Metabolic Pathways in T Cell Fate and Function: Valerie A. Gerriets and Jeffrey C. Rathmell

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Review

Metabolic pathways in T cell fate and


function
Valerie A. Gerriets and Jeffrey C. Rathmell
Department of Pharmacology and Cancer Biology, Department of Immunology, Sarah Stedman Nutrition and Metabolism Center,
Duke University, Durham, NC 27710, USA

T cell growth and function must be tightly regulated to Increased metabolism is a crucial part of activation because
provide protection against foreign pathogens, while if T cells fail to increase glucose metabolism due to inade-
avoiding autoimmunity and immunodeficiency. It is quate nutrients or direct metabolic inhibition, activation
now apparent that T cell metabolism is highly dynamic and proliferation are suppressed [10–12]. Conversely, in-
and has a tremendous impact on the ability of T cells to creased glucose uptake can enhance T cell activation and
grow, activate and differentiate. Specific metabolic path- proliferation [10]. Although altered metabolic needs and
ways provide energy and biosynthetic precursors that activity certainly follow changes in signaling and prolifera-
must support specific cell functions, as effector, regula- tion rate, evidence is now emerging that the regulation of T
tory, memory, and alloreactive T cells have distinct cell metabolism is also intimately linked to T cell function
metabolic needs in immunity and inflammation. Here, and differentiation. In each stage of the life of a T cell,
we review the signaling pathways that control metabo- whether it is naı̈ve, activated, antigen-experienced, or aner-
lism and how the metabolic phenotypes of T cell sub- gic, cell metabolism must be matched to the function of that
types integrate with T cell function. Ultimately, these particular T cell (Table 1). This review focuses on how
metabolic differences may provide new opportunities to metabolic pathways integrate with T cell function and the
modulate the immune response and treat inflammatory possibility that manipulation of metabolic pathways may
and autoimmune diseases. provide a new approach to modulate the immune response.

Immunometabolism: linking metabolism and immune Metabolic dysregulation and T cell activation
function Activation of effector T cells leads to increased glucose
Although most normal healthy cells use oxidative phos- uptake, glycolysis, and lipid synthesis to support growth
phorylation as an energy source, Otto Warburg observed and proliferation (Figure 1). Just as altered metabolism
almost a century ago that cancer cells are glycolytic and can promote or support cancer cell growth and survival,
produce lactic acid even in the presence of oxygen [1,2]. improperly controlled T cell metabolism can lead to chronic
Although less efficient at producing ATP, this transforma- T cell activation and inflammatory disease. Indeed, direct
tion from oxidative metabolism to aerobic glycolysis manipulation of glucose metabolism in vivo has been
enables cancer cells to acquire the biosynthetic precursors shown to modulate inflammatory disease. Overexpression
necessary to support rapid growth and proliferation [3]. of the glucose transporter Glut1 leads to increased glucose
Based on this phenotype, the field of cancer biology is now uptake and glycolysis, and transgenic expression of Glut1
actively seeking to inhibit aerobic glycolysis characteristic specifically in T cells leads to increased T cell proliferation,
of tumor metabolism, as a treatment for cancer [4]. Al- survival and cytokine production [10,13]. These cells are
though fundamental to transformed cells, this metabolic larger than normal T cells and less dependent on co-
phenotype is not unique to tumors. Indeed, it is now clear stimulation for interleukin (IL)-2 production. In addition,
that this same metabolic transition to aerobic glycolysis is T cells from aged Glut1 transgenic mice exhibit elevated
crucial in T cell activation, independent of any transfor- surface levels of several activation markers, including
mation event, and that alterations in metabolism can CD25, CD44 and CD69. They also produce more IL-2
significantly affect normal T cell function. Modulation of and interferon (IFN)g than control cells. Ultimately, Glut1
lymphocyte metabolism may, therefore, also be beneficial transgenic mice develop lymphadenopathy and a mild
in the treatment of immune disorders. generalized inflammatory disorder [10,13].
Lymphocyte stimulation leads to an exceptionally high Failure of effector T cells to upregulate glucose metab-
rate of cell growth and proliferation. To fuel the energetic olism properly results in decreased cytokine production,
and biosynthetic demands of rapid clonal expansion, acti- proliferation, and can lead to apoptosis [10,11,14–16]. If T
vated T cells rapidly upregulate glucose uptake and glycol- cells survive this metabolic stress, inhibition of metabo-
ysis [5–8]. This process is co-stimulation dependent and lism during T cell activation can lead to anergy [9].
failure to receive sufficient co-stimulation leads not only Glucose metabolism in particular is essential for T cells,
to functional inactivation or anergy but also to metabolic as glucose deprivation prevents T cell function despite the
anergy, with a reduction in glucose metabolism [9]. presence of other metabolic fuels [10,11,14]. Except in
rare instances, glucose is generally available in the ex-
Corresponding author: Rathmell, J.C. (jeff.rathmell@duke.edu). tracellular environment and the availability of glucose for
168 1471-4906/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. doi:10.1016/j.it.2012.01.010 Trends in Immunology, April 2012, Vol. 33, No. 4
Review Trends in Immunology April 2012, Vol. 33, No. 4

Table 1. Metabolic differences between T cell subsets.


T cell subset Glucose utilization Primary metabolic phenotype Immune setting identified References
Naı̈ve Low Mix of fuels oxidized in the mitochondria [5,8]
CD4+ or CD8+ effector High Aerobic glycolysis Inflammation [5,7,10]
Lactate production Autoimmunity
(Infection?)
Treg Low Lipid oxidation Asthma [12,13]
CD8+ memory Low? Lipid oxidation [64]
Alloreactive Very high Aerobic glycolysis GVHD [24,25,27]
Increased OXPHOS
Anergic Low Failure to upregulate glucose metabolism [9]

lymphocyte metabolism is regulated instead by glucose shown to produce high levels of lactate and overexpress
uptake and the expression and trafficking patterns of pyruvate dehydrogenase kinase (PDK)1. PDK1 acts to
glucose transporters (Gluts) [17,18]. Glut1 is expressed inhibit pyruvate dehydrogenase and thus restrict en-
at a low level in naı̈ve T cells and is rapidly induced by trance of pyruvate into the mitochondrial citric acid cycle;
Myc following T cell receptor (TCR) activation [10,19]. instead promoting aerobic glycolysis and the production
Glut1 trafficking is also highly regulated, with Glut1 of lactic acid [22]. Treatment of CD4+ T cells isolated from
protein remaining in intracellular vesicles until T cell asthma patients with the PDK1 inhibitor dichloroacetate
activation. CD28 co-stimulation to further activate (DCA) promotes pyruvate oxidation in the mitochondria
the PI3K/Akt/mTOR pathway (PI3K: phosphoinositide and prevents inflammatory cytokine production and T cell
3-kinase; mTOR: mammalian target of rapamycin), in proliferation. As in human patients, T cells from mice in
particular, provides a signal for Glut1 expression and models of asthma produce high levels of lactate. Treat-
cell surface localization [7]. Constitutive Akt expression ment of these mice with DCA reduces lactate production
leads to a higher level of Glut1 protein on the surface of and inhibits airway inflammation in vivo. Inhibition of
the cell [20], whereas PI3K inhibition or Akt-deficiency aerobic glycolysis with DCA also inhibits collagen-
lowers glucose uptake [17,18,21]. induced arthritis in female mice [23].
Mitochondria have many metabolic roles and dysregu-
Metabolism and inflammatory disease lation of mitochondrial metabolism is also associated with
Targeting glucose metabolism has been shown to be autoimmune and inflammatory disease. There is increas-
effective to reduce T cell effector function. Treatment of ing evidence that chronically stimulated autoreactive T
mice with the glycolytic inhibitor 2-deoxyglucose, for cells preferentially meet their metabolic demands through
example, can suppress experimental autoimmune mitochondrial oxidative phosphorylation (OXPHOS) rath-
encephalomyelitis (EAE) [12]. In humans, chronically er than aerobic glycolysis. Autoreactive splenocytes in mice
activated T cells in allergic asthma patients have been with active lupus preferentially convert glucose to CO2
rather than lactate, increasing glucose oxidation by 40%
over control splenocytes and relying on mitochondrial
Glut1
metabolism for ATP synthesis in vivo [24]. Similarly,
chronically stimulated mouse T cells proliferating in re-
Glucose sponse to alloantigen in the context of graft-versus-host
PPP
disease (GVHD) greatly increase both glycolysis and
Lipids Treg and
Ribose+
Ribose+ memory CD8
CD
D8
OXPHOS compared to normal T cells [25]. This altered
Glycolysis NADPH metabolic phenotype with chronic stimulation also occurs
in humans with autoimmune disease, because T cells from
ATP
ATP+pyruvate
Citrate
patients with systemic lupus erythematosus (SLE) have
Lip
pids
Lipids
mitochondrial abnormalities including elevated mitochon-
drial transmembrane potential, increased mitochondrial
ET/OXPHOS
S
Lactate mass and depletion of ATP [26,27].
TCA β-Ox This preference of chronically stimulated or alloreactive
lymphocytes for OXPHOS may allow selective targeting of
mitochondrial metabolism. Accordingly, the small mole-
Effector T cells cule Bz-423 that partially inhibits the mitochondrial F0F1
ATPase preferentially induces apoptosis of alloreactive T
TRENDS in Immunology
cells, with minimal impact on healthy lymphocytes [25].
Figure 1. Metabolic differences between effector, Treg and memory T cells. Upon Although Bz-423 does not appear to reduce cellular capac-
stimulation, CD4+ and CD8+ effector T cells upregulate glucose uptake and ity for ATP generation significantly, it does result in su-
glycolysis to provide biosynthetic precursors necessary for growth and
proliferation. Glucose enters the pentose phosphate pathway (PPP) to generate
peroxide generation that leads to apoptosis of alloreactive
ribose and NADPH necessary for nucleotide synthesis and is converted to citrate in cells [28]. This sensitivity is probably due to limited anti-
the citric acid cycle (TCA) for lipid synthesis. Lactate is produced as a byproduct of oxidant reserves in these oxidative cells. In vivo, Bz-423
the rapid rate of glycolysis in effector T cells. Treg and CD8+ memory cells instead
burn lipids through b-oxidation (b-ox) in the mitochondria and generate ATP
treatment reduces GVHD clinical scores and improves
through electron transport and oxidative phosphorylation (ET/OXPHOS). survival compared to vehicle treatment without impairing
169
Review Trends in Immunology April 2012, Vol. 33, No. 4

reconstitution in the spleen or thymus after bone marrow Box 1. Key players in T cell metabolism and differentiation
transplant [25].
Myc. Myc is a proto-oncogenic transcription factor that regulates
Nutrient excess or deprivation can also alter immune
genes involved in cell metabolism, proliferation and apoptosis. Myc
responses. Obesity has long been associated with low- regulates both glucose and glutamine metabolism to generate
grade inflammation, with macrophages and other immune biosynthetic precursors necessary for cell growth and proliferation
cells infiltrating the adipose tissue of obese mice and [37]. Myc has recently been shown to play a role in upregulating
humans [29]. Both the immune cells and the adipose tissue glucose and glutamine metabolism soon after T cell activation [38].
HIF1a. Hypoxia-inducible factor (HIF1)a is a transcription factor
itself produce inflammatory cytokines, such as tumor ne-
involved in the hypoxic response. HIF1a is constitutively transcribed
crosis factor (TNF)a, that continue the inflammatory re- and translated, but under normoxic conditions it is rapidly
sponse, and obese patients have an increased risk for a degraded. Under hypoxic conditions, the degradation of HIF1a is
wide array of inflammatory diseases [30,31]. By contrast, inhibited and it translocates to the nucleus where it upregulates
malnutrition is immunosuppressive and associated with glycolytic genes [39]. Importantly, HIF1a functions to direct glucose
away from the mitochondria; instead, upregulating glycolysis and
increased mortality from infectious disease [32]. Further- glucose uptake to support aerobic glycolysis. HIF1a is highly
more, anorexic patients with severe malnutrition have expressed in Th17 and is important in the balance between Th17
extremely low CD4+ T cell counts [33]. Mice fed a low and Treg differentiation [12,40].
protein diet also have decreased CD8+ memory T cells ERRa. Estrogen-related receptor (ERR)a is a hormone nuclear
and these cells have defects in homeostatic proliferation receptor that regulates energy metabolism pathways. There is no
known endogenous ligand and there is structural evidence that
and impaired recall response to a secondary infection [34]. ERRa is constitutively active without a ligand, instead regulated by
Although the root of weakened immunity in malnutrition its interactions with coactivators and corepressors including
is not entirely clear, it may be that the metabolic burden of peroxisomal proliferator-activated receptor g1 a and b (PGC1a/b)
an immune response is sufficient that it benefits animals to [43]. In T cells, ERRa promotes mitochondrial metabolism and is
link and balance metabolic and immune systems. In sup- important in T cell activation. Suppression of ERRa, either
genetically or pharmacologically, reduces Teff generation and
port of this link, a variety of hormones and neuropeptides, alleviates EAE in vivo [41].
including insulin and leptin, play a role in both metabolic mTOR/AMPK. Mammalian target of rapamycin (mTOR) is a serine/
and immune processes [35]. These systems are even con- threonine kinase responsible for integrating nutrient and energy
trolled by the same tissue in some organisms, such as status and regulating cell survival, growth and proliferation. mTOR
signals through two distinct complexes, mTORC1 and mTORC2,
Drosophila, where the fat body functions similarly to the
which have distinct functions in T cell differentiation [46,47].
mammalian immune system, adipose tissue and liver [36]. Inhibition of all mTOR activity prevents differentiation into Teff
and promotes the generation of Treg [50]. AMP-activated kinase
Metabolic reprogramming in T cell activation (AMPK) promotes catabolic processes and can phosphorylate
Mechanisms that control T cell metabolic reprogramming tuberous sclerosis 2 (TSC2), which in turn inhibits mTORC1 activity
[59]. Treating mice with metformin, which results in the activation of
are now coming to light, and many of the same oncogenes
AMPK, promotes Treg generation in a model of allergic asthma [13]
important in cancer metabolism are also crucial to drive T and promotes memory T cell generation after L. monocytogenes
cell metabolic transformations, most notably Myc, hypoxia infection and rechallenge [62].
inducible factor (HIF)1a, estrogen-related receptor (ERR)
a, and the mTOR pathway (Box 1). The proto-oncogenic
transcription factor Myc is known to promote transcription proven to be crucial to support generation of T cell effector
of genes for the cell cycle as well as aerobic glycolysis and function. In particular, the PI3K/Akt/mTOR pathway is
glutamine metabolism [37,38]. Recently, Myc has been important for T cell metabolism and is a potent target for T
shown to play an essential role to induce the expression cell immunosuppression. Inhibition or deficiency in this
of glycolytic and glutamine metabolism genes in the initial pathway leads to T cell anergy despite the presence of co-
hours of T cell activation [19]. In a similar fashion, the stimulation [44] and the mTOR inhibitor rapamycin is
transcription factor HIF1a can upregulate glycolytic genes used clinically as an immunosuppressive to treat a variety
to allow cancer cells to survive under hypoxic conditions of inflammatory disorders and to prevent transplant rejec-
[39]. The role for HIF1a in T cells is more complex, howev- tion [45,46]. Importantly, this pathway has been shown to
er, as HIF1a does not appear to play a role to promote play a key role to induce aerobic glycolysis and anabolic
glycolysis in initial T cell activation. Rather, HIF1a influ- processes in cell growth. Activation of mTOR complex 1
ences the balance of Th17:Treg cells (Th: T helper; Treg: T (mTORC1) induces genes involved in glycolysis, the pen-
regulatory) at later times [12,40]. In addition to glycolytic tose phosphate pathway and sterol and lipid biosynthesis
genes, other metabolic pathways, including mitochondrial [47].
pathways, must be upregulated during T cell activation to
fully support biosynthesis and cell growth. Consistent with Glucose is selectively required for effector CD4+ T cells
this diversity of metabolic demands, we recently showed a After early activation events, the cytokine environment
potential role for the nuclear hormone receptor ERRa in T promotes CD4+ T cells to differentiate into Teff (Th1, Th2
cell metabolism and activation [41]. ERRa is a well-known or Th17) or Treg subsets; each with a distinct functional role
mitochondrial regulator that is associated with a variety of in immunity. Th1 cells are involved with cell-mediated
cancers, and our data support a role for ERRa in mitochon- immunity, Th2 with humoral immunity, and Th17 with
drial metabolism of the glycolytic product, pyruvate, in mucosal immunity and inflammation [48], whereas Tregs
stimulated T cells [41–43]. limit inappropriate or excessive immune responses by sup-
In addition to transcriptional regulation, post-transla- pressing Teff cells [49]. Modulation of differentiation into
tional control of glucose uptake and glycolysis has also specific Teff or Treg subsets may, therefore, provide a way to
170
Review Trends in Immunology April 2012, Vol. 33, No. 4

(a) mTORC1 and mTORC2, with mTORC1 being downstream


Th1 of Akt and regulated by PI3K, whereas mTORC2 acts to
T-bet promote Akt activation. Together, these pathways inte-
C1
OR grate signals from the environment and contribute to T cell
mT
metabolism, activation and differentiation [52]. T cell spe-
Th2 cific mTOR knockouts fail to become Teffs even when
RC2
mTO GATA3 stimulated with the appropriate cytokines [50] and instead
Naïve ERRα Activated default to the Treg phenotype. Consistent with these find-
CD4+ CD4+ T cell HIF1
T cell Myc mTOR α , mT
ORC1
ings, the mTORC1 inhibitor rapamycin promotes both
Th17 fatty acid oxidation [53,54] and the generation of Tregs
AM RORγt [55]. Conversely, constitutive activation of the mTOR path-
PK
way instead promotes glycolysis and impairs the genera-
tion of Tregs [56]. This pathway has even more complicated
Treg roles in T cell metabolism and differentiation, because
FoxP3
mTORC1 and mTORC2 have specific effects on Teff sub-
sets. Ras homolog enriched in brain (Rheb)-deficient T
(b) cells, which lack mTORC1 activity, fail to become Th1
Naïve Activated
CD8+
Akt AMPK Memory and Th17 but can still differentiate into Th2, whereas
CD4+ T cell CD8+ T cell
T cell mTOR Traf6 mTORC2-deficient T cells differentiate into Th1 and
Th17 but not Th2 [57]. The role of cell metabolism relative
TRENDS in Immunology to altered signaling in these cell fate decisions remains
Figure 2. Signaling molecules involved in T cell metabolism and differentiation. (a) unclear, but this is a topic worth exploring given the broad
Activation of a naı̈ve CD4+ T cell requires appropriate TCR and co-stimulation, as impact of mTOR on metabolic pathways.
well as metabolic reprogramming and the signaling molecules mTOR, Myc and
The AMP-activated kinase (AMPK) promotes catabolic
ERRa contribute to this metabolic transition. An activated CD4+ T cell can
differentiate into an effector T cell (Th1, Th2 or Th17) or Treg depending on the processes [58] and can phosphorylate tuberous sclerosis 2
cytokine environment. mTORC1 is important for Th1 and Th17 differentiation, protein (TSC2), which in turn inhibits mTORC1 activity
mTORC2 for Th2, HIF1a for Th17 and AMPK for Treg differentiation. (b) Activation
of a naı̈ve CD8+ T cell also requires appropriate TCR and co-stimulation, as well as
[59]. Treg cells have high levels of phospho-AMPK and
metabolic reprogramming and the signaling molecules Akt and mTOR contribute treatment with metformin, an indirect activator of AMPK,
to this metabolic transition. TRAF6 and AMPK are involved in the transition from induces the generation of Tregs in a model of allergic
an effector to memory CD8+ T cell.
asthma [13]. Interestingly, although Akt is downstream
of PI3K and can regulate Glut1 and glycolysis [17], there is
evidence that it is not essential to coordinate metabolic
regulate immunity in the context of inflammatory disorders changes after initial activation of CD8+ T cells [60]. In this
[12,13,40,41,50]. Consistent with the distinct roles of CD4+ T setting, Akt instead regulates the expression of chemo-
cell subsets in immunity, the metabolic needs of CD4+ kines and adhesion molecules important for cell migration.
subsets differ and Teff and Treg are now known to utilize In addition to the mTOR pathway, HIF1a may also play
specific metabolic programs that may allow metabolic mod- a role in upregulating Teff glucose metabolism. Although
ulation of immunity (Figures 1 and 2a). not required in the first day of stimulation [19], HIF1a has
Although Teffs depend on glucose to support the rapid been shown to modulate the balance between Th17 and
growth and proliferation that accompanies clonal expan- Treg by upregulating glycolytic metabolism in Th17 cells in
sion, it is becoming increasingly evident that Tregs do not an mTOR-dependent manner [12]. In vitro, HIF1a defi-
share this same requirement. Tregs express low levels of ciency promotes Treg generation by stabilizing the Treg
cell surface Glut1 [13] and exhibit lower levels of glucose transcription factor forkhead box P3 (FoxP3) and directly
uptake and glycolysis; instead primarily relying on lipid inhibits IL-17 production [40]. In vivo, mice with HIF1a-
oxidation for energy [12,13]. Further, the addition of exog- deficient T cells have an increased number of Tregs and are
enous fatty acids promotes Treg generation while suppres- resistant to EAE [40]. The picture remains complex, but
sing Teff formation. Although lipids can have direct roles in together it appears that glucose metabolism is required for
signaling pathways in addition to providing a metabolic Teff survival and function but is dispensable and perhaps
fuel, the high rate of lipid oxidation and ability of Tregs to inhibitory to Treg generation.
persist even when glycolysis is inhibited suggest that lipid
metabolism can support Tregs [12,13]. In addition, al- Metabolism in CD8+ memory T cells
though inhibition of ERRa leads to a broad loss of glucose At the end of an immune response, the majority of effector
metabolism and inhibits T cell differentiation both in vitro T cells die by apoptosis, whereas a small percentage persist
and in vivo, the addition of exogenous fatty acids selec- as memory T cells [61]. Long-lived quiescent memory cells
tively rescues Treg, but not Teff generation. Together, are unlikely to have the same metabolic requirements as
these data suggest that Tregs are less reliant than Teffs effector T cells during an immune response. Pathways that
on glucose as a fuel [41]. control the generation of memory cells are of great interest
The PI3K/mTOR pathway plays a crucial role in pro- and TNF receptor associated factor (TRAF)6 has recently
moting the glucose metabolism required for effector T cell been found to be important in the transition from the
differentiation while at the same time inhibiting Treg effector to memory phenotype [62]. Importantly, TRAF6-
generation [47,51]. mTOR signals through two complexes, deficient cells have been shown to have defects switching
171
Review Trends in Immunology April 2012, Vol. 33, No. 4

from a glycolytic metabolism to oxidative fatty acid metab- References


1 Warburg, O. (1956) On the origin of cancer cells. Science 123, 309–314
olism, suggesting that this metabolic transition is required
2 Koppenol, W.H. et al. (2011) Otto Warburg’s contributions to current
for memory T cells (Figure 2b). In addition, the cytokine IL- concepts of cancer metabolism. Nat. Rev. Cancer 11, 325–337
15, which is important for memory T cells, promotes both 3 Vander Heiden, M.G. et al. (2009) Understanding the Warburg effect:
mitochondrial biogenesis and the expression of carnitine the metabolic requirements of cell proliferation. Science 324, 1029–
palmitoyl transferase (CPT)1a, a metabolic enzyme in- 1033
4 Vander Heiden, M.G. (2011) Targeting cancer metabolism: a
volved in fatty acid oxidation (FAO) [63]. Retroviral ex-
therapeutic window opens. Nat. Rev. Drug Discov. 10, 671–684
pression of CPT1A in T cells increases the survival and 5 Fox, C.J. et al. (2005) Fuel feeds function: energy metabolism and the T-
recall response of antigen-specific CD8+ T cells in vivo. Like cell response. Nat. Rev. Immunol. 5, 844–852
CD4+ Treg cells, memory CD8+ T cells exhibit high levels of 6 Maciver, N.J. et al. (2008) Glucose metabolism in lymphocytes is a
phospho-AMPK and treatment with metformin to activate regulated process with significant effects on immune cell function and
survival. J. Leukoc. Biol. 84, 949–957
AMPK and induce FAO further promote memory T cell 7 Frauwirth, K.A. et al. (2002) The CD28 signaling pathway regulates
generation in vivo [62]. Furthermore, treatment of mice glucose metabolism. Immunity 16, 769–777
with rapamycin during T cell contraction following viral 8 Jones, R.G. and Thompson, C.B. (2007) Revving the engine: signal
infections can enhance the formation of memory T cells and transduction fuels T cell activation. Immunity 27, 173–178
accelerate the transition from effector to memory T cells 9 Zheng, Y. et al. (2009) Anergic T cells are metabolically anergic. J.
Immunol. 183, 6095–6101
[64]. Importantly, these memory cells have a better recall 10 Jacobs, S.R. et al. (2008) Glucose uptake is limiting in T cell activation
response and are more protective following rechallenge and requires CD28-mediated Akt-dependent and independent
with virus. Memory cells have the ability to proliferate pathways. J. Immunol. 180, 4476–4486
rapidly following rechallenge and although the metabolic 11 Cham, C.M. and Gajewski, T.F. (2005) Glucose availability regulates
IFN-gamma production and p70S6 kinase activation in CD8+ effector
phenotype during this transition has not been described, it
T cells. J. Immunol. 174, 4670–4677
probably involves a shift back to glycolysis. 12 Shi, L.Z. et al. (2011) HIF1alpha-dependent glycolytic pathway
orchestrates a metabolic checkpoint for the differentiation of TH17
Concluding remarks and Treg cells. J. Exp. Med. 208, 1367–1376
13 Michalek, R.D. et al. (2011) Cutting edge: distinct glycolytic and lipid
It is now clear that T cell metabolism is intimately linked to
oxidative metabolic programs are essential for effector and regulatory
T cell survival, function and differentiation. Metabolic CD4+ T cell subsets. J. Immunol. 186, 3299–3303
differences between different types of T cells may, there- 14 Greiner, E.F. et al. (1994) Glucose is essential for proliferation and the
fore, provide a new direction for modulation of the immune glycolytic enzyme induction that provokes a transition to glycolytic
response. Just as effector, regulatory and memory T cells energy production. J. Biol. Chem. 269, 31484–31490
15 Coloff, J.L. et al. (2011) Akt requires glucose metabolism to suppress
play unique immunologic roles, they have distinct meta- puma expression and prevent apoptosis of leukemic T cells. J. Biol.
bolic profiles to provide the proper balance of energy and Chem. 286, 5921–5933
biosynthetic precursors to function properly. Effector T 16 Alves, N.L. et al. (2006) The Noxa/Mcl-1 axis regulates susceptibility to
cells require high levels of glucose to allow for rapid growth apoptosis under glucose limitation in dividing T cells. Immunity 24,
and cell division upon activation. This increase in glucose 703–716
17 Wieman, H.L. et al. (2007) Cytokine stimulation promotes glucose
metabolism is controlled by many of the same metabolic uptake via phosphatidylinositol-3 kinase/Akt regulation of Glut1
regulators that play an important role in cancer, including activity and trafficking. Mol. Biol. Cell 18, 1437–1446
PI3K/mTOR, HIF1a, Myc and ERRa. Treg and memory 18 Wofford, J.A. et al. (2008) IL-7 promotes Glut1 trafficking and glucose
CD8+ T cells instead mainly utilize fatty acids for energy. uptake via STAT5-mediated activation of Akt to support T-cell
survival. Blood 111, 2101–2111
Distinct programs for fuel utilization may serve additional
19 Wang, R. et al. (2011) The transcription factor Myc controls metabolic
purposes, because glucose is also used for protein glycosyl- reprogramming upon T lymphocyte activation. Immunity 35, 871–882
ation and acetyl CoA, the product of FAO, is the precursor 20 Rathmell, J.C. et al. (2003) Activated Akt promotes increased resting T
for acetylation of protein and DNA. The promotion of FAO cell size, CD28-independent T cell growth, and development of
in Treg cells may modulate lipid raft formation to alter autoimmunity and lymphoma. Eur. J. Immunol. 33, 2223–2232
21 Juntilla, M.M. et al. (2007) Akt1 and Akt2 are required for alphabeta
signaling pathways, lead to the breakdown of inflammato-
thymocyte survival and differentiation. Proc. Natl. Acad. Sci. U.S.A.
ry and lipid signaling molecules, or potentially influence 104, 12105–12110
epigenetic modifications. These differences in metabolism 22 Ostroukhova, M. et al. (2011) The role of low-level lactate production in
may provide an opportunity to modulate the balance be- airway inflammation in asthma. Am. J. Physiol. Lung Cell. Mol.
tween effector and regulatory T cells or to inhibit auto- Physiol. DOI: 10.1152/ajplung.00221.2011
23 Bian, L. et al. (2009) Dichloroacetate alleviates development of
reactive and inflammatory T cells with minimal effect on collagen II-induced arthritis in female DBA/1 mice. Arthritis Res.
healthy lymphocytes. Targeting T cell metabolism may, Ther. 11, R132
therefore, provide new directions to modulate the immune 24 Wahl, D.R. et al. (2010) Characterization of the metabolic phenotype of
response and treat an array of inflammatory diseases or to chronically activated lymphocytes. Lupus 19, 1492–1501
potentially impact T cell responses to infection. 25 Gatza, E. et al. (2011) Manipulating the bioenergetics of alloreactive T
cells causes their selective apoptosis and arrests graft-versus-host
disease. Sci. Transl. Med. 3, 67ra68
Acknowledgments 26 Gergely, P., Jr et al. (2002) Persistent mitochondrial hyperpolarization,
We thank Drs. Andrew Macintyre and Ryan Michalek for constructive increased reactive oxygen intermediate production, and cytoplasmic
comments and suggestions. National Institutes of Health Grants R01 alkalinization characterize altered IL-10 signaling in patients with
AI063345 and R01 HL108006 (to J.C.R.); the American Asthma systemic lupus erythematosus. J. Immunol. 169, 1092–1101
Foundation (J.C.R. is the Bernard Osher Fellow of the American 27 Gergely, P., Jr et al. (2002) Mitochondrial hyperpolarization and ATP
Asthma Foundation); the Lupus Research Institute (J.C.R); and the depletion in patients with systemic lupus erythematosus. Arthritis
Leukemia and Lymphoma Society (J.C.R.). Rheum. 46, 175–190

172
Review Trends in Immunology April 2012, Vol. 33, No. 4

28 Blatt, N.B. et al. (2008) Bz-423 superoxide signals apoptosis via 47 Duvel, K. et al. (2010) Activation of a metabolic gene regulatory
selective activation of JNK, Bak, and Bax. Free Radic. Biol. Med. network downstream of mTOR complex 1. Mol. Cell 39, 171–183
45, 1232–1242 48 Annunziato, F. and Romagnani, S. (2009) Heterogeneity of human
29 Gregor, M.F. and Hotamisligil, G.S. (2011) Inflammatory mechanisms effector CD4+ T cells. Arthritis Res. Ther. 11, 257
in obesity. Ann. Rev. Immunol. 29, 415–445 49 Sakaguchi, S. et al. (2008) Regulatory T cells and immune tolerance.
30 Agrawal, A. et al. (2011) Emerging interface between metabolic Cell 133, 775–787
syndrome and asthma. Am. J. Respir. Cell Mol. Biol. 44, 270–275 50 Delgoffe, G.M. et al. (2009) The mTOR kinase differentially regulates
31 Delgado, J. et al. (2008) Obesity and asthma. J. Investig. Allergol. Clin. effector and regulatory T cell lineage commitment. Immunity 30,
Immunol. 18, 420–425 832–844
32 Shears, P. (1991) Epidemiology and infection in famine and disasters. 51 Powell, J.D. et al. (2011) Regulation of immune responses by mTOR.
Epidemiol. Infect. 107, 241–251 Ann. Rev. Immunol. Mar 24 (Epub ahead of print)
33 Saito, H. et al. (2007) Malnutrition induces dissociated changes in 52 Laplante, M. and Sabatini, D.M. (2009) mTOR signaling at a glance. J.
lymphocyte count and subset proportion in patients with anorexia Cell Sci. 122, 3589–3594
nervosa. Int. J. Eat. Disord. 40, 575–579 53 Brown, N.F. et al. (2007) The mammalian target of rapamycin
34 Iyer, S.S. et al. (2012) Protein energy malnutrition impairs homeostatic regulates lipid metabolism in primary cultures of rat hepatocytes.
proliferation of memory CD8 T cells. J. Immunol. 188, 77–84 Metab. Clin. Exp. 56, 1500–1507
35 Matarese, G. and La Cava, A. (2004) The intricate interface between 54 Sipula, I.J. et al. (2006) Rapamycin-mediated inhibition of
immune system and metabolism. Trends Immunol. 25, 193–200 mammalian target of rapamycin in skeletal muscle cells reduces
36 Hoffmann, J.A. and Reichhart, J.M. (2002) Drosophila innate glucose utilization and increases fatty acid oxidation. Metab. Clin.
immunity: an evolutionary perspective. Nat. Immunol. 3, 121–126 Exp. 55, 1637–1644
37 Dang, C.V. et al. (2008) The interplay between MYC and HIF in cancer. 55 Battaglia, M. et al. (2005) Rapamycin selectively expands
Nat. Rev. Cancer 8, 51–56 CD4+CD25+FoxP3+ regulatory T cells. Blood 105, 4743–4748
38 Dang, C.V. (2010) Rethinking the Warburg effect with Myc 56 Haxhinasto, S. et al. (2008) The AKT-mTOR axis regulates de novo
micromanaging glutamine metabolism. Cancer Res. 70, 859–862 differentiation of CD4+Foxp3+ cells. J. Exp. Med. 205, 565–574
39 Semenza, G.L. (2009) Regulation of oxygen homeostasis by hypoxia- 57 Delgoffe, G.M. et al. (2011) The kinase mTOR regulates the
inducible factor 1. Physiology (Bethesda) 24, 97–106 differentiation of helper T cells through the selective activation of
40 Dang, E.V. et al. (2011) Control of T(H)17/T(reg) balance by hypoxia- signaling by mTORC1 and mTORC2. Nat. Immunol. 12, 295–303
inducible factor 1. Cell 146, 772–784 58 Mihaylova, M.M. and Shaw, R.J. (2011) The AMPK signalling pathway
41 Michalek, R.D. et al. (2011) Estrogen-related receptor-alpha is a coordinates cell growth, autophagy and metabolism. Nat. Cell Biol. 13,
metabolic regulator of effector T-cell activation and differentiation. 1016–1023
Proc. Natl. Acad. Sci. U.S.A. 108, 18348–18353 59 Inoki, K. et al. (2003) TSC2 mediates cellular energy response to control
42 Chang, C.Y. et al. (2011) The metabolic regulator ERRalpha, a cell growth and survival. Cell 115, 577–590
downstream target of HER2/IGF-1R, as a therapeutic target in 60 Macintyre, A.N. et al. (2011) Protein kinase B controls transcriptional
breast cancer. Cancer Cell 20, 500–510 programs that direct cytotoxic T cell fate but is dispensable for T cell
43 Villena, J.A. and Kralli, A. (2008) ERRalpha: a metabolic function for metabolism. Immunity 34, 224–236
the oldest orphan. Trends Endocrinol. Metab. 19, 269–276 61 D’Cruz, L.M. et al. (2009) Surviving the crash: transitioning from
44 Powell, J.D. et al. (1999) Inhibition of cell cycle progression by effector to memory CD8+ T cell. Semin. Immunol. 21, 92–98
rapamycin induces T cell clonal anergy even in the presence of 62 Pearce, E.L. et al. (2009) Enhancing CD8 T-cell memory by modulating
costimulation. J. Immunol. 162, 2775–2784 fatty acid metabolism. Nature 460, 103–107
45 Cutler, C. and Antin, J.H. (2010) Sirolimus immunosuppression for 63 van der Windt, G.J. et al. (2011) Mitochondrial respiratory capacity is a
graft-versus-host disease prophylaxis and therapy: an update. Curr. critical regulator of CD8(+) T cell memory development. Immunity
Opin. Hematol. 17, 500–504 DOI: 10.1016/j.immuni.2011.12.007
46 Young, D.A. and Nickerson-Nutter, C.L. (2005) mTOR—beyond 64 Araki, K. et al. (2009) mTOR regulates memory CD8 T-cell
transplantation. Curr. Opin. Pharmacol. 5, 418–423 differentiation. Nature 460, 108–112

173

You might also like