Signaling via the Akt/mammalian target of rapamycin pathway influences CD4+ T cell differentiation; low levels favor regulatory T cell induction and high levels favor Th induction. Although the lipid phosphatase phosphatase and tensin homolog (PTEN) suppresses Akt activity, the control of PTEN activity is poorly studied in T cells. In this study, we identify multiple mechanisms that regulate PTEN expression. During Th induction, PTEN function is suppressed via lower mRNA levels, lower protein levels, and an increase in C-terminal phosphorylation. Conversely, during regulatory T cell induction, PTEN function is maintained through the stabilization of PTEN mRNA transcription and sustained protein levels. We demonstrate that differential Akt/mammalian target of rapamycin signaling regulates PTEN transcription via the FoxO1 transcription factor. A mathematical model that includes multiple modes of PTEN regulation recapitulates our experimental findings and demonstrates how several feedback loops determine differentiation outcomes. Collectively, this work provides novel mechanistic insights into how differential regulation of PTEN controls alternate CD4+ T cell fate outcomes.

Ligand binding to the TCR induces a cascade of signaling events that culminates in gene transcription, T cell proliferation, and differentiation. We (1) and others (2, 3) have demonstrated that Foxp3-expressing regulatory T cells (Tregs) are induced when naive T cells are exposed to low Ag doses, and it has been shown that Tregs expanded following stimulation with low-dose Ag prevent autoimmune diabetes in vivo (4, 5). Treg induction is inversely correlated with the degree of signaling via the Akt/mammalian target of rapamycin (mTOR) pathway (1, 6, 7).

We developed a mathematical model to determine how TCR signal strength contributes to Treg induction (8). Regulation of the phosphatase and tensin homolog (PTEN) protein emerged as a critical control point in Treg induction (8). Expression and function of PTEN are tightly regulated via transcriptional control and posttranslational modifications (PTMs) (9, 10). Naive T cells express high levels of PTEN and following T cell activation PTEN protein disappears (11), but the mechanism for this downregulation is not known. Tregs are known to express higher levels of PTEN (12) and this changes the signaling pathways triggered following IL-2 activation (13).

In this study, we identify multiple modes of PTEN regulation that affect CD4+ T cell differentiation. Th induction is characterized by reduced PTEN transcription, degradation of PTEN protein, and higher C-terminal phosphorylation. However, in Treg induction, PTEN mRNA and protein levels are maintained. Additionally, we identify a loop connecting Akt/mTOR signaling to the regulation of PTEN via FoxO1 as a novel mechanism for regulating CD4+ T cell differentiation. Taken together, these findings define how TCR signal strength differentially tunes PTEN function to drive Th versus Treg differentiation.

C57BL/6 mice were obtained from The Jackson Laboratory. All mice were housed in a specific pathogen-free facility at the University of Pittsburgh and treated under Institutional Animal Care and Use Committee–approved guidelines in accordance with approved protocols.

CD4+ T cells were isolated from C57BL/6 spleens using a CD4+ negative selection kit (Miltenyi Biotec). In some experiments CD25+ T cells were removed using CD25 microbeads. T cell activation assays were performed with low-dose (0.25 μg/ml) or high-dose (1 μg/ml) plate-bound anti-CD3 mAb in the presence of soluble anti-CD28 mAb (1 μg/ml). For inhibition studies, T cells were treated for 1 h with the following drugs prior to stimulation: caspase inhibitor (ZVAD, 80 μM), PTEN inhibitor (SF1670, 10 μM), and Akt inhibitor (Akti1/2, 10 μM).

Activated CD4+ T cells were stained with the following mAbs: anti–CD3-allophycocyanin-eFluor 780, anti–CD4-allophycocyanin, anti–CD25-PE, anti–Foxp3-Pacific Blue (eBioscience), and anti–pS6 (Ser235)-FITC (Cell Signaling Technology) using buffers from eBioscience. The stained cells were analyzed on an LSR II flow cytometer and data were analyzed with the FlowJo software package.

Western blotting was performed using the following Abs from Cell Signaling Technology: β-actin, PTEN (138G6), phospho-PTEN (Ser380/Thr382/383), FoxO1 (C29H4), phospho-FoxO1 (Thr24), and ubiquitin.

Total RNA was isolated from CD4+ T cells the Aurum total RNA mini kit (Bio-Rad). Quantitative RT-PCR (qPCR) was performed on a StepOnePlus real-time PCR system using SYBR Green (Applied Biosystems). The primer pair used was 5′-ACACCGCCAAATTTAACTGC-3′ and 5′-TACACCAGTCCGTCCCTTTC-3′.

Chromatin immunoprecipitation (ChIP) for FoxO1 was performed using the ChIP-IT high sensitivity kit (Active Motif) and an anti-FoxO1 Ab (Cell Signaling Technology). qPCR was used to quantify the amount of the PTEN 5 kb upstream element (primer pair used: 5′-CCTTGCATCAGCCATTAAAGAAA-3′ and 5′-CTGCTGTACCAGGCTATATGTG-3′) associated with FoxO1.

A murine FoxO1 small interfering RNA (siRNA) kit (Origene) was used to knock down FoxO1 expression. The siRNAs were introduced into isolated CD4+ T cells using a standard protocol (Lonza Nucleofector kit for mouse T cells). Western blot analysis for FoxO1, PTEN, and actin was performed after 48 h of incubation.

Nuclear and cytoplasmic fractions of activated T cells were generated using the NE-PER nuclear and cytoplasmic kit (Thermo Scientific) following the manufacturer’s instructions.

Simulations of PTEN levels in cells destined to become Th cells or Tregs were performed as described (8). A minimized version was specified as a Boolean model using the logical rules shown in Supplemental Fig. 2. For the simulations performed in this study, the logical rules were translated into the BioNetGen rule-based modeling language using an automated tool called Boolean2BNGL included in the BioNetGen package (14). The simulations, performed according to the general asynchronous update scheme (15), used a modified version of Gillespie’s (16) direct stochastic simulation algorithm.

All statistical calculations were performed in the GraphPad Prism 6 software package. Except where indicated, all analyses used the two-way ANOVA analysis with a Bonferroni postanalysis correction. Some analyses used the one-way ANOVA or Student t test. A p value <0.05 was considered significant.

Our computational model of Th/Treg differentiation (8) predicted that, in developing Th cells, PTEN rapidly decreased and remained absent, whereas PTEN was only transiently downmodulated in cells destined to become Tregs (Fig. 1A). To test this prediction, we performed T cell activation assays with low and high doses of anti-CD3 mAb, which drive Treg and Th differentiation, respectively (Supplemental Fig. 1A, 1B) (1, 4, 17). Western blot analysis of PTEN in activated CD4+ T cells confirmed the model predictions, and PTEN levels were markedly and persistently reduced with high-dose stimulation, but only transiently reduced with low-dose stimulation (Fig. 1B, 1C). Similar results were obtained with CD25-depleted CD4+ T cells (Supplemental Fig. 1C, 1D)

FIGURE 1.

TCR signal strength regulates PTEN protein levels. (A) PTEN activity in simulations of CD4 T cell differentiation leading to Th (solid line) and Treg fate (dotted line) reported as percentage of simulation trajectories. (B) PTEN protein levels were tracked by Western blotting of activated CD4+ T cells under low-dose (upper panel) and high-dose (bottom panel) conditions. (C) Actin-normalized PTEN values were plotted. (D and E) CD4+ T cells were activated under low-dose TCR stimulation in the presence (dashed line) or absence (solid line) of a PTEN inhibitor (SF1670). The development of Th cells (CD25+Foxp3) (D) or Tregs (CD25+Foxp3+) (E) was assessed by flow cytometry on gated CD3+CD4+ T cells. Results in (C)–(E) represent the mean ± SEM from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 1.

TCR signal strength regulates PTEN protein levels. (A) PTEN activity in simulations of CD4 T cell differentiation leading to Th (solid line) and Treg fate (dotted line) reported as percentage of simulation trajectories. (B) PTEN protein levels were tracked by Western blotting of activated CD4+ T cells under low-dose (upper panel) and high-dose (bottom panel) conditions. (C) Actin-normalized PTEN values were plotted. (D and E) CD4+ T cells were activated under low-dose TCR stimulation in the presence (dashed line) or absence (solid line) of a PTEN inhibitor (SF1670). The development of Th cells (CD25+Foxp3) (D) or Tregs (CD25+Foxp3+) (E) was assessed by flow cytometry on gated CD3+CD4+ T cells. Results in (C)–(E) represent the mean ± SEM from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

To determine whether PTEN activity regulated CD4+ T cell differentiation, activation assays were performed in the presence of a PTEN inhibitor. PTEN inhibition of T cells stimulated with high-dose anti-CD3 resulted in the development of Th cells and enhancement of Akt/mTOR signaling as expected (Supplemental Fig. 1E). However, under low-dose stimulation, PTEN inhibition enhanced Th (Fig. 1D) and suppressed Treg (Fig. 1E) induction. These results are consistent with recent studies using PTEN short hairpin RNA, showing decreased Treg induction and increased pS6 in cells with PTEN knockdown (13, 18).

TCR signaling perturbs PTEN protein levels (Fig. 1) (13, 18), but the molecular mechanisms that regulate PTEN protein levels in T cells are unexplored. The rapidity of PTEN degradation in Th and Treg conditions suggests that PTMs contribute to the regulation of PTEN turnover. In other cell types, polyubiquitination of PTEN by the E3 ubiquitin ligase Nedd4 leads to proteosomal degradation (19). In Treg conditions, PTEN was not ubiquitinated (Fig. 2A), whereas in Th conditions, PTEN was extensively ubiquitinated (Fig. 2B).

FIGURE 2.

PTEN is regulated by ubiquitination, phosphorylation, and caspase degradation. PTEN was immunoprecipitated from CD4+ T cells activated under low (A) or high (B) TCR stimulation and probed for total ubiquitin (UB). (C) T cells were activated with low dose in the presence or absence of a pan-caspase inhibitor (ZVAD), and PTEN levels were determined by Western blot. (D) Phosphorylation of the PTEN C terminus was assessed by Western blotting under low- and high-dose TCR stimulation [quantitated in (E)]. Results represent the mean ± SEM from three independent experiments. ***p < 0.001.

FIGURE 2.

PTEN is regulated by ubiquitination, phosphorylation, and caspase degradation. PTEN was immunoprecipitated from CD4+ T cells activated under low (A) or high (B) TCR stimulation and probed for total ubiquitin (UB). (C) T cells were activated with low dose in the presence or absence of a pan-caspase inhibitor (ZVAD), and PTEN levels were determined by Western blot. (D) Phosphorylation of the PTEN C terminus was assessed by Western blotting under low- and high-dose TCR stimulation [quantitated in (E)]. Results represent the mean ± SEM from three independent experiments. ***p < 0.001.

Close modal

During Treg induction, PTEN is degraded soon after T cell activation, yet PTEN is not ubiquitinated (Figs. 1, 2A), suggesting a different mechanism for the early degradation of PTEN. Multiple caspases, including caspase-3 and caspase-9, are known to cleave PTEN (20), and caspases function in TCR signaling (21, 22). Pan-caspase inhibition with the inhibitor ZVAD blocked degradation of PTEN under Treg conditions (Fig. 2C), demonstrating that caspase-mediated degradation of PTEN occurs during CD4+ T cell activation.

Constitutive phosphorylation of the C terminus of PTEN suppresses its enzymatic activity (23, 24), and casein kinase 2 has been shown to phosphorylate these sites in T cells (25, 26). We explored the phosphorylation state of PTEN during T cell differentiation. PTEN was phosphorylated at a cluster of sites, including Ser380, Thr382, and Thr383 during both Treg and Th induction (Fig. 2D), but relative levels of PTEN phosphorylation were significantly higher during Th induction (Fig. 2E). To our knowledge, these results are the first demonstration in T cells that levels of PTEN and enzymatic activity are controlled by multiple PTMs, which are differentially regulated by TCR signal strength.

High levels of TCR signaling have been correlated with a reduction in PTEN mRNA (13, 18). Therefore, we followed the expression of PTEN mRNA during Th and Treg induction. PTEN mRNA decreased in both Treg and Th activation conditions (Fig. 3A); however, whereas ∼20% of PTEN mRNA remained under Treg conditions, little PTEN mRNA was detected under Th conditions.

FIGURE 3.

FoxO1 regulates PTEN transcription. (A) PTEN mRNA levels were measured during Treg (black bars) and Th (hashed bars) induction. SDs were determined from three independent experiments. (B) ChIP experiments were performed to measure the association of FoxO1 and a 5 kb region upstream of the PTEN promoter at time = 0 (T = 0) and after 12 h of stimulation under Th (High) and Treg (Low) conditions. Results were normalized to unstimulated T cells (T = 0). A one-way ANOVA was performed. (C) Western blot analysis of PTEN, FoxO1, and actin protein levels in CD4 T cells treated with 3 FoxO1 siRNAs (A, B, and C) and scrambled control (S) for 48 h. (D) Total levels of FoxO1 and phospho-Thr24 FoxO1 were monitored by Western blotting. (E) Cellular localization of FoxO1 during low- and high-dose TCR stimulation determined by biochemically isolating nuclear (N) and cytoplasmic (C) fractions and Western blotting for FoxO1. Anti-actin Ab identifies the cytoplasmic fraction. (F) A reduced logical model of PTEN and Akt dynamics was developed (left panel). Two key feedback loops can be identified: negative feedback, in which Akt inhibits mTORC2 activity (top right panel), and positive feedback, in which Akt inactivates FoxO1 (bottom right panel). (G) Average levels of total PTEN (far left panel), active Akt (left panel), and nuclear FoxO1 (right panel) from the reduced model, and average levels of Foxp3 (far right panel) from the full model with inclusion of new regulatory elements from the reduced model for no TCR stimulation (thick solid line), low TCR stimulation (thin solid line), and high stimulation (dashed line) as a function of simulated time computed from 10,000 simulated trajectories. **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 3.

FoxO1 regulates PTEN transcription. (A) PTEN mRNA levels were measured during Treg (black bars) and Th (hashed bars) induction. SDs were determined from three independent experiments. (B) ChIP experiments were performed to measure the association of FoxO1 and a 5 kb region upstream of the PTEN promoter at time = 0 (T = 0) and after 12 h of stimulation under Th (High) and Treg (Low) conditions. Results were normalized to unstimulated T cells (T = 0). A one-way ANOVA was performed. (C) Western blot analysis of PTEN, FoxO1, and actin protein levels in CD4 T cells treated with 3 FoxO1 siRNAs (A, B, and C) and scrambled control (S) for 48 h. (D) Total levels of FoxO1 and phospho-Thr24 FoxO1 were monitored by Western blotting. (E) Cellular localization of FoxO1 during low- and high-dose TCR stimulation determined by biochemically isolating nuclear (N) and cytoplasmic (C) fractions and Western blotting for FoxO1. Anti-actin Ab identifies the cytoplasmic fraction. (F) A reduced logical model of PTEN and Akt dynamics was developed (left panel). Two key feedback loops can be identified: negative feedback, in which Akt inhibits mTORC2 activity (top right panel), and positive feedback, in which Akt inactivates FoxO1 (bottom right panel). (G) Average levels of total PTEN (far left panel), active Akt (left panel), and nuclear FoxO1 (right panel) from the reduced model, and average levels of Foxp3 (far right panel) from the full model with inclusion of new regulatory elements from the reduced model for no TCR stimulation (thick solid line), low TCR stimulation (thin solid line), and high stimulation (dashed line) as a function of simulated time computed from 10,000 simulated trajectories. **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

The mechanisms used to maintain PTEN mRNA levels under Treg induction conditions are not known. In our T cell differentiation model (8), the recovery of PTEN in Treg conditions arises from a link coupling Foxp3 to PTEN. However, recent data implicate the transcription factor FoxO1 in the induction of Treg, and phosphorylation of FoxO1 by Akt was established as a mechanism by which Akt/mTOR signaling inhibits Treg differentiation (27).

Previous ChIP sequencing data identified a FoxO1 binding site 5 kb upstream of the PTEN promoter (28), and we hypothesized that FoxO1 might be responsible for maintenance of PTEN transcription in Treg conditions. We performed ChIP assays to determine whether FoxO1 bound to this region. In both resting CD4+ T cells and under Treg conditions, FoxO1 bound the 5 kb upstream element, but it did not exhibit significant binding under Th conditions (Fig. 3B). siRNA knockdown of FoxO1 resulted in a concomitant decrease in PTEN protein levels, consistent with a positive role for FoxO1 in the regulation of PTEN levels (Fig. 3C).

Previous work demonstrated that unphosphorylated FoxO1 is localized in the nucleus and that Akt-mediated phosphorylation of Thr24 drives FoxO1 into the cytoplasm (29). We found that FoxO1 remained unphosphorylated under Treg conditions, but it was rapidly phosphorylated under Th conditions (Fig. 3D). As expected, FoxO1 remained in the nucleus in resting CD4+ T cells and under Treg conditions, but it was largely in the cytoplasm following high-dose stimulation (Fig. 3E), consistent with our observations on regulation of PTEN transcription.

Using components and interactions from our previous model of T cell differentiation (8), we developed a more limited model focusing on the regulation of PTEN, Akt, and FoxO1 (Fig. 3F, left). Simulations exhibited rapid and sustained loss of PTEN (Fig. 3G) at high Ag dose, as well as transient and incomplete loss of PTEN at low dose, in agreement with experimental findings. Under both conditions there was a rapid initial increase in Akt activity (Fig. 3G), sustained by positive feedback involving the loss of nuclear FoxO1 (Fig. 3F, bottom right). At high dose, the activation of casein kinase 2 blocked PTEN activity and sustained Akt activation. At low dose, PTEN activity rebounded as negative feedback through mTOR complex 2 (mTORC2)–decreased Akt activity (Fig. 3F, top right), increasing nuclear FoxO1 and, consequently, PTEN transcription. PTEN activity then further suppressed Akt, permitting full restoration of PTEN expression. Insertion of our reduced model back into our previous model of T cell differentiation resulted in sustained induction of Foxp3 in ∼70% of cells at low dose and only transient induction of Foxp3 at high dose (Fig. 3G), in agreement with results from the previous model and experiments (8).

Transient suppression of PTEN at low dose arises from the inclusion of two regulatory mechanisms in the model: the requirement for MEK1 to activate PTEN (30), and inhibition of mTORC2 by Akt (31). Prior to MEK1 activation by TCR (32), PTEN is inactive, allowing the transient induction of Akt, even at low dose, which may be necessary for T cell activation and proliferation regardless of differentiation outcome. Subsequent activation of PTEN by MEK1 then inhibits Akt in concert with the negative feedback provided by indirect Akt inhibition of mTORC2 (31).

To further elucidate the role of Akt in the regulation of FoxO1, we treated CD4+ T cells with an Akt inhibitor (Akti1/2) simultaneously with activation by a high dose of stimulation. The efficacy of the Akt inhibitor was confirmed by a reduction in pS6 expression (Fig. 4A). Under high TCR stimulation alone, we observed sustained phosphorylation of FoxO1 (Fig. 4B) and cytoplasmic localization of FoxO1 at 12 h (Fig. 4C). We also observed loss of FoxO1 binding to the 5 kb element upstream of the PTEN promoter (Fig. 4D) and loss of PTEN mRNA expression (Fig. 4E). Addition of the Akt inhibitor, alternatively, resulted in only transient phosphorylation of FoxO1 (Fig. 4B), which remained in the nucleus at 12 h (Fig. 4C) and was accompanied by sustained binding of FoxO1 upstream of the PTEN promoter and significantly higher PTEN mRNA expression. These results show that Akt is an important regulator of PTEN expression, acting through the transcription factor FoxO1.

FIGURE 4.

Akt regulates FoxO1 phosphorylation and localization. T cells were stimulated under high-dose conditions in the presence or absence of an Akt inhibitor (Akti1/2). (A) Phosphorylation of pS6 (PS6) was monitored by flow cytometry. An example of untreated (left panel) and Akt inhibitor–treated (right panel) T cells stimulated for 12 h are depicted. (B) Phosphorylation of Thr24 on FoxO1 was monitored by Western blotting at the indicated time points. (C) Nuclear and cytoplasmic fractions were Western blotted for total FoxO1 protein. (D) PTEN mRNA levels were determined by qPCR for T cells stimulated for 12 h with high dose (High) and high dose–treated with Akt inhibitor (H+iAkt). (E) ChIP experiments measured the association of FoxO1 and the 5 kb upstream of the PTEN promoter under Th (High) and Th with Akt inhibitor (High+iAkt) conditions. A t test was performed in (D) and (E). The multiple mechanisms identified that differentially regulate PTEN under Treg and Th conditions are illustrated in (F). ***p < 0.001.

FIGURE 4.

Akt regulates FoxO1 phosphorylation and localization. T cells were stimulated under high-dose conditions in the presence or absence of an Akt inhibitor (Akti1/2). (A) Phosphorylation of pS6 (PS6) was monitored by flow cytometry. An example of untreated (left panel) and Akt inhibitor–treated (right panel) T cells stimulated for 12 h are depicted. (B) Phosphorylation of Thr24 on FoxO1 was monitored by Western blotting at the indicated time points. (C) Nuclear and cytoplasmic fractions were Western blotted for total FoxO1 protein. (D) PTEN mRNA levels were determined by qPCR for T cells stimulated for 12 h with high dose (High) and high dose–treated with Akt inhibitor (H+iAkt). (E) ChIP experiments measured the association of FoxO1 and the 5 kb upstream of the PTEN promoter under Th (High) and Th with Akt inhibitor (High+iAkt) conditions. A t test was performed in (D) and (E). The multiple mechanisms identified that differentially regulate PTEN under Treg and Th conditions are illustrated in (F). ***p < 0.001.

Close modal

In conclusion, we have demonstrated that differential regulation of PTEN determines CD4+ T cell fate. In Th differentiation, stimulated by high TCR strength, multiple mechanisms suppress PTEN activity by lowering protein levels and enzymatic activity (Fig. 4F). Low TCR signal strength, resulting in Treg induction, also involves early downregulation of PTEN but requires that PTEN activity is re-established at longer times because inhibition of PTEN under these conditions reduces Treg and increases Th induction. Additionally, we identify a critical feedback loop driven by differential Akt/mTOR signaling that regulates PTEN transcription via the FoxO1 transcription factor. By combining biochemical and computational modeling approaches, our work defines how differential regulation of PTEN can produce alternate CD4+ T cell fate outcomes.

We thank Leonard Harris for help with the Boolean2BNGL package.

This work was supported by National Institutes of Health Grant P41 GM103712 and National Science Foundation Grant 0926181 (to J.R.F. and N.M.-Z.), National Institutes of Health Grants GM080398 and AI095730 (to L.P.K.), and by National Institutes of Health Postdoctoral Training Grant T32 AI089443 (to W.F.H.) and Predoctoral Training Grant T32 EB009403 (to R.P.S.).

The online version of the article contains supplemental material.

Abbreviations used in this article:

ChIP

chromatin immunoprecipitation

mTOR

mammalian target of rapamycin

mTORC2

mTOR complex 2

PTEN

phosphatase and tensin homolog

PTM

posttranslational modification

qPCR

quantitative RT-PCR

siRNA

small interfering RNA

Treg

regulatory T cell.

1
Turner
M. S.
,
Kane
L. P.
,
Morel
P. A.
.
2009
.
Dominant role of antigen dose in CD4+Foxp3+ regulatory T cell induction and expansion.
J. Immunol.
183
:
4895
4903
.
2
Gottschalk
R. A.
,
Corse
E.
,
Allison
J. P.
.
2010
.
TCR ligand density and affinity determine peripheral induction of Foxp3 in vivo.
J. Exp. Med.
207
:
1701
1711
.
3
Kretschmer
K.
,
Apostolou
I.
,
Hawiger
D.
,
Khazaie
K.
,
Nussenzweig
M. C.
,
von Boehmer
H.
.
2005
.
Inducing and expanding regulatory T cell populations by foreign antigen.
Nat. Immunol.
6
:
1219
1227
.
4
Turner
M. S.
,
Isse
K.
,
Fischer
D. K.
,
Turnquist
H. R.
,
Morel
P. A.
.
2014
.
Low TCR signal strength induces combined expansion of Th2 and regulatory T cell populations that protect mice from the development of type 1 diabetes.
Diabetologia
57
:
1428
1436
.
5
Daniel
C.
,
Weigmann
B.
,
Bronson
R.
,
von Boehmer
H.
.
2011
.
Prevention of type 1 diabetes in mice by tolerogenic vaccination with a strong agonist insulin mimetope.
J. Exp. Med.
208
:
1501
1510
.
6
Haxhinasto
S.
,
Mathis
D.
,
Benoist
C.
.
2008
.
The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells.
J. Exp. Med.
205
:
565
574
.
7
Sauer
S.
,
Bruno
L.
,
Hertweck
A.
,
Finlay
D.
,
Leleu
M.
,
Spivakov
M.
,
Knight
Z. A.
,
Cobb
B. S.
,
Cantrell
D.
,
O’Connor
E.
, et al
.
2008
.
T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR.
Proc. Natl. Acad. Sci. USA
105
:
7797
7802
.
8
Miskov-Zivanov
N.
,
Turner
M. S.
,
Kane
L. P.
,
Morel
P. A.
,
Faeder
J. R.
.
2013
.
The duration of T cell stimulation is a critical determinant of cell fate and plasticity.
Sci. Signal.
6
:
ra97
.
9
Shi
Y.
,
Paluch
B. E.
,
Wang
X.
,
Jiang
X.
.
2012
.
PTEN at a glance.
J. Cell Sci.
125
:
4687
4692
.
10
Song
M. S.
,
Salmena
L.
,
Pandolfi
P. P.
.
2012
.
The functions and regulation of the PTEN tumour suppressor.
Nat. Rev. Mol. Cell Biol.
13
:
283
296
.
11
Newton
R. H.
,
Turka
L. A.
.
2012
.
Regulation of T cell homeostasis and responses by pten.
Front. Immunol.
3
:
151
.
12
Francisco
L. M.
,
Salinas
V. H.
,
Brown
K. E.
,
Vanguri
V. K.
,
Freeman
G. J.
,
Kuchroo
V. K.
,
Sharpe
A. H.
.
2009
.
PD-L1 regulates the development, maintenance, and function of induced regulatory T cells.
J. Exp. Med.
206
:
3015
3029
.
13
Bensinger
S. J.
,
Walsh
P. T.
,
Zhang
J.
,
Carroll
M.
,
Parsons
R.
,
Rathmell
J. C.
,
Thompson
C. B.
,
Burchill
M. A.
,
Farrar
M. A.
,
Turka
L. A.
.
2004
.
Distinct IL-2 receptor signaling pattern in CD4+CD25+ regulatory T cells.
J. Immunol.
172
:
5287
5296
.
14
Faeder
J. R.
,
Blinov
M. L.
,
Hlavacek
W. S.
.
2009
.
Rule-based modeling of biochemical systems with BioNetGen.
Methods Mol. Biol.
500
:
113
167
.
15
Saadatpour
A.
,
Albert
I.
,
Albert
R.
.
2010
.
Attractor analysis of asynchronous Boolean models of signal transduction networks.
J. Theor. Biol.
266
:
641
656
.
16
Gillespie
D.
1976
.
A general method for numerically simulating the stochastic time evolution of coupled chemical reactions.
J. Comput. Phys.
22
:
403
434
.
17
Gabryšová
L.
,
Christensen
J. R.
,
Wu
X.
,
Kissenpfennig
A.
,
Malissen
B.
,
O’Garra
A.
.
2011
.
Integrated T-cell receptor and costimulatory signals determine TGF-β-dependent differentiation and maintenance of Foxp3+ regulatory T cells.
Eur. J. Immunol.
41
:
1242
1248
.
18
Gomez-Rodriguez
J.
,
Wohlfert
E. A.
,
Handon
R.
,
Meylan
F.
,
Wu
J. Z.
,
Anderson
S. M.
,
Kirby
M. R.
,
Belkaid
Y.
,
Schwartzberg
P. L.
.
2014
.
Itk-mediated integration of T cell receptor and cytokine signaling regulates the balance between Th17 and regulatory T cells.
J. Exp. Med.
211
:
529
543
.
19
Wang
X.
,
Trotman
L. C.
,
Koppie
T.
,
Alimonti
A.
,
Chen
Z.
,
Gao
Z.
,
Wang
J.
,
Erdjument-Bromage
H.
,
Tempst
P.
,
Cordon-Cardo
C.
, et al
.
2007
.
NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN.
Cell
128
:
129
139
.
20
Torres
J.
,
Rodriguez
J.
,
Myers
M. P.
,
Valiente
M.
,
Graves
J. D.
,
Tonks
N. K.
,
Pulido
R.
.
2003
.
Phosphorylation-regulated cleavage of the tumor suppressor PTEN by caspase-3: implications for the control of protein stability and PTEN-protein interactions.
J. Biol. Chem.
278
:
30652
30660
.
21
Su
H.
,
Bidère
N.
,
Zheng
L.
,
Cubre
A.
,
Sakai
K.
,
Dale
J.
,
Salmena
L.
,
Hakem
R.
,
Straus
S.
,
Lenardo
M.
.
2005
.
Requirement for caspase-8 in NF-κB activation by antigen receptor.
Science
307
:
1465
1468
.
22
Puga
I.
,
Rao
A.
,
Macian
F.
.
2008
.
Targeted cleavage of signaling proteins by caspase 3 inhibits T cell receptor signaling in anergic T cells.
Immunity
29
:
193
204
.
23
Bolduc
D.
,
Rahdar
M.
,
Tu-Sekine
B.
,
Sivakumaren
S. C.
,
Raben
D.
,
Amzel
L. M.
,
Devreotes
P.
,
Gabelli
S. B.
,
Cole
P.
.
2013
.
Phosphorylation-mediated PTEN conformational closure and deactivation revealed with protein semisynthesis.
eLife
2
:
e00691
.
24
Vazquez
F.
,
Ramaswamy
S.
,
Nakamura
N.
,
Sellers
W. R.
.
2000
.
Phosphorylation of the PTEN tail regulates protein stability and function.
Mol. Cell. Biol.
20
:
5010
5018
.
25
Miller
S. J.
,
Lou
D. Y.
,
Seldin
D. C.
,
Lane
W. S.
,
Neel
B. G.
.
2002
.
Direct identification of PTEN phosphorylation sites.
FEBS Lett.
528
:
145
153
.
26
Sestero
C. M.
,
McGuire
D. J.
,
De Sarno
P.
,
Brantley
E. C.
,
Soldevila
G.
,
Axtell
R. C.
,
Raman
C.
.
2012
.
CD5-dependent CK2 activation pathway regulates threshold for T cell anergy.
J. Immunol.
189
:
2918
2930
.
27
Kerdiles
Y. M.
,
Stone
E. L.
,
Beisner
D. R.
,
McGargill
M. A.
,
Ch’en
I. L.
,
Stockmann
C.
,
Katayama
C. D.
,
Hedrick
S. M.
.
2010
.
Foxo transcription factors control regulatory T cell development and function.
Immunity
33
:
890
904
.
28
Ouyang
W.
,
Liao
W.
,
Luo
C. T.
,
Yin
N.
,
Huse
M.
,
Kim
M. V.
,
Peng
M.
,
Chan
P.
,
Ma
Q.
,
Mo
Y.
, et al
.
2012
.
Novel Foxo1-dependent transcriptional programs control Treg cell function.
Nature
491
:
554
559
.
29
Brunet
A.
,
Bonni
A.
,
Zigmond
M. J.
,
Lin
M. Z.
,
Juo
P.
,
Hu
L. S.
,
Anderson
M. J.
,
Arden
K. C.
,
Blenis
J.
,
Greenberg
M. E.
.
1999
.
Akt promotes cell survival by phosphorylating and inhibiting a forkhead transcription factor.
Cell
96
:
857
868
.
30
Zmajkovicova
K.
,
Jesenberger
V.
,
Catalanotti
F.
,
Baumgartner
C.
,
Reyes
G.
,
Baccarini
M.
.
2013
.
MEK1 is required for PTEN membrane recruitment, AKT regulation, and the maintenance of peripheral tolerance.
Mol. Cell
50
:
43
55
.
31
Julien
L. A.
,
Carriere
A.
,
Moreau
J.
,
Roux
P. P.
.
2010
.
mTORC1-activated S6K1 phosphorylates Rictor on threonine 1135 and regulates mTORC2 signaling.
Mol. Cell. Biol.
30
:
908
921
.
32
Gorentla
B. K.
,
Wan
C. K.
,
Zhong
X. P.
.
2011
.
Negative regulation of mTOR activation by diacylglycerol kinases.
Blood
117
:
4022
4031
.

The authors have no financial conflicts of interest.

Supplementary data