cysteine,
BP1 through a common effector mechanism. J Biol Chem 1998;273: glutamine, phenylalanine, tryptophan and arginine in T-cell function
14484–94. and cancer? Pathol Oncol Res 2015;21:9–17.
66. Melnik BC. Leucine signaling in the pathogenesis of type 2 diabetes and 85. Hsiung YC, Liu JJ, Hou YC, Yeh CL, Yeh SL. Effects of dietary gluta-
obesity. World J Diabetes 2012;3:38–53. mine on the homeostasis of CD4+ T cells in mice with dextran sulfate
67. Pedroso JA, Zampieri TT, Donato J Jr. Reviewing the effects of L-leucine sodium-induced acute colitis. PLoS One 2014;9:e84410.
supplementation in the regulation of food intake, energy balance, and 86. Hou YC, Wu JM, Wang MY, Wu MH, Chen KY, Yeh SL, Lin MT. Glu-
glucose homeostasis. Nutrients 2015;7:3914–37. tamine supplementation attenuates expressions of adhesion molecules
68. Gil JH, Kim CK. Effects of different doses of leucine ingestion following and chemokine receptors on T cells in a murine model of acute colitis.
eight weeks of resistance exercise on protein synthesis and hypertrophy Mediators Inflamm 2014.
of skeletal muscle in rats. J Exerc Nutrition Biochem 2015;19:31–8. 87. Zheng Y, Delgoffe GM, Meyer CF, Chan W, Powell JD. Anergic T cells
69. Laplante M, Sabatini DM. mTOR signaling at a glance. J Cell Sci 2009; are metabolically anergic. J Immunol 2009;183:6095–101.
122:3589–94. 88. Hidayat S, Yoshino K, Tokunaga C, Hara K, Matsuo M, Yonezawa K.
70. Laplante M, Sabatini DM. mTOR signaling in growth control and dis- Inhibition of amino acid-mTOR signaling by a leucine derivative in-
ease. Cell 2012;149:274–93. duces G1 arrest in Jurkat cells. Biochem Biophys Res Commun 2003;
71. Waickman AT, Powell JD. mTOR, metabolism, and the regulation of 301:417–23.
T-cell differentiation and function. Immunol Rev 2012;249:43–58. 89. Powell JD, Lerner CG, Schwartz RH. Inhibition of cell cycle progression
72. Waickman AT, Powell JD. Mammalian target of rapamycin integrates by rapamycin induces T cell clonal anergy even in the presence of cos-
diverse inputs to guide the outcome of antigen recognition in T cells. timulation. J Immunol 1999;162:2775–84.
J Immunol 2012;188:4721–9. 90. Gao W, Lu Y, El Essawy B, Oukka M, Kuchroo VK, Strom TB. Con-
73. Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled trasting effects of cyclosporine and rapamycin in de novo generation
L, Sabatini DM. The rag GTPases bind raptor and mediate amino acid of alloantigen-specific regulatory T cells. Am J Transplant 2007;7:
signaling to mTORC1. Science 2008;320:1496–501. 1722–32.
74. Han JM, Jeong SJ, Park MC, Kim G, Kwon NH, Kim HK, Ha SH, Ryu 91. Hsieh MM, Kang EM, Fitzhugh CD, Link MB, Bolan CD, Kurlander R,
SH, Kim S. Leucyl-tRNA synthetase is an intracellular leucine sensor Childs RW, Rodgers GP, Powell JD, Tisdale JF. Allogeneic hematopoietic
for the mTORC1-signaling pathway. Cell 2012;149:410–24. stem-cell transplantation for sickle cell disease. N Engl J Med 2009;361:
75. Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini DM. 2309–17.
Ragulator-rag complex targets mTORC1 to the lysosomal surface and is 92. Cobbold SP, Adams E, Farquhar CA, Nolan KF, Howie D, Lui KO,
necessary for its activation by amino acids. Cell 2010;141:290–303. Fairchild PJ, Mellor AL, Ron D, Waldmann H. Infectious tolerance
76. Gao HN, Hu H, Zheng N, Wang JQ. Leucine and histidine indepen- via the consumption of essential amino acids and mTOR signaling.
dently regulate milk protein synthesis in bovine mammary epithelial Proc Natl Acad Sci USA 2009;106:12055–60.
cells via mTOR signaling pathway. J Zhejiang Univ Sci B 2015;16: 93. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of
560–72. differentiation and function. Annu Rev Immunol 2012;30:531–64.
77. Lynch CJ, Fox HL, Vary TC, Jefferson LS, Kimball SR. Regulation of 94. Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR,
amino acid-sensitive TOR signaling by leucine analogues in adipocytes. Elstrom RL, June CH, Thompson CB. The CD28 signaling pathway
J Cell Biochem 2000;77:234–51. regulates glucose metabolism. Immunity 2002;16:769–77.
78. Lo YC, Lee CF, Powell JD. Insight into the role of mTOR and metab- 95. Yeung SJ, Pan J, Lee MH. Roles of p53, MYC and HIF-1 in regulating
olism in T cells reveals new potential approaches to preventing graft re- glycolysis— the seventh hallmark of cancer. Cell Mol Life Sci 2008;65:
jection. Curr Opin Organ Transplant 2014;19:363–71. 3981–99.
79. Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton 96. Wang ZQ, Faddaoui A, Bachvarova M, Plante M, Gregoire J, Renaud MC,
MR, Xiao B, Worley PF, Powell JD. The kinase mTOR regulates the dif- Sebastianelli A, Guillemette C, Gobeil S, Macdonald E, et al. BCAT1 ex-
ferentiation of helper T cells through the selective activation of signaling pression associates with ovarian cancer progression: possible implications
by mTORC1 and mTORC2. Nat Immunol 2011;12:295–303. in altered disease metabolism. Oncotarget 2015;6:31522–43.
80. Park Y, Jin HS, Lopez J, Elly C, Kim G, Murai M, Kronenberg M, Liu 97. Chang IW, Wu WJ, Wang YH, Wu TF, Liang PI, He HL, Yeh BW, Li CF.
YC. TSC1 regulates the balance between effector and regulatory T cells. BCAT1 overexpression is an indicator of poor prognosis in patients
J Clin Invest 2013;123:5165–78. with urothelial carcinomas of the upper urinary tract and urinary
81. Klysz D, Tai X, Robert PA, Craveiro M, Cretenet G, Oburoglu L, Mongellaz bladder. Histopathology 2015 Jul 14(Epub ahead of print; DOI:
C, Floess S, Fritz V, Matias MI, et al. Glutamine-dependent alpha- 10.1111/his.12778).
ketoglutarate production regulates the balance between T helper 98. Phillips MM, Sheaff MT, Szlosarek PW. Targeting arginine-dependent
1 cell and regulatory T cell generation. Sci Signal 2015;8:ra97. cancers with arginine-degrading enzymes: opportunities and chal-
82. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, McCormick lenges. Cancer Res Treat 2013;45:251–62.
LL, Fitzgerald P, Chi H, Munger J, Green DR. The transcription factor myc 99. Platten M, von Knebel Doeberitz N, Oezen I, Wick W, Ochs K. Cancer
controls metabolic reprogramming upon T lymphocyte activation. Immu- immunotherapy by targeting IDO1/TDO and their downstream effec-
nity 2011;35:871–82. tors. Front Immunol 2014;5:673.

Leucine and immunity 805S

You might also like