mtor signalling and metabolic regulation of t cell differentiation

7
Available online at www.sciencedirect.com mTOR signalling and metabolic regulation of T cell differentiation Christian Peter, Herman Waldmann and Stephen P Cobbold T cells constantly monitor energy status and nutrient levels in order to adjust metabolic pathways according to their nutritional status and other environmental stimuli. It is increasingly evident that the regulation of cellular metabolism is tightly coupled to T cell differentiation that ultimately determines the cellular fate. The mammalian target of Rapamycin (mTOR) pathway has emerged as a key player in sensing these nutritional/energetic signals and in addition, acts as a major integrator of growth factor induced signals, so placing mTOR at the core of a signalling network controlling metabolism and cellular fate. The mTOR pathway has been shown to play an important role in determining the differentiation of CD4 + T cells into inflammatory and regulatory subsets, in the induction of anergy, in the development of CD8 + memory T cells and the regulation of T cell trafficking. Address Sir William Dunn School of Pathology, Oxford University, South Parks Road, Oxford OX1 3RE, United Kingdom Corresponding author: Waldmann, Herman ([email protected]) Current Opinion in Immunology 2010, 22:655–661 This review comes from a themed issue on Immunogenetics and transplantation Edited by Terry Strom and Allan D. Kirk Available online 15th September 2010 0952-7915/$ – see front matter # 2010 Elsevier Ltd. All rights reserved. DOI 10.1016/j.coi.2010.08.010 T cell activation and mTOR regulated metabolic changes T cell activation is intimately linked to changes in the mode of energy generation, providing the basis for the energy demanding processes concurrent with T cell differentiation. Whereas resting T cells generate most of their energy by oxidative phosphorylation, T cell activation is characterized by a marked increase in gly- colysis [1]. This mode of energy generation is termed ‘aerobic glycolysis’ since it occurs despite a seemingly adequate oxygen supply. What causes this switch remains unclear. Based on the finding that increased mTOR activity can stimulate oxygen consumption by direct interaction with mitochondria or through an increased energetic load caused, for example, by increased protein synthesis, one could speculate that mTOR is involved in these metabolic changes [2,3]. In order to generate vast amounts of ATP from glucose and provide sufficient amino acids (AA) and lipids for protein and membrane synthesis respectively, T cells have to co-ordinately and rapidly up-regulate import of these reactants. In the case of glucose this might be achieved through AMP-activated protein kinase (AMPK) activation initiated by T cell receptor engagement and via Ca 2 + /calmodulin-dependent kinase kinase (CAMKK2) [4,5]. This might represent a mechanism by which T cells can anticipate rising energy demands required by T cell activation. Concurrently, activated T cells up- regulate surface expression of glucose transporter Glut1 by increasing its trafficking to meet the increasing demand for glucose. The rise in surface localisation of Glut1 seems to be dependent on PI3K/Akt activation resulting from TCR signalling and CD28 costimulation [6]. Glucose uptake seems to be enhanced further through mTOR-mediated activation of Glut1 [7]. In addition to increased glucose consumption activated T cells require enhanced protein synthesis and other anabolic pathways to facilitate cell growth, division and differentiation, thereby necessitating enhanced uptake of nutrients. T cell receptor and CD28 engagement induce Akt signalling via mTOR which leads to higher surface expression of a variety of nutrient transporters and receptors including AA transporters and the trans- ferrin receptor [8]. The fact that mTOR was originally discovered in response to the development of rapamy- cin as an immunosuppressive agent [9] highlights the crucial importance of the mTOR signalling pathway in T cell mediated immune responses, and further suggests that nutrient availability and metabolic regu- lation are intimately linked to T cell activation and differentiation. The sensing of nutrional and energetic status by mTOR mTOR signalling, as a function of the TORC1 complex [10 ], has been implicated as a major integrator of signals related to cellular nutrient and energy status. The mol- ecular mechanisms by which TORC1 controls the nutri- tional status of cells have only begun to be understood. In the first instance, cell intrinsic energy availability is sensed by AMPK, which is activated by rising AMP/ ATP ratios and hypoxia. AMPK negatively regulates TORC1 activity through activation of the TSC1/2 com- plex. This AMPK mediated sensing mechanism enables TORC1 to adjust metabolic pathways and other vital energy consuming cellular processes, such as protein synthesis with respect to the cellular energy status (Figure 1). www.sciencedirect.com Current Opinion in Immunology 2010, 22:655661

Upload: christian-peter

Post on 26-Jun-2016

215 views

Category:

Documents


3 download

TRANSCRIPT

Page 1: mTOR signalling and metabolic regulation of T cell differentiation

Available online at www.sciencedirect.com

mTOR signalling and metabolic regulation of T cell differentiationChristian Peter, Herman Waldmann and Stephen P Cobbold

T cells constantly monitor energy status and nutrient levels in

order to adjust metabolic pathways according to their

nutritional status and other environmental stimuli. It is

increasingly evident that the regulation of cellular metabolism is

tightly coupled to T cell differentiation that ultimately

determines the cellular fate. The mammalian target of

Rapamycin (mTOR) pathway has emerged as a key player in

sensing these nutritional/energetic signals and in addition, acts

as a major integrator of growth factor induced signals, so

placing mTOR at the core of a signalling network controlling

metabolism and cellular fate. The mTOR pathway has been

shown to play an important role in determining the

differentiation of CD4+ T cells into inflammatory and regulatory

subsets, in the induction of anergy, in the development of CD8+

memory T cells and the regulation of T cell trafficking.

Address

Sir William Dunn School of Pathology, Oxford University, South Parks

Road, Oxford OX1 3RE, United Kingdom

Corresponding author: Waldmann, Herman

([email protected])

Current Opinion in Immunology 2010, 22:655–661

This review comes from a themed issue on

Immunogenetics and transplantation

Edited by Terry Strom and Allan D. Kirk

Available online 15th September 2010

0952-7915/$ – see front matter

# 2010 Elsevier Ltd. All rights reserved.

DOI 10.1016/j.coi.2010.08.010

T cell activation and mTOR regulatedmetabolic changesT cell activation is intimately linked to changes in the

mode of energy generation, providing the basis for the

energy demanding processes concurrent with T cell

differentiation. Whereas resting T cells generate most

of their energy by oxidative phosphorylation, T cell

activation is characterized by a marked increase in gly-

colysis [1]. This mode of energy generation is termed

‘aerobic glycolysis’ since it occurs despite a seemingly

adequate oxygen supply. What causes this switch remains

unclear. Based on the finding that increased mTOR

activity can stimulate oxygen consumption by direct

interaction with mitochondria or through an increased

energetic load caused, for example, by increased protein

synthesis, one could speculate that mTOR is involved in

these metabolic changes [2,3].

www.sciencedirect.com

In order to generate vast amounts of ATP from glucose

and provide sufficient amino acids (AA) and lipids for

protein and membrane synthesis respectively, T cells

have to co-ordinately and rapidly up-regulate import of

these reactants. In the case of glucose this might be

achieved through AMP-activated protein kinase (AMPK)

activation initiated by T cell receptor engagement and via

Ca2+/calmodulin-dependent kinase kinase (CAMKK2)

[4,5]. This might represent a mechanism by which T

cells can anticipate rising energy demands required by T

cell activation. Concurrently, activated T cells up-

regulate surface expression of glucose transporter Glut1

by increasing its trafficking to meet the increasing

demand for glucose. The rise in surface localisation of

Glut1 seems to be dependent on PI3K/Akt activation

resulting from TCR signalling and CD28 costimulation

[6]. Glucose uptake seems to be enhanced further

through mTOR-mediated activation of Glut1 [7].

In addition to increased glucose consumption activated

T cells require enhanced protein synthesis and other

anabolic pathways to facilitate cell growth, division and

differentiation, thereby necessitating enhanced uptake

of nutrients. T cell receptor and CD28 engagement

induce Akt signalling via mTOR which leads to higher

surface expression of a variety of nutrient transporters

and receptors including AA transporters and the trans-

ferrin receptor [8]. The fact that mTOR was originally

discovered in response to the development of rapamy-

cin as an immunosuppressive agent [9] highlights the

crucial importance of the mTOR signalling pathway in

T cell mediated immune responses, and further

suggests that nutrient availability and metabolic regu-

lation are intimately linked to T cell activation and

differentiation.

The sensing of nutrional and energeticstatus by mTORmTOR signalling, as a function of the TORC1 complex

[10�], has been implicated as a major integrator of signals

related to cellular nutrient and energy status. The mol-

ecular mechanisms by which TORC1 controls the nutri-

tional status of cells have only begun to be understood. In

the first instance, cell intrinsic energy availability is

sensed by AMPK, which is activated by rising AMP/

ATP ratios and hypoxia. AMPK negatively regulates

TORC1 activity through activation of the TSC1/2 com-

plex. This AMPK mediated sensing mechanism enables

TORC1 to adjust metabolic pathways and other vital

energy consuming cellular processes, such as protein

synthesis with respect to the cellular energy status

(Figure 1).

Current Opinion in Immunology 2010, 22:655–661

Page 2: mTOR signalling and metabolic regulation of T cell differentiation

656 Immunogenetics and transplantation

Figure 1

The mTORC1 signalling pathway. The serine/threonine protein kinase mTOR forms two structurally and functionally distinct complexes TORC1 and

TORC2. TORC1, the Rapamycin sensitive branch of the mTOR pathway, consists of the scaffolding protein Raptor, FKBP12 (the molecular target of

Rapamycin) and three further subunits, mLST8, PRAS40 and Deptor. TORC1 activity is critically controlled by a small GTPase, Rheb, whose activity is

inhibited by a GTPase activating protein, TSC2 in complex with TSC1. Consequently, inactivation of the TSC1/2 complex by upstream signals

emerging from the PI3K/Akt axis and the Erk pathway activate TORC1. Many accessory molecules as well as growth factors signal via the PI3K/Akt

and Erk pathways and therefore inevitably activate TORC1. Akt can further promote TORC1 activity independent of TSC1/2 by phosphorylating

PRAS40, a negative regulator of TORC1 activity. Furthermore TORC1 can be activated via the WNT pathway, in a signalling cascade involving GSK3.

Energy deficiencies as well as low oxygen levels lead to the activation of AMPK, which phosphorylates and activates the TSC1/2 complex resulting in

TORC1 inhibition. Furthermore, TORC1 activity is dependent on sufficient levels of amino acids, in a process involving recently discovered Rag A–D

proteins and the Ragulator complex. Active TORC1 controls a variety of cellular processes ranging from autophagy, lipid synthesis and cell cycle

progression to mitochondrial biogenesis and protein synthesis. The latter is at least in part controlled by the TORC1 downstream targets 4E-BP1 and

S6K1. Abbreviations: mTOR, mammalian target of Rapamycin; TORC, mTOR complex; Raptor, regulatory-associated companion of mTOR; FKBP12,

FK506 binding protein 12; mLST8, mammalian lethal with Sec13 protein 8; PRAS40, prolin-rich Akt substrate 40kDa; Deptor, DEP-domain-containing

mTOR-interacting protein; Rheb, Ras homologue enriched in brain; TSC, tuberous sclerosis protein; PI3K, Phosphatidylinositol 3-kinase; Akt, AKT8

virus oncogene cellular homologue; Erk, Extracellular signal-regulated kinase; GSK3, Glycogen synthase kinase 3; AMPK, AMP-activated protein

kinase; Rag A–D, Ras-related GTP-binding-proteins A–D; (EIF)4E-BP1, Eukaryotic translation initiation factor 4E-binding protein 1; S6K1, (ribosomal)

S6 kinase 1.

A second key nutrient sensed by TORC1 is the avail-

ability of AA. This component of TORC1 activity

requires a number of recently identified proteins (the

Ras-related GTP-binding-proteins A–D; Rag A–D) that

control the intracellular localisation and thereby the acti-

vation of TORC1 (Figure 2) [11�,12�]. These Rag

proteins form heterodimers that interact with TORC1

Current Opinion in Immunology 2010, 22:655–661

via raptor when the RagB component of the RagB/D

complex is in its GTP-bound form. AA induce the loading

of RagB with GTP and thereby facilitate the interaction

of the Rag complex with TORC1. Recently an additional,

trimeric protein complex called Ragulator was identified

and shown to interact with TORC1 and the Rag

GTPases. Comprising the MAPKSP1, ROBLD3, and

www.sciencedirect.com

Page 3: mTOR signalling and metabolic regulation of T cell differentiation

mTOR signalling and metabolic regulation of T cell differentiation Peter, Waldmann and Cobbold 657

Figure 2

Putative model for the amino acid mediated regulation of TORC1 activation. Amino acid induced translocation of mTOR is thought to regulate its

activity, in a process that involves the Rag A–D and the trimeric Ragulator complex. Amino acids facilitate loading of Rag B with GTP, thereby

permitting the binding of the Rag proteins to Raptor in TORC1. In conjunction with the Ragulator complex the Rag proteins are essential for the

movement of TORC1 to Rhab7+ lysosmal compartments, in which the TORC1 activator Rheb resides. Abbreviations: Rag A–D, Ras-related GTP-

binding-proteins A–D; mTOR, mammalian target of Rapamycin; Rheb, Ras homologue enriched in brain

c11orf59 gene products, Ragulator was demonstrated to be

essential for the localisation of TORC1 and the Rag

complex to a Rab7+ lysosomal compartment containing

the crucial TORC1 activator Rheb [13�]. Despite recent

advances in the understanding of TORC1 activation by

AA, the molecular sensor of AA within or outside the cells

has yet to be identified, although recent studies in Dro-

sophila have revealed a potential role of proton-assisted

amino acid-transporters (PAT) in sensing and transmit-

ting an AA signal to TORC1 [14,15]. Considering the fact

that TORC1 can regulate the activity of AA transporters

this model also might allow TORC1 signals to feed back

on its sensors [16,17].

mTOR and T cell differentiationThe differentiation of T cells is a highly complex process

initiated by multiple signals emerging from the TCR,

cytokine receptors and co-stimulatory molecules. These

signals and the microenvironment in which a T cell is

activated direct its fate towards the ever growing number

of functionally distinct T cell subsets. By integrating the

nutritional and energetic status of the cells into the

signalling events induced by T cell activation in the local

environment, the mTOR complexes allow the cells to

www.sciencedirect.com

asses the wide spectrum of environmental stimuli as a

whole, so impacting the outcome of the differentiation

process.

It is widely accepted that the balance between effector T

cells and regulatory T cells is critical to the resulting

immune response. Studies performed by Delgoffe and

colleagues highlight the importance of mTOR in con-

trolling this delicate and important balance. Despite

displaying normal up-regulation of activation markers

and IL-2 production, T cells deficient in mTOR failed

to differentiate into inflammatory Th1, Th2 and Th17

subsets when activated. Furthermore the same activation

conditions led to the differentiation of Foxp3+ regulatory

T cells, indicating the importance of the mTOR pathway

in skewing CD4+ helper T cell fate [18��]. A recent study

utilizing Rictor knockout T cells demonstrates that the

differentiation of Th1 and Th2 is highly dependent on

TORC2 signalling leading to activation of Akt and PKC-u

respectively, whereas the development of Th17 cells

seems to be unimpaired by the lack of TORC2 activity

[19��]. Along these lines Kopf et al. confirmed that Rapa-

mycin prevented the differentiation of Th17 whereas

Foxp3 induction was promoted [20]. This role of the

Current Opinion in Immunology 2010, 22:655–661

Page 4: mTOR signalling and metabolic regulation of T cell differentiation

658 Immunogenetics and transplantation

mTOR pathway in promoting regulatory T cell differen-

tiation is underlined by experiments demonstrating that

components upstream of TORC1, such as PI3K and Akt

as well as TORC1 itself, negatively regulate Foxp3

expression [21�,22�]. Although these results suggest that

TORC1 inhibition is the crucial component allowing

Foxp3 to be induced, a number of caveats hinder a

definite conclusion. Although both studies indicate the

involvement of the mTOR pathway by demonstrating

that the observed effect is, for the most part, sensitive to

Rapamycin, neither group compared TORC1 or TORC2

activity. As the two complexes are thought to be recipro-

cally regulated and yet prolonged treatment with the

allosteric TORC1 inhibitor Rapamycin may result in

TORC2 inhibition, a straight forward analysis seems

difficult [23]. The interpretation of the findings of Hax-

hinasto et al. is further complicated given that Akt is also a

downstream target of TORC2, which might explain why

Rapamycin could not fully rescue the negative effect of

constitutively active Akt on Foxp3 induction [21�]. Along

these lines, Delgoffe et al. demonstrate that TORC2

inhibition might have a more important contribution to

the induction than previously assumed. Based on the

finding that T cells devoid of the TORC1 activator Rheb

failed to express Foxp3 under conditions that induced the

transcription factor in mTOR�/� T cells, the authors

postulated that TORC2 inhibition might be crucial in

facilitating Foxp3 induction by releasing Smad3 from an

inhibitory phosphorylation mediated via the TORC2

target Akt [18��].

We have demonstrated that TORC1 inhibition, by star-

ving T cells of essential AA, represents a physiological

mechanism that synergizes with TCR and TGFb sig-

nalling to induce Foxp3 expression. Furthermore, our

studies revealed that this depletion originates from the

induction of EAA consuming enzymes in APCs during

their recognition by regulatory T cells. The resulting

depletion of EAA then induces further Foxp3 during

antigen presentation to conventional T cells (Tconv),

converting them to Treg, which provides a mechanism

by which Foxp3+ Treg can mediate infectious tolerance

[24�]. Since EAA depletion is thought to specifically

inhibit TORC1 and not affect TORC2 activity, these

results seem to contradict the hypothesis that TORC2

inhibition is key to Foxp3 induction. These obvious

discrepancies might be explained by variations in the

quality and specificity of mTOR inhibition that is

caused by Rapamycin, EAA starvation and genetic

deletion. Furthermore the development of an ATP

competitive mTOR inhibitor (Torin1) has revealed

Rapamycin resistant TORC1 functions that might be

involved in regulating Foxp3 [25]. Therefore, the differ-

ential contribution of the TORCs to the induction of

Foxp3 is still unclear and downstream targets of the

TORC complexes involved in the induction have yet to

be identified. A possible mechanism that links mTOR

Current Opinion in Immunology 2010, 22:655–661

and Foxp3 expression was proposed by experiments

demonstrating the importance of the Foxo transcription

factors in the TGFb mediated induction of Foxp3 [26–28]. Harada and colleagues indicate that the PI3K-Akt-

mTOR axis might have a crucial role in controlling the

activity of the Foxo factors as inhibition of PI3K in T

cells lacking Cbl-b, a molecule known to reduce PI3K

activation, restored Foxp3 induction in these cells [26].

The activity of the Foxos is mainly controlled by their

translocation between nucleus and cytoplasm, which is

at least in part dependent on phosphorylation events

mediated by Akt [29]. Considering the accumulating

evidence that Akt and mTOR are important regulators

of Foxp3 expression and since PI3K inhibition inevita-

bly reduces Akt and mTOR activity, one can speculate

that these two downstream targets of PI3K modulate the

activity of Foxo factors to control Foxp3 expression.

Moreover, it will in due course be interesting to deter-

mine whether other conditions known to inhibit mTOR

such as glucose deprivation can also facilitate Foxp3

induction. Along these lines, Jurkat T cells seem to up-

regulate Foxp3 in a HIF-1a dependent manner when

cultured under hypoxic conditions known to activate

AMPK.

Further evidence for the importance of the mTOR path-

way in the induction and maintenance of Foxp3+ regu-

latory T cells in vivo comes from studies implicating

programmed death 1 protein (PD1). Ligation of PD1

during T cell activation in vitro was shown to inhibit

TORC1 and TORC2 and lead to an increase of Foxp3+

cells [30]. In addition to Foxp3 induction, several groups

have described that TORC1 inhibition by Rapamycin

favours Treg expansion over effector T cells (Teff) by

differentially regulating signalling and sensitivity to apop-

tosis [31–35]. It therefore seems likely that a combination

of inductive events and preferential expansion of Foxp3+

Treg contributes to the shift of T cell fate towards regu-

latory subsets.

T cell metabolism and anergyBesides the induction of regulatory T cells, T cell anergy

is thought to be another crucial tolerance inducing mech-

anism. The term anergy describes the secondary unre-

sponsiveness of T cells to a normally activating stimulus

via the TCR and CD28 with respect to cytokine pro-

duction and proliferation. This phenomenon was

ascribed to a lack of costimulation via CD28 during

the primary exposure to antigen resulting in suboptimal

activation of the Akt-mTOR pathway and incomplete

recruitment of transcription factors, particularly those

involved in IL-2 transcription. This leads to the induc-

tion of an anergic gene expression profile and failure of

the cell to enter the cell cycle [36]. Recently a number of

groups have extended this model by demonstrating that

signal 1 (TCR) and signal 2 (costimulation) are both

necessary to fully activate mTOR and to adjust the

www.sciencedirect.com

Page 5: mTOR signalling and metabolic regulation of T cell differentiation

mTOR signalling and metabolic regulation of T cell differentiation Peter, Waldmann and Cobbold 659

cellular metabolism in order to meet the demands that

cell division and differentiation impose on the cell.

Zheng and colleagues further demonstrated that anergy

induction, by signal 1 alone, not only results in metabo-

lically anergic cells but that nutrient availability sensed

by the mTOR pathway controls anergy induction [37�].The authors convincingly show that anergy is associated

with a failure to up-regulate nutrient transporters such as

CD71 (transferrin receptor; iron uptake), CD98 (amino

acid uptake) and Glut1 (glucose uptake) and an inability

to induce glycolytic pathways. The blockade of leucine,

glucose and energy metabolism using N-acetyl-leucine

amide (NALA), 2-deoxyglucose (2DG) and 5-aminoimi-

dazole-4-carboxamide ribonucleoside (AICAR) were

able to induce anergy even in the presence of sufficient

signal 1 plus 2.

Besides these striking effects of mTOR inhibition that

seem to explain the potent immunosuppressive effects of

Rapamycin in vivo, recent reports indicate that the bac-

terial metabolite can also promote the development of

CD8+ memory T cells [38�,39��,40]. Administration of

Rapamycin in vivo enhances not only the magnitude, but

also the quality, of highly functional memory CD8+ cells.

Araki et al. were able to prove that this effect of Rapa-

mycin was due to TORC1 inhibition in CD8+ T cells (and

not other immune cells) by performing a series of knock-

down experiments targeting TORC1 components raptor

and FKBP12 as well as TORC1 downstream targets S6K1

and eIF4E in CD8+ cells [38�]. Similarly, Rao and col-

leagues demonstrate that inhibition of TORC1 reverses

the IL-12 induced skewing of CD8+ cell differentiation

into effector rather than memory type cells by disruption

of persistent T-bet expression. The reduced levels of the

transcription factor T-bet augment the induction of yet

another transcription factor, Eomesodermin, that has

been shown to favour CD8+ memory precursor cell

generation [39,41]. Additionally, the pharmacological

activation of the negative TORC1 regulator AMPK by

the anti-diabetic drug metformin, was able to reverse the

defects in lipid metabolism and memory cell develop-

ment seen in TRAF6 deficient T cells. This study

elegantly highlights the connection between lipid

metabolism, growth factor signalling and the mTOR

pathway [40]. Further evidence for the role of mTOR

in regulating CD8+ T cell differentiation comes from

studies investigating the role of the Wnt pathway [42].

Wnt signalling, which is known to activate TORC1

signalling by preventing GSK3 mediated TSC1/2 phos-

phorylation [43], was demonstrated to arrest effector T

cell differentiation at the expense of increased CD8+

memory stem cells [42]. The remarkable difference in

the response of CD8+ and CD4+ T cells to mTOR

inhibition suggests that mTOR signalling might play a

different role in controlling cellular fate in these two

subsets. Araki et al. speculate that this discrepancy could

be explained by the incomplete inhibition of mTOR

www.sciencedirect.com

signalling by Rapamycin compared to genetic deletion

[44].

mTOR signalling and T cell traffickingWhile the majority of studies have focused on mTOR

inhibition and its role in the induction of Foxp3 in per-

ipheral T cells, Liu and colleagues identified lysopho-

spholipid sphingosine 1-phosphate (S1P) as novel

regulator of the Akt-mTOR axis in T cells [45]. The

S1P receptor is known to be an important regulator of T

cell trafficking and exit from the lymph nodes, and is the

inhibitory target of the novel immunosuppressive agent

FTY720 [46]. Interestingly, by activating mTOR S1P re-

ceptor signalling inhibits Foxp3 induction and suppressive

acitivity in thymocytes in a Rapamycin sensitive manner.

Further, Liu et al. demonstrated differential control of S1P

receptor expression upon stimulation of Tconv (abrupt

decrease) opposed to Treg (slower decrease). With the Treg

being released from S1P induced mTOR activity later,

Tconv can initiate the immune response relatively unim-

peded [45].

Sinclair et al. have investigated the effects of the PI3K-

mTOR axis on the expression of CD62L and CCR7, two

other important regulators of lymphocyte trafficking.

TORC1 inhibition by Rapamycin prevented IL-2

induced down-regulation of CD62L mRNA and CCR7

surface expression on activated CD8+ T cells in vitro, so

enabling these cells to retain their homing capacity for

secondary lymphoid organs in vivo [47�]. Similar to the

effects of FTY720, this sequestration of CD8+ effector T

cells away from sites of inflammation might also contrib-

ute to the immunosuppressive effects of Rapamycin invivo. Studies investigating the chemotactic response of

PBMC derived CD4+ cells treated with Rapamycin also

demonstrate that TORC1 signalling is necessary for

CCR5 mediated chemotaxis. In this case TORC1 activity

allows translation of a subset of mRNAs necessary for

chemotaxis (including cyclin D1 and MMP-9) through its

effect on the downstream target 4E-BP1 [48].

ConclusionsIn summary, T cells link instructive signals emerging

from the local microenvironment to their nutritional and

energetic status primarily by using a common route, via

mTOR, to control the functional fate of the cell. This

might represent a mechanism that allows lymphocytes

not only to differentiate according to their instructed fate

but also to fine-tune their response according to their

metabolic competence. The activation status of mTOR in

T cells has been demonstrated to crucially influence the

balance of inflammatory and tolerogenic T cell subset

differentiation. Understanding how the mTOR com-

plexes regulate T cell fate in greater detail opens up

the prospect of developing novel therapeutic regimens

(based on mTOR and other metabolic inhibitors) to treat

diseases in which this balance needs to be modified.

Current Opinion in Immunology 2010, 22:655–661

Page 6: mTOR signalling and metabolic regulation of T cell differentiation

660 Immunogenetics and transplantation

AcknowledgementsWe thank the Medical Research Council and the Edward Penley AbrahamTrust for their support.

References and recommended readingPapers of particular interest published within the period of review havebeen highlighted as:

� of special interest

�� of outstanding interest

1. Buttgereit F, Brand MD, Muller M: ConA induced changes inenergy metabolism of rat thymocytes. Biosci Rep 1992,12:109-114.

2. Ramanathan A, Schreiber SL: Direct control of mitochondrialfunction by mTOR. Proc Natl Acad Sci U S A 2009,106:22229-22232.

3. Schieke SM, Phillips D, McCoy JP Jr, Aponte AM, Shen RF,Balaban RS, Finkel T: The mammalian target of rapamycin(mTOR) pathway regulates mitochondrial oxygenconsumption and oxidative capacity. J Biol Chem 2006,281:27643-27652.

4. Hurley RL, Anderson KA, Franzone JM, Kemp BE, Means AR,Witters LA: The Ca2+/calmodulin-dependent protein kinasekinases are AMP-activated protein kinase kinases. J Biol Chem2005, 280:29060-29066.

5. Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M,Johnstone SR, Carlson M, Carling D: Ca2+/calmodulin-dependent protein kinase kinase-beta acts upstream ofAMP-activated protein kinase in mammalian cells.Cell Metab 2005, 2:21-33.

6. Jacobs SR, Herman CE, Maciver NJ, Wofford JA, Wieman HL,Hammen JJ, Rathmell JC: Glucose uptake is limiting in T cellactivation and requires CD28-mediated Akt-dependent andindependent pathways. J Immunol 2008, 180:4476-4486.

7. Wieman HL, Wofford JA, Rathmell JC: Cytokine stimulationpromotes glucose uptake via phosphatidylinositol-3 kinase/Akt regulation of Glut1 activity and trafficking. Mol Biol Cell2007, 18:1437-1446.

8. Edinger AL, Thompson CB: Akt maintains cell size and survivalby increasing mTOR-dependent nutrient uptake. Mol Biol Cell2002, 13:2276-2288.

9. Hay N, Sonenberg N: Upstream and downstream of mTOR.Genes Dev 2004, 18:1926-1945.

10.�

Laplante M: Sabatini DM: mTOR signaling at a glance. J Cell Sci2009, 122:3589-3594.

Excellent and concise review on upstream signals feeding into the mTORpathway and the downstream effectors of the two mTOR complexes.

11.�

Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC,Bar-Peled L, Sabatini DM: The Rag GTPases Bind Raptor andMediate Amino Acid Signaling to mTORC1. Science 2008,320:1496-1501.

12.�

Kim E, Goraksha-Hicks P, Li L, Neufeld TP, Guan K-L: Regulationof TORC1 by Rag GTPases in nutrient response. Nat Cell Biol2008, 10:935-945.

13.�

Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, SabatiniDM: Ragulator–Rag complex targets mTORC1 to thelysosomal surface and is necessary for its activation by aminoacids. Cell 2010, 141:290-303.

This series of papers of three papers has highlighted the role of the Rag–Ragulator complex in the amino acid dependent translocation of TORC1,enabling the activation of the mTOR complex by amino acids.

14. Goberdhan DC, Meredith D, Boyd CA, Wilson C: PAT-relatedamino acid transporters regulate growth via a novelmechanism that does not require bulk transport of aminoacids. Development 2005, 132:2365-2375.

15. Heublein S, Kazi S, Ogmundsdottir MH, Attwood EV, Kala S,Boyd CA, Wilson C, Goberdhan DC: Proton-assisted amino-acidtransporters are conserved regulators of proliferation and

Current Opinion in Immunology 2010, 22:655–661

amino-acid-dependent mTORC1 activation. Oncogene 2010doi: 10.1038/onc. 2010.177.

16. Reynolds B, Laynes R, Ogmundsdottir MH, Boyd CA,Goberdhan DC: Amino acid transporters and nutrient-sensingmechanisms: new targets for treating insulin-linkeddisorders? Biochem Soc Trans 2007, 35:1215-1217.

17. Goberdhan DC, Ogmundsdottir MH, Kazi S, Reynolds B,Visvalingam SM, Wilson C, Boyd CA: Amino acid sensing andmTOR regulation: inside or out? Biochem Soc Trans 2009,37:248-252.

18.��

Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B,Worley PF, Kozma SC, Powell JD: The mTOR kinasedifferentially regulates effector and regulatory T cell lineagecommitment. Immunity 2009, 30:832-844.

Using CD4+ T cell specific knock outs of mTOR and Rheb, the authorshighlight the importance mTOR in regulating the differentiation of inflam-matory and regulatory T cell subsets. The fact that mTOR deficient T cellsstimulated in the absence of TGFb turn on Foxp3, whereas Rheb deficientcells failed to do so, indicates that the role of TORC2 might be moreimportant in regulating Foxp3 expression than TORC1.

19.��

Keunwook Lee PG, Srdjan Dragovic, Charles Spencer, SebastianJoyce, Nigel Killeen, Mark A, Magnuson, Mark Boothby::Mammalian target of rapamycin protein complex 2 regulatesdifferentiation of Th1 and Th2 cell subsets via distinctsignaling pathways. Immunity 2010, 32:743-753.

Using CD4+ T cell specific knock outs of Rictor, the authors highlight theimportance TORC2 signalling for the differentiation of Th1 and Th2 cells.They further demonstrate that TORC2 regulates these differentiationprocesses via its downstream targets Akt and PKCu.

20. Kopf H, de la Rosa GM, Howard OM, Chen X: Rapamycin inhibitsdifferentiation of Th17 cells and promotes generation ofFoxP3+ T regulatory cells. Int Immunopharmacol 2007,7:1819-1824.

21.�

Haxhinasto S, Mathis D, Benoist C: The AKT-mTOR axisregulates de novo differentiation of CD4+Foxp3+ cells.J Exp Med 2008, 205:565-574.

This paper along with Sauer et al. [22] was amongst the first to provideevidence that Akt and the mTOR pathway regulate Foxp3 induction in Tcells. The authors demonstrate that expression of a constitutively activeAkt prevents TGFb mediated Foxp3 induction and that this effect can befor the most part reversed by Rapamycin.

22.�

Sauer S, Bruno L, Hertweck A, Finlay D, Leleu M, Spivakov M,Knight ZA, Cobb BS, Cantrell D, O’Connor E et al.: T cell receptorsignaling controls Foxp3 expression via PI3K, Akt, and mTOR.Proc Natl Acad Sci 2008, 105:7797-7802.

The authors demonstrate that premature determination of TCR signallingfollowed by PI3K and mTOR inhibition confers Foxp3 expression in T cellsand thymocytes, thereby establishing the link between mTOR and Foxp3expression.

23. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley AF,Markhard AL, Sabatini DM: Prolonged rapamycin treatmentinhibits mTORC2 assembly and Akt/PKB. Mol Cell 2006,22:159-168.

24.�

Cobbold SP, Adams E, Farquhar CA, Nolan KF, Howie D,Lui KO, Fairchild PJ, Mellor AL, Ron D, Waldmann H: Infectioustolerance via the consumption of essential amino acidsand mTOR signaling. Proc Natl Acad Sci U S A 2009,106:12055-12060.

Our own work has established that inhibition of TORC1 by EAA starvationcan lead to the induction of Foxp3 and could represent a mechanism thatexplains infectious tolerance.

25. Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y,Reichling LJ, Sim T, Sabatini DM, Gray NS: An ATP-competitivemammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J Biol Chem 2009,284:8023-8032.

26. Harada Y, Elly C, Ying G, Paik JH, Depinho RA, Liu YC:Transcription factors Foxo3a and Foxo1 couple the E3 ligaseCbl-b to the induction of Foxp3 expression in inducedregulatory T cells. J Exp Med 2010, 207:1381-1391.

27. Ouyang W, Beckett O, Ma Q, Paik JH, DePinho RA, Li MO: Foxoproteins cooperatively control the differentiation of Foxp3+regulatory T cells. Nat Immunol 2010, 11:618-627.

www.sciencedirect.com

Page 7: mTOR signalling and metabolic regulation of T cell differentiation

mTOR signalling and metabolic regulation of T cell differentiation Peter, Waldmann and Cobbold 661

28. Merkenschlager M, von Boehmer H: PI3 kinase signalling blocksFoxp3 expression by sequestering Foxo factors. J Exp Med2010, 207:1347-1350.

29. Hedrick SM: The cunning little vixen: Foxo and the cycle of lifeand death. Nat Immunol 2009, 10:1057-1063.

30. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ,Kuchroo VK, Sharpe AH: PD-L1 regulates the development,maintenance, and function of induced regulatory T cells.J Exp Med 2009, 206:3015-3029.

31. Basu S, Golovina T, Mikheeva T, June CH, Riley JL: Cutting edge:Foxp3-mediated induction of pim 2 allows human T regulatorycells to preferentially expand in rapamycin. J Immunol 2008,180:5794-5798.

32. Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J,Kaupper T, Roncarolo MG: Rapamycin promotes expansion offunctional CD4+CD25+FOXP3+ regulatory T cells of bothhealthy subjects and type 1 diabetic patients. J Immunol 2006,177:8338-8347.

33. Battaglia M, Stabilini A, Roncarolo MG: Rapamycin selectivelyexpands CD4+CD25+FoxP3+ regulatory T cells. Blood 2005,105:4743-4748.

34. Strauss L, Czystowska M, Szajnik M, Mandapathil M,Whiteside TL: Differential responses of human regulatoryT cells (Treg) and effector T cells to rapamycin. PLoS One 2009,4:e5994.

35. Strauss L, Whiteside TL, Knights A, Bergmann C, Knuth A,Zippelius A: Selective survival of naturally occurring humanCD4+CD25+Foxp3+ regulatory T cells cultured withrapamycin. J Immunol 2007, 178:320-329.

36. Wells AD: New insights into the molecular basis of T cellanergy: anergy factors, avoidance sensors, and epigeneticimprinting. J Immunol 2009, 182:7331-7341.

37.�

Zheng Y, Delgoffe GM, Meyer CF, Chan W, Powell JD: Anergic Tcells are metabolically anergic. J Immunol 2009, 183:6095-6101.

This study expanded the widely accepted two-signal model of T cellactivation by emphasizing the importance of the up-regulation of themetabolic machinery by these signals. The experiments convincinglydemonstrate that anergic T cells are metabolically anergic and thatblockade of metabolic pathways can render T cells anergic.

38.�

Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA,Bachmann MF, Larsen CP, Ahmed R: mTOR regulates memoryCD8 T-cell differentiation. Nature 2009, 460:108-112.

The authors demonstrate that administration of Rapamycin in LCMVinfected mice exerts immunostimulatory effects by enhancing the quan-tity and quality of CD8+ memory T cells. Furthermore, they establish that

www.sciencedirect.com

this is a T cell intrinsic effect by demonstrating that knock down of mTORpathway components can induce similar differentiation patterns.

39.��

Rao RR, Li Q, Odunsi K, Shrikant PA: The mTOR kinasedetermines effector versus memory CD8+ T cell fate byregulating the expression of transcription factors T-bet andEomesodermin. Immunity 2010, 32:67-78.

Rao et al. provide evidence that TORC1 inhibition by Rapamycin pro-motes memory CD8+ T cell differentiation by blocking IL-12 inducedsustained T-bet expression and promoting Eomesodermin expression.

40. Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS,Jones RG, Choi Y: Enhancing CD8 T-cell memory bymodulating fatty acid metabolism. Nature 2009, 460:103-107.

41. Intlekofer AM, Takemoto N, Wherry EJ, Longworth SA,Northrup JT, Palanivel VR, Mullen AC, Gasink CR, Kaech SM,Miller JD et al.: Effector and memory CD8+ T cell fate coupledby T-bet and eomesodermin. Nat Immunol 2005, 6:1236-1244.

42. Gattinoni L, Zhong XS, Palmer DC, Ji Y, Hinrichs CS, Yu Z,Wrzesinski C, Boni A, Cassard L, Garvin LM et al.: Wnt signalingarrests effector T cell differentiation and generates CD8+memory stem cells. Nat Med 2009, 15:808-813.

43. Inoki K, Ouyang H, Zhu T, Lindvall C, Wang Y, Zhang X, Yang Q,Bennett C, Harada Y, Stankunas K et al.: TSC2 integratesWnt and energy signals via a coordinated phosphorylationby AMPK and GSK3 to regulate cell growth. Cell 2006,126:955-968.

44. Araki K, Youngblood B, Ahmed R: The role of mTOR in memoryCD8 T-cell differentiation. Immunol Rev 2010, 235:234-243.

45. Liu G, Burns S, Huang G, Boyd K, Proia RL, Flavell RA, Chi H:The receptor S1P1 overrides regulatory T cell-mediatedimmune suppression through Akt-mTOR. Nat Immunol 2009,10:769-777.

46. Kunzendorf U, Ziegler E, Kabelitz D: FTY720 – the first compoundof a new promising class of immunosuppressive drugs.Nephrol Dial Transplant 2004, 19:1677-1681.

47.�

Sinclair LV, Finlay D, Feijoo C, Cornish GH, Gray A, Ager A,Okkenhaug K, Hagenbeek TJ, Spits H, Cantrell DA:Phosphatidylinositol-3-OH kinase and nutrient-sensing mTORpathways control T lymphocyte trafficking. Nat Immunol 2008,9:513-521.

The studies performed by Sinclair et al. revealed that inhibition of thePI3K-mTOR axis can prevent activation and cytokine induced down-regulation of CD62L and CCR7 surface expression.

48. Murooka TT, Rahbar R, Platanias LC, Fish EN: CCL5-mediatedT-cell chemotaxis involves the initiation of mRNA translationthrough mTOR/4E-BP1. Blood 2008, 111:4892-4901.

Current Opinion in Immunology 2010, 22:655–661