Naive T cells receive stimulation from the positive selecting ligand in the periphery for their survival. This stimulation does not normally lead to overt activation of T cells, as the T cells remain largely quiescent until they receive either antigenic or lymphopenic stimuli. The underlying mechanism responsible for survival and quiescence of the naive T cells remains largely unknown. In this study, we report that T cell-specific deletion of Tsc1, a negative regulator of mammalian target of rapamycin, resulted in both spontaneous losses of quiescence and cellularity, especially within the CD8 subset. The Tsc1-deficient T cells have increased cell proliferation and apoptosis. Tsc1 deletion affects the survival and quiescence of T cells in the absence of antigenic stimulation. Loss of quiescence but not cellularity was inhibited by rapamycin. Our data demonstrate that tuberous sclerosis complex–mammalian target of rapamycin maintains quiescence and survival of T cells.

Lymphocytes are generally referred to as “resting” cells prior to antigenic stimulation. However, these cells are active at molecular levels, as evidenced by constitutive phosphorylation of the TCR ζ-chain in resting T cells (1). Requirements for engagement of Ag receptors in T cell survival are inferred by the need for BCRs or TCRs for their respective survivals (2, 3). Moreover, accumulating data also support the view that survival of naive T cells requires the positive-selecting MHC (4, 5). In addition to antigenic stimulation, T cells undergo homeostatic proliferation in a lymphopenic environment (6). In the absence of Ag and lymphopenia, self-MHC ligands do not lead to overt activation of T cells. Thus, T cells remain largely quiescent in the absence of antigenic or lymphopenic stimuli. These observations raise two interesting questions. First, is there an active mechanism that maintains the quiescence of naive T cells? Second, does the mechanism that maintains the quiescence of T cells also control their survival?

The tuberous sclerosis complex (TSC)–mammalian target of rapamycin (mTOR) pathway has emerged as a central regulator for cellular metabolism (711). More recent studies have revealed two functionally distinct complexes with different components, TORC1 and TORC2 (1214). Among the different components, TOR is associated with Raptor and others to form TORC1 (15). Because rapamycin–FKBP12 selectively binds to TORC1 (16, 17), it is specifically inhibited by rapamycin (12), although prolonged exposure to rapamycin may also affect TORC2 (18). In contrast, the Rictor–TOR complex formed the core of TORC2 (12). Although the TSC negatively regulates TORC1 function, recent studies suggest that defects in the TSC result in reduced TORC2 function, either directly or indirectly (14, 19, 20).

A critical role for the mTOR pathway in T cell activation was deduced from the fact that rapamycin, which specifically targets mTOR, has been used as an immunosuppressant in transplantation (21). Additionally, a role for mTOR in lymphocyte homing has also been reported (22). More recent studies have indicated that the mTOR pathway regulates generation of effector T cells and regulatory T cells (23, 24). Surprisingly, in vivo administration of rapamycin potently induced memory T cells in the presence of Ags (25, 26) and rejuvenated the aging hematopoietic stem cells (27). Because all reports on mTOR and T cells focus on T cell responses to Ag, it is of interest to establish the role of this pathway in the quiescence and survival of naive T cells in the absence of antigenic stimulation. In this study, we report that targeted mutation of Tsc1, an important regulator of mTOR, leads to loss of quiescence and cellularity in T cells, particularly in the CD8 subset. Interestingly, loss of quiescence and cellularity are mediated by distinct mechanisms.

Tsc1fl/fl mice (28) were provided by Dr. David J. Kwiatkowski and backcrossed for six generations onto a C57BL/6 background in the animal facility at University of Michigan. CD4-Cre (29) mice were from Taconic (Hudson, NY), and Lck-Cre (30) mice were from the The Jackson Laboratory. Mice were housed under specific pathogen-free conditions. All studies involving mice have been approved by University Laboratory Animal Use and Care Committee at the University of Michigan.

The Abs and annexin V used in the study were obtained from eBioscience and BD Biosciences unless otherwise noted. Alexa Fluor 488-conjugated p-S6 Abs were obtained from Cell Signaling Technology. Intracellular staining of p-S6 was carried out as described (11, 27). Cell apoptosis and death were analyzed by staining cells with FITC- or allophycocyanin-conjugated annexin V in conjunction with 7-aminoactinomycin D (BD Pharmingen) according to the manufacturer’s protocol.

Mice were treated with either vehicle (5% polyethylene glycol 400, 5% Tween 80 in PBS) or rapamycin (4 mg/kg) every other day for 2–7 wk or as specified in the text. The splenocytes and thymocytes were analyzed at the specified time points.

Thymocytes were sorted into four subsets (CD4+CD8+, CD4CD8+, CD4+CD8, and CD4CD8) by FACSAria. The efficacy of Tsc1 deletion in genomic DNA was quantitated by real-time PCR. Genomic DNA was isolated using a PicoPure DNA extraction kit (Arcturus, Mountain View, CA), and quantitative PCR was performed with previously described TSC1 primers F4536 and F4830 using an ABI 7500 real-time PCR system (Applied Biosystems, Foster City, CA) (31).

In vivo BrdU labelings were performed according BD Pharmingen’s suggested protocol. Briefly, 8-wk-old Cre+ and CreTsc1fl/fl littermates were i.p. injected with 200 μl 10 mg/ml BrdU in PBS followed by feeding the mice with drinking water containing 1 mg/ml BrdU for 24 h before mice were euthanized. Single-cell suspensions were prepared from thymus and spleen. Cells were stained with anti-CD4 and anti-CD8 Abs from eBioscience (San Diego, CA). BrdU+ populations were stained using FITC BrdU flow kits from BD Pharmingen according to the manufacturer’s recommended protocol. FACS analyses were preformed on a BD Biosciences LSRII.

To isolate lymphocytes, lungs were first perfused by injecting 10 ml PBS through the right ventricle of the heart with dissected hepatic vein and excised. The lung tissues were then cut into 1- to 2-mm3 pieces in 1× HBSS and incubated in 1 mg/ml collagenase II (Invitrogen, Carlsbad, CA) in 1× HBSS for 1 h at 37°C. The digested tissues were minced with frosted microscope slides and pass through a 70-μm cell strainer. Cells were then washed twice with staining buffer (1× HBSS with 2% FBS and 0.04% NaN3) before staining with Abs. Anti-CD45 Abs were included in the Ab cocktails. Cells were gated on CD45+ population when analyzed by flow cytometry.

Peyer’s patches were removed and minced with glass slides to get single cell suspension for further analysis. Intraepithelial lymphocytes (IEL) were isolated according to protocols described in Lycke and Lefrançois (32). Briefly, the small intestines were cut open horizontally and rinsed at least four times in ice-cold CMF solution (1× HBSS, Ca2+- and Mg2+-free, 10 mM HEPES, 25 mM sodium bicarbonate [pH 7.2], 2% FBS). Cleaned small intestines were cut into 0.5-cm pieces with scissors. IEL were extracted by rotating at 37°C with 20 ml CMF solution with 5 mM EDTA for 20 min. The cells in the supernatants were collected after the tissues had settled down. The extraction procedure was repeated one more time. The supernatants were combined and cells were washed twice with RPMI 1640 supplemented with 10% FBS, 100 U/ml penicillin, 100 U/ml streptomycin, 2 mM l-glutamine, and 0.0005% 2-ME and resuspended in 8 ml 44% Percoll (GE Healthcare) in PBS solution, which were overloaded onto 5 ml 67% Percoll PBS solution and then centrifuged at 600 × g for 20 min at room temperature with low acceleration and deceleration settings. IEL were collected by carefully harvesting 1–2 cm of the 44% and 67% Percoll interface.

A Student t test was used to determine the statistical significance of the differences. All tests are two-tailed. Significance was defined as follows: *0.01 < p ≤ 0.05, **0.001 < p ≤ 0.01, ***p ≤ 0.001.

To study the function of Tsc1in T cells, we backcrossed the floxed Tsc1 allele into the C57BL/6 background for six generations and induced deletion by crossing the Tsc1fl/fl mice (31) to the Tsc1fl/fl;Lck-Cre+/− mice or Tsc1fl/fl;CD4-Cre+/− mice that express the recombinase Cre under the control of either proximal Lck (30) or CD4 promoter (29). The Cre+ and Cre littermates were used for the study.

To verify the efficacy of gene deletion, we sorted the thymic CD4CD8, CD4+CD8+, CD3+CD4+CD8, and CD3+CD4CD8+ subsets from Lck-Cre+ and Lck-Cre- Tsc1fl/fl littermates. Real-time PCR using genomic DNA as templates was used to determine the relative amounts of deleted and undeleted DNA forms. As shown in Fig. 1A, the efficacy of deletion reached ∼90% among CD4+CD8+, CD3+CD4+CD8, and CD3+CD4CD8+ subsets. Because the Lck promoter is activated at DN3 (29), little deletion is observed within the CD4CD8 subset (Fig. 1A). Consistent with the functional role of Tsc1 in cell size regulation, the Cre+ thymocytes were significantly larger than the Cre thymocytes (Fig. 1B). A mild but statistically significant decrease in the percentage of the CD3+CD8+CD4 and CD3+CD8CD4+ thymocytes was observed in Cre+ mice (Fig. 1C).

FIGURE 1.

Tsc1 deletion increase sizes of thymocytes but does not abrogate T cell development. A, Efficacy of deletion of the Tsc1 gene by lck-Cre. Data shown are the relative abundance of floxed region of the Tsc1 gene as measured by real-time PCR. The DNA was isolated with FACS-purified thymocyte subsets. B, Tsc1 deletion increases the size of thymocytes, based on forward scatters. C, Distribution of CD4 and CD8 expression in Tsc1fl/fl;lck-Cre+ and Tsc1fl/fl;lck-Cre littermates. FACS profiles of a representative thymus in each group are shown in the left, while the percentage of CD3+CD4+CD8 and the CD3+CD4CD8+ subsets are shown in the right. Data shown are 12-d-old littermates. Similar results were observed in mice ranging from 2 to 20 wk old. *0.01 < p ≤ 0.05.

FIGURE 1.

Tsc1 deletion increase sizes of thymocytes but does not abrogate T cell development. A, Efficacy of deletion of the Tsc1 gene by lck-Cre. Data shown are the relative abundance of floxed region of the Tsc1 gene as measured by real-time PCR. The DNA was isolated with FACS-purified thymocyte subsets. B, Tsc1 deletion increases the size of thymocytes, based on forward scatters. C, Distribution of CD4 and CD8 expression in Tsc1fl/fl;lck-Cre+ and Tsc1fl/fl;lck-Cre littermates. FACS profiles of a representative thymus in each group are shown in the left, while the percentage of CD3+CD4+CD8 and the CD3+CD4CD8+ subsets are shown in the right. Data shown are 12-d-old littermates. Similar results were observed in mice ranging from 2 to 20 wk old. *0.01 < p ≤ 0.05.

Close modal

In the spleen, the percentage of CD4 and CD8 T cells was further reduced in Cre+ mice (Fig. 2A). The percentage of CD4 T cells was reduced by ∼2-fold, whereas those of the CD8 T cells was reduced by ∼2- to 5-fold (Fig. 2B). As a result, an increase of the CD4/CD8 ratio was observed after deletion of Tsc1 (Fig. 2B). The cell loss can be observed in both young (2-wk-old) and adult mice (Fig. 2B). Corresponding to the dramatic reduction of CD8 percentage of T cells, the numbers of CD8 T cells were also significantly reduced at all time points tested (Fig. 2C). Owing to larger variations in cell counting, the reduction of CD4 T cells was significant only in the 2-wk-old mice (Fig. 2C). In addition to the change of T cell numbers, the remaining T cells were more active as judged by their cell surface markers. As shown in Fig. 2D and 2E, an overall increase of CD44hi and CD62Llo T cells was observed in the Tsc1-deficient mice. Similar changes in the cell number and phenotypes were observed in lymph nodes (data not shown).

FIGURE 2.

Tsc1 deletion causes losses in both quiescence and cellularity of mature T cells. A, FACS profiles of CD4 and CD8 T cell subsets in the spleen. B, Summary data on percentage of CD4 (upper panel) and CD8 (middle panel) and CD4/CD8 ratios in the spleen. Data shown in the bar graphs are means ± SD. C, Number of CD4+ and CD8+ T cells in the spleen, as calculated by the number of mononuclear cells in spleen times the percentage of T cell subsets. D, FACS profiles of CD44 and CD62L in spleen T cells from 2-mo-old mice. E, Summary data involving 4- to 5–mo-old mice. Data shown are percentages of naive (CD44loCD62Lhi), central memory (CD44hiCD62Lhi), and effector memory T cells (CD44hiCD62Llo). The increased activation of T cells is substantially the same in mice ranging from 2 to 20 wk old. *0.01 < p ≤ 0.05, **0.001 < p ≤ 0.01, ***p ≤ 0.001.

FIGURE 2.

Tsc1 deletion causes losses in both quiescence and cellularity of mature T cells. A, FACS profiles of CD4 and CD8 T cell subsets in the spleen. B, Summary data on percentage of CD4 (upper panel) and CD8 (middle panel) and CD4/CD8 ratios in the spleen. Data shown in the bar graphs are means ± SD. C, Number of CD4+ and CD8+ T cells in the spleen, as calculated by the number of mononuclear cells in spleen times the percentage of T cell subsets. D, FACS profiles of CD44 and CD62L in spleen T cells from 2-mo-old mice. E, Summary data involving 4- to 5–mo-old mice. Data shown are percentages of naive (CD44loCD62Lhi), central memory (CD44hiCD62Lhi), and effector memory T cells (CD44hiCD62Llo). The increased activation of T cells is substantially the same in mice ranging from 2 to 20 wk old. *0.01 < p ≤ 0.05, **0.001 < p ≤ 0.01, ***p ≤ 0.001.

Close modal

To substantiate our basic observation, we also used CD4-Cre to delete the Tsc1 gene and carried out a parallel analysis. The data, presented in Supplemental Fig. 1, support the conclusion that deletion of the Tsc1 gene in T cell lineages caused loss of both quiescence and cellularity in mature T cells. One exception is that the central memory CD8 T cells, which are CD44hi CD62Lhi, were found to be increased in Lck-Cre+ mice (Fig. 2E, middle lower panel) but were decreased in CD4-Cre+ mice with Tsc1 deletion (Supplemental Fig. 1E, middle lower panel). This difference remains to be explained. Because CD4-Cre is known to be more efficient in gene deletion than Lck-Cre, one potential explanation is that heterozygous deletion of Tsc1 by Lck-Cre results in partial activation that leads to production of central memory, whereas more complete deletion by CD4-Cre results in a higher level of T cell activation, leading to a transition to effector phenotype.

We measured the percentage of proliferating T cells by BrdU incorporation. As shown in Fig. 3A, whereas deletion of Tsc1 showed a slight trend of increased T cell proliferation in the thymus, this is statistically insignificant. In contrast, the peripheral CD4 and CD8 T cells from Cre+ mice had a significantly higher percentage of BrdU+ cells. Therefore, deletion of Tsc1 increased proliferation of T cells in the periphery (Fig. 3B, 3C).

FIGURE 3.

Deletion of Tsc1 gene increases proliferation of spleen T cells. The CD4-Cre+ and Cre littermates of the Tsc1fl/fl mice were pulsed with BrdU for 24 h. Thymii and spleens were harvested and stained for CD4, CD8, and BrdU. A, Summary data of BrdU+ percentage cells among CD4+CD8 (upper panel) and CD4CD8+ (lower panel) thymocytes. B, Representative histograms depicting distribution of BrdU staining among spleen CD4+ and CD8+ cells. C, Summary data of BrdU+ percentage cells among CD4+ (upper panel) and CD8+ (lower panel) spleen cells.

FIGURE 3.

Deletion of Tsc1 gene increases proliferation of spleen T cells. The CD4-Cre+ and Cre littermates of the Tsc1fl/fl mice were pulsed with BrdU for 24 h. Thymii and spleens were harvested and stained for CD4, CD8, and BrdU. A, Summary data of BrdU+ percentage cells among CD4+CD8 (upper panel) and CD4CD8+ (lower panel) thymocytes. B, Representative histograms depicting distribution of BrdU staining among spleen CD4+ and CD8+ cells. C, Summary data of BrdU+ percentage cells among CD4+ (upper panel) and CD8+ (lower panel) spleen cells.

Close modal

To understand the mechanism of the loss of Tsc1−/− T cells, we stained the thymocytes (Fig. 4A) and the spleen cells (Fig. 4B) with annexin V and 7-aminoactinomycin D, in conjunction with CD4 and CD8. Representative FACS profiles are shown in the left panels of Fig. 4A and 4B while the summary data involving five to eight mice per group are shown in the right panels. As shown in Fig. 4A and 4B, significant increases of the apoptotic T cells were observed among the CD8 T cells. Additionally, Tsc1 deletion caused loss of mitochondria potential in both CD4 and CD8 T cells, an early indicator of apoptosis (Fig. 4C). The lack of increase in apoptosis of CD4 T cells may be explained by efficient removal of apoptotic cells in vivo (33). To confirm this prediction, we cultured spleen cells in vitro for 6 h. As shown in Fig. 4D, Tsc1 deletion doubled the percentage of apoptotic CD4 and CD8 T cells in the culture. Lack of increase in apoptotic Tsc1−/− CD4 T cells by annexin V staining may therefore be attributable to clearance in vivo.

FIGURE 4.

Tsc1 deletion increases apoptosis of mature CD8 T cells in both thymus and spleen. A, Apoptosis of CD3+CD4CD8+ and CD3+CD4+CD8 thymocytes. B, Apoptosis of mature T cells in the spleen. Representative profiles are shown on the left while summary of data involving 11- to 13-d-old mice are shown on the right. C, Tsc1 deletion caused loss of mitochondria potential in both CD4 and CD8 T cells. Spleen cells from CD4-Cre+ and Cre littermates of the Tsc1fl/fl mice were stained with DiOC6(3) to measure mitochondrial potential. Data shown are historgrams depicting distribution of mitochondrial potential of CD4+ T cells, CD8+ T cells, or non-T spleen cells. Similar increases were observed in lymph node T cells. D, Apoptosis of spleen T cells after 6 h in vitro culture. Data shown are representative profiles and were repeated more than five times. *0.01 < p ≤ 0.05, **0.001 < p ≤ 0.01, ***p ≤ 0.001.

FIGURE 4.

Tsc1 deletion increases apoptosis of mature CD8 T cells in both thymus and spleen. A, Apoptosis of CD3+CD4CD8+ and CD3+CD4+CD8 thymocytes. B, Apoptosis of mature T cells in the spleen. Representative profiles are shown on the left while summary of data involving 11- to 13-d-old mice are shown on the right. C, Tsc1 deletion caused loss of mitochondria potential in both CD4 and CD8 T cells. Spleen cells from CD4-Cre+ and Cre littermates of the Tsc1fl/fl mice were stained with DiOC6(3) to measure mitochondrial potential. Data shown are historgrams depicting distribution of mitochondrial potential of CD4+ T cells, CD8+ T cells, or non-T spleen cells. Similar increases were observed in lymph node T cells. D, Apoptosis of spleen T cells after 6 h in vitro culture. Data shown are representative profiles and were repeated more than five times. *0.01 < p ≤ 0.05, **0.001 < p ≤ 0.01, ***p ≤ 0.001.

Close modal

It has been suggested that activated effector memory T cells preferentially home to mucosal tissues such as the lung and the gut (34). To rule out the possibility that the reduced cellularity in spleen was due to increased homing to mucosal tissues, we examined the CD4 and CD8 T cells from the lung and Peyer’s patches and IEL from small intestine. As shown in Fig. 5A and 5B, the percentages of CD4 and CD8 T cells were comparable in lung and Peyer’s patches between Cre+ and Cre mice, whereas the T cells in spleen were decreased in Cre+ mice. Histological examination of lung (data not shown) and intestine (Supplemental Fig. 2A) showed no increased lymphocyte infiltration. Moreover, the frequency of CD4 and CD8 T cells among IEL was similar in number between Cre+ and Cre mice (Supplemental Fig. 2B). Furthermore, in the OT-1 transgenic mice, no increase of transgenic T cells was attributable to deficiency of TSC1 (Supplemental Fig. 2C). Therefore, loss of mature T cells in the spleen and lymph node was not due to their migration in the mucosal tissue. Perhaps because activated T cells have a tendency to migrate to target tissues, no reduction in T cell frequencies was observed in lung and Peyer’s patches.

FIGURE 5.

Deletion of Tsc1 does not increase T cells in mucosal tissues. A, FACS profiles depicting distribution of CD4 and CD8 markers among CD45+ mononuclear cells from spleen, Peyer’s patch, and lung. B, Summary data of percentage of CD4 and CD8 T cells in the three organs (n = 3–4). This experiment was reproduced twice. *0.01 < p ≤ 0.05, ***p ≤ 0.001.

FIGURE 5.

Deletion of Tsc1 does not increase T cells in mucosal tissues. A, FACS profiles depicting distribution of CD4 and CD8 markers among CD45+ mononuclear cells from spleen, Peyer’s patch, and lung. B, Summary data of percentage of CD4 and CD8 T cells in the three organs (n = 3–4). This experiment was reproduced twice. *0.01 < p ≤ 0.05, ***p ≤ 0.001.

Close modal

Thus, deletion of Tsc1 specifically in T cell lineages caused loss of quiescence and cellularity in mature T cells by increasing the proliferation and apoptosis.

An interesting issue is whether the impact of Tsc1 deletion on naive T cells depends on antigenic stimulation. This issue cannot be resolved in mice with polyclonal T cells, as many of them will be stimulated by environmental Ags. We therefore bred the floxed Tsc1 allele into transgenic mice expressing OT-1 TCR, which is specific for OVA (35). By allelic exclusion, the T cells expressing high levels of transgenic α-chain are likely to be specific for the OVA presented by MHC class I and are thus less likely to recognize environmental Ags. As shown in Fig. 6A, although the mice were not challenged with the OVA, a 3-fold reduction of Vα2hiCD8+ cells was observed in the spleen of the Cre+ mice. This corresponded to an increased apoptosis among the transgenic T cells (Fig. 6B). Additionally, the CD8 T cells are more activated as demonstrated by a 3-fold loss in naive T cells and a 5-fold increase in CD44hiCD62Llo cells (Fig. 6C). Because the loss of quiescence was more robust than what was observed in mice with polyclonal T cell repertoire, the data demonstrated that the spontaneous activation was not induced by Ags, even though a small fraction of the Vα2hi cells may still have rearranged endogenous Tcra locus.

FIGURE 6.

Ag-independent loss of quiescence and cellularity in Tsc1-deficient OT-1. A, Reduced cellularity of CD8+Vα2hi transgenic T cells. B, Tsc1 deletion leads to spontaneous apoptosis of transgenic T cells in the absence of nominal Ag. C, Tsc1 deficiency results in spontaneous activation of the transgenic T cells in the absence of nominal Ags. Data shown are for splenocytes from 2- to 10-mo-old littermates and were repeated twice. Similar data were obtained in more than five independent experiments using PBL from mice ranging from 6 to 12 wk old. *0.01 < p ≤ 0.05, ***p ≤ 0.001.

FIGURE 6.

Ag-independent loss of quiescence and cellularity in Tsc1-deficient OT-1. A, Reduced cellularity of CD8+Vα2hi transgenic T cells. B, Tsc1 deletion leads to spontaneous apoptosis of transgenic T cells in the absence of nominal Ag. C, Tsc1 deficiency results in spontaneous activation of the transgenic T cells in the absence of nominal Ags. Data shown are for splenocytes from 2- to 10-mo-old littermates and were repeated twice. Similar data were obtained in more than five independent experiments using PBL from mice ranging from 6 to 12 wk old. *0.01 < p ≤ 0.05, ***p ≤ 0.001.

Close modal

Tsc1 forms a complex with Tsc2 to regulate mTOR function (711). To determine whether increased mTOR activation in T cells is responsible for losses in both quiescence and cellularity, we treated the Tsc1fl/fl;Lck-Cre+ mice with rapamycin or vehicle. As shown in Fig. 7A, 2 wk rapamycin treatment (4 mg/kg, once every other day) effectively inhibited mTOR activation of the Tsc1fl/fl;Lck-Cre+ T cells, based on intracellular staining of p-S6. Additionally, the treatment restored the cell size of both CD4 and CD8 T cells (Fig. 7B). Furthermore, rapamycin treatment increased the percentage of naive T cells (Fig. 7C). Surprisingly, rapamycin treatment failed to restore cellularity of mature T cells (Fig. 7D). Neither extending the treatment to 7 wk nor decreasing the dose of rapamycin to 75 μg/kg had any appreciable effect on T cell cellularity (Supplemental Fig. 3). Correspondingly, the percentage of apoptotic cells was also unaffected (Supplemental Fig. 3). To confirm that the loss of cellularity was not rescued by rapamycin, we cultured wild-type and mutant T cells in the presence of a wide range of concentrations of rapamycin. As shown in Fig. 7E, rapamycin treatment did not reduce apoptosis of spleen T cells in vitro. The differential effect of rapamycin on apoptosis and cellular activation indicated that loss of quiescence and cellularity may be mediated by different mechanisms.

FIGURE 7.

Loss of quiescence but not cellularity is rescued by rapamycin treatment. A, Validation of rapamycin effect by reduction of p-S6. Data shown in the left panel are mean fluorescence intensity. The middle panel shows representative histograms depicting p-S6 distribution among CD4 and CD8 T cells in vehicle control or rapamycin-treated spleen CD4 and CD8 T cells. The right panel shows Western blot data demonstrating the reduction of p-S6 in rapamycin-treated CD4-Cre+;Tsc1fl/fl splenic T cells. B, Rapamycin treatment restores the cell size of CD4 and CD8 T cells. C, Rapamycin restores the percentage of naive T cells in the Tsc1fl/fl;Lck-Cre+ mice. D, Rapamycin treatment does not increase CD4 and CD8 T cell cellularity in vivo. Similar data were obtained when the duration of treatment extends to 7 wk. E, Rapamycin has no effect on apoptosis of mature T cells in vitro. The data were reproduced twice. *0.01 < p ≤ 0.05, **0.001 < p ≤ 0.01.

FIGURE 7.

Loss of quiescence but not cellularity is rescued by rapamycin treatment. A, Validation of rapamycin effect by reduction of p-S6. Data shown in the left panel are mean fluorescence intensity. The middle panel shows representative histograms depicting p-S6 distribution among CD4 and CD8 T cells in vehicle control or rapamycin-treated spleen CD4 and CD8 T cells. The right panel shows Western blot data demonstrating the reduction of p-S6 in rapamycin-treated CD4-Cre+;Tsc1fl/fl splenic T cells. B, Rapamycin treatment restores the cell size of CD4 and CD8 T cells. C, Rapamycin restores the percentage of naive T cells in the Tsc1fl/fl;Lck-Cre+ mice. D, Rapamycin treatment does not increase CD4 and CD8 T cell cellularity in vivo. Similar data were obtained when the duration of treatment extends to 7 wk. E, Rapamycin has no effect on apoptosis of mature T cells in vitro. The data were reproduced twice. *0.01 < p ≤ 0.05, **0.001 < p ≤ 0.01.

Close modal

Taken together, we showed both spontaneous activation and loss of cellularity of naive T cells after activation of mTOR by Tsc1 deletion. Although the Tsc1 gene controls both survival and quiescence, the downstream effector mechanisms for the two functions may be distinct. On one hand, because rapamycin effectively inhibited activation but not loss of cellularity, it is likely that loss of cellularity was not due to loss of quiescence. On the other hand, although loss of T cells may create lymphopenia that leads to homeostatic proliferation and acquisition of memory markers, the fact that Tsc1 deletion increased T cells with effector memory markers suggests that homeostatic proliferation alone, which primarily resulted in central memory T cells (6), cannot fully account for the spontaneous T cell activation .

A recent report demonstrated that FoxO1 maintains survival and trafficking of naive T cells by regulating expression of Sell, Klf2, and Il7r (36). Because none of the genes are repressed by Tsc1 deletion (Supplemental Fig. 4), it is likely that the FoxO and Tsc mediate nonoverlapping functions, both necessary for survival and quiescence of naive T cells. Additional studies are needed to elucidate how Tsc is stimulated to maintain naive T cells. One of our groups has shown that the Tsc is activated by AMP-activated protein kinase (8). Because AMP-activated protein kinase is known to be activated by TCR signaling (37), it would be of interest to test whether such activation may play a role in T cell quiescence and survival.

Interestingly, deletion of Tsc1, much like that of FoxO1 (36), reduced the number of CD8 but not CD4 T cells. The functional dichotomy of the two subsets remains to be explained. Because the TCR–MHC interaction is necessary for survival of CD8 (2, 4, 38) but apparently not CD4 T cells (39), it is intriguing that both FoxO1 and Tsc1 play major roles in mediating the protection conferred by TCR–MHC interaction.

Regardless of the mechanism, our data provide a direct link between T cell survival and mTOR activation. The molecular link helps to explain how T cells survive under physiological conditions. Moreover, because inflammatory cytokines are potent inducers of mTOR activation (27, 40), our results presented in this study may provide new insights on how inflammation associates with loss of T cells during infections (41, 42).

We thank Darla Kroft for editorial assistance.

This work was supported by National Institutes of Health Grants AG036690 and GM091648 (to P.Z.) and AI64350 and CA58033 (to Y.L.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

IEL

intraepithelial lymphocytes

mTOR

mammalian target of rapamycin

TSC

tuberous sclerosis complex.

1
van Oers
N. S.
,
Killeen
N.
,
Weiss
A.
.
1994
.
ZAP-70 is constitutively associated with tyrosine-phosphorylated TCRζ in murine thymocytes and lymph node T cells.
Immunity
1
:
675
685
.
2
Polic
B.
,
Kunkel
D.
,
Scheffold
A.
,
Rajewsky
K.
.
2001
.
How αβ T cells deal with induced TCRα ablation.
Proc. Natl. Acad. Sci. USA
98
:
8744
8749
.
3
Lam
K. P.
,
Kühn
R.
,
Rajewsky
K.
.
1997
.
In vivo ablation of surface immunoglobulin on mature B cells by inducible gene targeting results in rapid cell death.
Cell
90
:
1073
1083
.
4
Freitas
A. A.
,
Rocha
B.
.
1999
.
Peripheral T cell survival.
Curr. Opin. Immunol.
11
:
152
156
.
5
Tanchot
C.
,
Lemonnier
F. A.
,
Pérarnau
B.
,
Freitas
A. A.
,
Rocha
B.
.
1997
.
Differential requirements for survival and proliferation of CD8 naive or memory T cells.
Science
276
:
2057
2062
.
6
Cho
B. K.
,
Rao
V. P.
,
Ge
Q.
,
Eisen
H. N.
,
Chen
J.
.
2000
.
Homeostasis-stimulated proliferation drives naive T cells to differentiate directly into memory T cells.
J. Exp. Med.
192
:
549
556
.
7
Inoki
K.
,
Guan
K. L.
.
2006
.
Complexity of the TOR signaling network.
Trends Cell Biol.
16
:
206
212
.
8
Inoki
K.
,
Zhu
T.
,
Guan
K. L.
.
2003
.
TSC2 mediates cellular energy response to control cell growth and survival.
Cell
115
:
577
590
.
9
Sancak
Y.
,
Peterson
T. R.
,
Shaul
Y. D.
,
Lindquist
R. A.
,
Thoreen
C. C.
,
Bar-Peled
L.
,
Sabatini
D. M.
.
2008
.
The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1.
Science
320
:
1496
1501
.
10
Kim
E.
,
Goraksha-Hicks
P.
,
Li
L.
,
Neufeld
T. P.
,
Guan
K. L.
.
2008
.
Regulation of TORC1 by Rag GTPases in nutrient response.
Nat. Cell Biol.
10
:
935
945
.
11
Chen
C.
,
Liu
Y.
,
Liu
R.
,
Ikenoue
T.
,
Guan
K. L.
,
Liu
Y.
,
Zheng
P.
.
2008
.
TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species.
J. Exp. Med.
205
:
2397
2408
.
12
Sarbassov
D. D.
,
Ali
S. M.
,
Kim
D. H.
,
Guertin
D. A.
,
Latek
R. R.
,
Erdjument-Bromage
H.
,
Tempst
P.
,
Sabatini
D. M.
.
2004
.
Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton.
Curr. Biol.
14
:
1296
1302
.
13
Jacinto
E.
,
Facchinetti
V.
,
Liu
D.
,
Soto
N.
,
Wei
S.
,
Jung
S. Y.
,
Huang
Q.
,
Qin
J.
,
Su
B.
.
2006
.
SIN1/MIP1 maintains rictor-mTOR complex integrity and regulates Akt phosphorylation and substrate specificity.
Cell
127
:
125
137
.
14
Yang
Q.
,
Inoki
K.
,
Ikenoue
T.
,
Guan
K. L.
.
2006
.
Identification of Sin1 as an essential TORC2 component required for complex formation and kinase activity.
Genes Dev.
20
:
2820
2832
.
15
Hara
K.
,
Maruki
Y.
,
Long
X.
,
Yoshino
K.
,
Oshiro
N.
,
Hidayat
S.
,
Tokunaga
C.
,
Avruch
J.
,
Yonezawa
K.
.
2002
.
Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action.
Cell
110
:
177
189
.
16
Brown
E. J.
,
Albers
M. W.
,
Shin
T. B.
,
Ichikawa
K.
,
Keith
C. T.
,
Lane
W. S.
,
Schreiber
S. L.
.
1994
.
A mammalian protein targeted by G1-arresting rapamycin-receptor complex.
Nature
369
:
756
758
.
17
Sabatini
D. M.
,
Erdjument-Bromage
H.
,
Lui
M.
,
Tempst
P.
,
Snyder
S. H.
.
1994
.
RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs.
Cell
78
:
35
43
.
18
Sarbassov
D. D.
,
Ali
S. M.
,
Sengupta
S.
,
Sheen
J. H.
,
Hsu
P. P.
,
Bagley
A. F.
,
Markhard
A. L.
,
Sabatini
D. M.
.
2006
.
Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB.
Mol. Cell
22
:
159
168
.
19
Huang
J.
,
Wu
S.
,
Wu
C. L.
,
Manning
B. D.
.
2009
.
Signaling events downstream of mammalian target of rapamycin complex 2 are attenuated in cells and tumors deficient for the tuberous sclerosis complex tumor suppressors.
Cancer Res.
69
:
6107
6114
.
20
Huang
J.
,
Dibble
C. C.
,
Matsuzaki
M.
,
Manning
B. D.
.
2008
.
The TSC1-TSC2 complex is required for proper activation of mTOR complex 2.
Mol. Cell. Biol.
28
:
4104
4115
.
21
Abraham
R. T.
,
Wiederrecht
G. J.
.
1996
.
Immunopharmacology of rapamycin.
Annu. Rev. Immunol.
14
:
483
510
.
22
Sinclair
L. V.
,
Finlay
D.
,
Feijoo
C.
,
Cornish
G. H.
,
Gray
A.
,
Ager
A.
,
Okkenhaug
K.
,
Hagenbeek
T. J.
,
Spits
H.
,
Cantrell
D. A.
.
2008
.
Phosphatidylinositol-3-OH kinase and nutrient-sensing mTOR pathways control T lymphocyte trafficking.
Nat. Immunol.
9
:
513
521
.
23
Delgoffe
G. M.
,
Kole
T. P.
,
Zheng
Y.
,
Zarek
P. E.
,
Matthews
K. L.
,
Xiao
B.
,
Worley
P. F.
,
Kozma
S. C.
,
Powell
J. D.
.
2009
.
The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment.
Immunity
30
:
832
844
.
24
Rao
R. R.
,
Li
Q.
,
Odunsi
K.
,
Shrikant
P. A.
.
2010
.
The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin.
Immunity
32
:
67
78
.
25
Araki
K.
,
Turner
A. P.
,
Shaffer
V. O.
,
Gangappa
S.
,
Keller
S. A.
,
Bachmann
M. F.
,
Larsen
C. P.
,
Ahmed
R.
.
2009
.
mTOR regulates memory CD8 T-cell differentiation.
Nature
460
:
108
112
.
26
Pearce
E. L.
,
Walsh
M. C.
,
Cejas
P. J.
,
Harms
G. M.
,
Shen
H.
,
Wang
L. S.
,
Jones
R. G.
,
Choi
Y.
.
2009
.
Enhancing CD8 T-cell memory by modulating fatty acid metabolism.
Nature
460
:
103
107
.
27
Chen
C.
,
Liu
Y.
,
Liu
Y.
,
Zheng
P.
.
2009
.
mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells.
Sci. Signal.
2
:
ra75
.
28
Uhlmann
E. J.
,
Wong
M.
,
Baldwin
R. L.
,
Bajenaru
M. L.
,
Onda
H.
,
Kwiatkowski
D. J.
,
Yamada
K.
,
Gutmann
D. H.
.
2002
.
Astrocyte-specific TSC1 conditional knockout mice exhibit abnormal neuronal organization and seizures.
Ann. Neurol.
52
:
285
296
.
29
Tanigaki
K.
,
Tsuji
M.
,
Yamamoto
N.
,
Han
H.
,
Tsukada
J.
,
Inoue
H.
,
Kubo
M.
,
Honjo
T.
.
2004
.
Regulation of αβ/γδ T cell lineage commitment and peripheral T cell responses by Notch/RBP-J signaling.
Immunity
20
:
611
622
.
30
Gu
H.
,
Marth
J. D.
,
Orban
P. C.
,
Mossmann
H.
,
Rajewsky
K.
.
1994
.
Deletion of a DNA polymerase beta gene segment in T cells using cell type-specific gene targeting.
Science
265
:
103
106
.
31
Kwiatkowski
D. J.
,
Zhang
H.
,
Bandura
J. L.
,
Heiberger
K. M.
,
Glogauer
M.
,
el-Hashemite
N.
,
Onda
H.
.
2002
.
A mouse model of TSC1 reveals sex-dependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells.
Hum. Mol. Genet.
11
:
525
534
.
32
Lycke
N.
,
Lefrançois
L.
.
1996
.
Isolation of mouse small intestinal intraepithelial lymphocytes, Peyer’s patch and lamina propria
. In
Current Protocols in Immunology,
Vol. 13.19.1
.
Coligan
J. E.
,
Kruisbeek
A. M.
,
Margulies
D. H.
,
Shevach
E. M.
,
Strober
W.
, eds.
John Wiley and Sons
,
New York
.
33
Platt
N.
,
Suzuki
H.
,
Kodama
T.
,
Gordon
S.
.
2000
.
Apoptotic thymocyte clearance in scavenger receptor class A-deficient mice is apparently normal.
J. Immunol.
164
:
4861
4867
.
34
Johansson-Lindbom
B.
,
Agace
W. W.
.
2007
.
Generation of gut-homing T cells and their localization to the small intestinal mucosa.
Immunol. Rev.
215
:
226
242
.
35
Clarke
S. R.
,
Barnden
M.
,
Kurts
C.
,
Carbone
F. R.
,
Miller
J. F.
,
Heath
W. R.
.
2000
.
Characterization of the ovalbumin-specific TCR transgenic line OT-I: MHC elements for positive and negative selection.
Immunol. Cell Biol.
78
:
110
117
.
36
Kerdiles
Y. M.
,
Beisner
D. R.
,
Tinoco
R.
,
Dejean
A. S.
,
Castrillon
D. H.
,
DePinho
R. A.
,
Hedrick
S. M.
.
2009
.
Foxo1 links homing and survival of naive T cells by regulating L-selectin, CCR7 and interleukin 7 receptor.
Nat. Immunol.
10
:
176
184
.
37
Tamás
P.
,
Hawley
S. A.
,
Clarke
R. G.
,
Mustard
K. J.
,
Green
K.
,
Hardie
D. G.
,
Cantrell
D. A.
.
2006
.
Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes.
J. Exp. Med.
203
:
1665
1670
.
38
Takada
K.
,
Jameson
S. C.
.
2009
.
Self-class I MHC molecules support survival of naive CD8 T cells, but depress their functional sensitivity through regulation of CD8 expression levels.
J. Exp. Med.
206
:
2253
2269
.
39
Dorfman
J. R.
,
Stefanová
I.
,
Yasutomo
K.
,
Germain
R. N.
.
2000
.
CD4+ T cell survival is not directly linked to self-MHC-induced TCR signaling.
Nat. Immunol.
1
:
329
335
.
40
Chen
C.
,
Liu
Y.
,
Liu
Y.
,
Zheng
P.
.
2010
.
Mammalian target of rapamycin activation underlies HSC defects in autoimmune disease and inflammation in mice.
J. Clin. Invest.
120
:
4091
4101
.
41
Badovinac
V. P.
,
Porter
B. B.
,
Harty
J. T.
.
2004
.
CD8+ T cell contraction is controlled by early inflammation.
Nat. Immunol.
5
:
809
817
.
42
Favre
D.
,
Lederer
S.
,
Kanwar
B.
,
Ma
Z. M.
,
Proll
S.
,
Kasakow
Z.
,
Mold
J.
,
Swainson
L.
,
Barbour
J. D.
,
Baskin
C. R.
, et al
.
2009
.
Critical loss of the balance between Th17 and T regulatory cell populations in pathogenic SIV infection.
PLoS Pathog.
5
:
e1000295
.

The authors have no financial conflicts of interest.