Abstract
Pten, one of the most common tumor suppressors in human cancers, antagonizes signaling by the PI3K pathway. Mice with thymocyte-specific deletion of Pten rapidly developed peripheral lymphomas and autoimmunity, which may have been due to failed negative selection of thymocytes or from dysregulation of post-thymic T cells. We induced conditional deletion of Pten from CD4 helper T cells using a Cre knocked into the Tnfrsf4 (OX40) locus to generate OX40CrePtenf mice. Pten-deficient helper T cells proliferated more and produced higher concentrations of cytokines. The OX40CrePtenf mice had a general increase in the number of lymphocytes in the lymph nodes, but not in the spleen. When transferred into wild-type mice, Pten-deficient helper T cells enhanced anti-Listeria responses and the clearance of tumors under conditions in which wild-type T cells had no effect. Moreover, inflammatory responses were exaggerated and resolved later than in OX40CrePtenf mice than in wild type mice. However, in contrast to models of thymocyte-specific Pten deletion, lymphomas and autoimmunity were not observed, even in older OX40CrePtenf mice. Hence, loss of Pten enhances helper T cell function without obvious deleterious effects.
Introduction
CD4 T cells support the coordinated activation of other leukocytes during immune responses. For instance, CD4 T cells secrete inflammatory cytokines during contact hypersensitivity (CHS) reactions, augment tumor surveillance, help B cells during the germinal center reactions, and license dendritic cells to express high levels of MHC and costimulatory ligands (1-4). Full activation of naive CD4 T cells requires persistent stimulation of the TCR and CD28 over a period of about 24 hours. During this time, the T cells interact with antigen-presenting cells (APCs) and integrate signals needed to increase metabolism, grow in size and upregulate cytokine, chemokine and costimulatory receptors (5). After initial activation, CD4 T cells begin to divide in response to cytokines and express additional costimulatory receptors such as ICOS and OX40. They then differentiate into different T helper (Th) cell lineages which secrete cytokines such as interferon-γ (IFNγ), interleukin 4 (IL-4) or IL-17. Eventually, most CD4 Th cells die through apoptosis, but some survive as CD4 memory Th cells (6). Similarly, after initial activation, naïve CD8 T cells differentiate to become CTLs and the ones that survive the cycle of expansion and contraction become CD8 memory T cells (7).
The TCR and many costimulatory and cytokine receptors activate phosphoinositide 3-kinases (PI3Ks) (8). The class I PI3Ks (p110α, p110β, p110γ and p110δ) use PtdIns(4,5)P2 as their preferred substrate to generate the second messenger molecule PtdIns(3,4,5)P3, which helps activate PH domain containing proteins such as Akt. Akt phosphorylates Foxo transcription factors and proteins associated with mTOR complex activation (9, 10). PI3K activity is also required for optimal Erk phosphorylation in T cells (11). By these and other mechanisms, PI3Ks contribute to the proliferation, growth, survival, cytokine production, trafficking, and homeostasis of CD4 T cells (8-16). In T cells, roles for p110α and p110β have not been reported, but the p110γ isoform is activated by G-protein coupled receptors and regulates basal motility in the lymph node (LN), chemotaxis of effector T cells to sites of inflammation and the survival of memory T cells (17-21). p110γ can also regulate chemotaxis in human T cells (22). p110δ is the functionally dominant isoform downstream of TCR, ICOS and the IL-2 receptors and controls Ag-specific events such as differentiation (11, 12, 23-25). PI3K activity remains high for several days after CD4 T cell activation (26), and using acute inhibition with an isoform-selective inhibitor, we demonstrated that p110δ activity is required beyond the first 24 hours following TCR activation to regulate cytokine production (16). We also demonstrated the p110δ is a major regulator of cytokine production in human T cells from healthy, atopic and arthritic individuals (16). Given the importance of Th cytokine production in supporting protective and pathological immune responses, it is important not only to understand how the PI3K pathway is activated, but also how it is sustained or curtailed.
Pten de-phosphorylates PtdIns(3,4,5)P3 to produce PtdIns(4,5)P2, thus terminating PI3K signaling. Pten is frequently lost or mutated in human cancers, including 20% of T cell acute lymphoblastic leukemias (27). Pten expression is also lost in commonly used human T cell lines such as Jurkat which may have confounded some studies where these cells are used to study T cell signaling (28, 29). Conditional deletion of Pten in thymocytes led to defective negative selection, progressive lymphoproliferation, autoimmunity and CD4 T cell lymphomas (30-32). Similar effects were observed in mice that over-expressed micro-RNAs that target Pten (33). After activation, peripheral Pten−/− T cells proliferated more, resisted apoptosis, and failed to contract after superantigen stimulation (30, 34). Pten−/− T cells also could be activated in the absence of CD28 co-stimulation and resisted CTLA4-Ig-induced anergy induction and so Pten was suggested to impose a threshold for activation during initial TCR sensing (32). Although Pten expression levels may modulate immediate responses downstream of the TCR in thymocytes and naïve T cells, the role of Pten during an ongoing immune response is not well understood.
To address the role of Pten in mature CD4 Th cells, we conditionally deleted the Pten gene in Th cells using a Cre-recombinase gene knocked into Tnfrsf4 (the gene that encodes OX40) (35). OX40 is transiently expressed about 24 hours after activation in the majority of CD4 T cells, but rarely in CD8 T cells (36). OX40 is also constitutively expressed on regulatory T cells (Tregs) (36). We found that deletion of Pten after TCR stimulation regulated the magnitude and duration of T cell responses, but not apoptosis or contraction. Furthermore, contrary to when Pten was deleted in thymocytes, lymphoma did not develop when the deletion occurred in activated Th cells. Instead, over-production of cytokines in these mice leads to altered homeostasis of the lymphocyte compartment, and enhanced inflammatory, anti-bacterial and anti-tumor responses.
Materials and Methods
Mice
All mice were maintained under specific pathogen-free conditions. All experiments were performed in accordance with U.K. Home Office regulations. OT2 (37), Rag1−/− (38), Rag2−/− (39), OX40Cre (35), Ptenf (40) and R26YFP (41) and R26tdRFP (42) mice were described previously
Reagents
Unless otherwise stated, all chemicals were from Sigma-Aldrich. IC87114 (IC)3 was synthesized as previously described (16) and anti-CD3 (2C11) was purified in house. IL-2 was synthesized by GSK. Cells were cultured in RPMI 1640 supplemented with 100 U/ml penicillin/streptomycin, 2 mM L-glutamine, 10 mM HEPES, 20 μM 2-ME, and 5% FCS at 5% CO2.
Antibodies, flow cytometry and Western blot
All Abs were from eBioscience except for Fas and CD4-V500 (BD); Pten, p-Akt and p-Erk (Cell Signal Technologies; and anti-rabbit Ig Alexa 647 (Invitrogen). To stain for transcription factors, Pten or intracellular cytokine, FoxP3 staining buffer set or IC Fixation Buffer Kits (both eBioscience) were used, respectively. To detect signaling molecules, CD4 T cells were purified with a Naïve CD4 T cell Kit (Miltenyi) and activated with 1μg/mL anti-CD3 on irradiated APCs. Every 24 hr, an aliquot of cells were treated with Fix Buffer I and Permeabilization Buffer 3 (BD) according to manufacturer’s instruction, and cells stained at the end of the time course. Apoptotic cells were detected with 7-AAD +/− Annexin V (BD), according to manufacturer’s instructions. Flow cytometry data was acquired with FACSCalibur or LSRII instruments (BD) and analyzed with FlowJo (Tree Star). Cells were sorted using a FACSAria (BD) machine. Cell counts were performed either using a CASYCounter or FlowCount Fluorospheres (Beckman Coulter).
Cell purification, proliferation and cytokine assays
CD4 T cells were purified, stimulated, and proliferation and cytokine production measured as described previously (12, 16). To measure cytokines by intracellular FACS or by Mouse Cytokine Array Panel A (R&D Systems), cells were stimulated with 1μg/mL PdBu and 5nM Ionomycin for 5 hrs and 10μg/mL Brefeldin A was added for the final 2hrs. For the Mouse Cytokine Array Panel A, lysates then were made from 5×107 cells and cytokines detected according to manufacturer’s instructions. Pixel density was analyzed using Aida Image Analysis software.
Treg suppression assay
Suppression assays with anti-CD3-coated APCs were performed as described previously (43). Suppression was calculated as the amount of proliferation when Tregs were present compared to when they were absent. (% suppression = (cpm responders with Tregs/cpm responders alone) * 100).
Autoantibody detection
Autoantibodies were detected using a Hep2 ANA kit (The Binding Site). MRL positive control serum was a gift from L. Martensson-Bopp.
Transwell assays
OT2 Ptenf and OT2 OX40CrePtenf cells were activated in vitro with OVA peptide for 3 days, and then live CD4 T cells were purified. Next, CD4 T cells were co-cultured at various ratios with CD45.1+ naïve WT splenocytes, either in direct contact or separated by 0.4μM Transwell inserts (Fisher). After three days, the numbers of CD45.1+ CD4, CD8 and B220 cells were calculated.
CHS assays
CHS assays were done as described previously (16). Briefly, trinitrochlorobenzene (TNCB4)-sensitized mice were re-challenged with TNCB and dosed twice daily with 30mg/kg IC or 1% methylcellulose vehicle control for 2 days. 26 days after the first challenge, the same ear was re-challenged to examine secondary responses. These mice had not received drug during the primary elicitation. Ear size was measured with a micrometer (Kroeplin).
Adoptive transfer assays
CD45.1 hosts were injected i.v. with 106 OT2 Ptenf or OT2 OX40CrePtenf CD4 T cells and the next day injected s.c. with PBS or 50 μg LPS O26:B6+ 1mg OVA. Skin draining LNs were harvested on day 3, 6, or 9 after immunization.
Listeria monocytogenes (Lm5) assays
CD45 mis-matched hosts were injected i.v. with PBS or 1.5×106 OT2 Rag2−/− or OT2 OX40CrePtenfRag2−/− cells. The following day, hosts were i.v. injected with 107 CFU of attenuated OVA-secreting LM (ActA-LM-OVA)(44). After 3 days, splenocytes were stained for intracellular cytokines. APC-labeled SIINFEKL-MHC Class I tetramer (Beckman-Coulter) were used to detect OVA-specific CD8 T cells.
Tumor assays
CD45.1+ hosts were injected i.v. with PBS or 2×105 CD45.2+ OT2 Ptenf Rag2−/− or OT2 OX40CrePtenfRag2−/− cells. The following day, 5×105 EG7 were injected s.c. into hosts. Tumors were palpable between days 10-11. On day 14 after tumor injection, mice were culled and tumors were weighed.
Statistics
Statistics were calculated with GraphPad or SSCS software. The following symbols are used on graphs and tables: * 0.05≥p>0.01 **0.01≥p>0.001 ***0.001≥p
Results
PI3K signaling in OX40CrePtenf T cells is sustained
In OX40Cre mice, Cre is expressed almost exclusively in activated CD4 T cells and Tregs and only 2-5% of CD8 T cells (35). Using reporter mice, we detected Cre activity in 20% of CD4+ T cells one day after activation, increasing to 80% after three days (Fig S1A). PCR analysis revealed nearly complete recombination of the Pten locus in YFP+, OX40CrePtenf CD4 T cells whereas no deletion was observed in YFP− cells. This showed that the YFP accurately reported Cre activity on the Ptenf gene (Fig S1B). Flow cytometry showed loss of Pten protein in OX40CrePtenf CD4+CD25+ and CD4+CD44+ T cells, but not in CD4+CD44− T cells (Fig. 1A). These results confirm that Pten was deleted in Treg and memory T cells, but not in naïve T cells. When stimulated with anti-CD3 and APCs, Akt and Erk phosphorylation were sustained for longer in OX40CrePtenf CD4 T cells compared to controls (Fig. 1B). This was consistent with the kinetics observed for Cre expression after activation (Fig S1A)
Figure 1. Loss of Pten leads to altered signalling and LN hyperplasia.
A) Pten was detected ex vivo by intracellular flow cytometry in CD25+, CD44lo and CD44hi CD4 T cells mice. n≥5 for each genotype. p values were determined with Student’s t-test. B) CD62L+CD4+ T cells were activated by irradiated APCs and anti-CD3, and the level of Akt and Erk phosphorylation were measured at 24 hr intervals. Representative histograms and graphs of median fluorescence intensities are on the right. Data shows mean±SD for three mice from each genotype, and represents one of three independent experiments. Repeated measures ANOVA shows statistical significance over the whole time course of 0.05≥p≥0.01 for both phospho-Akt and phospho-Erk. The results of Bonferroni post-tests also showed that phospho-Akt was strongly increased in OX40CrePtenf cells at day three. C,D) Blood, spleen, and LN cells from 5-10 week old Ptenf and OX40CrePtenf mice were stained for lineage and activation markers, and analyzed by flow cytometry. C) Representative histograms of activation markers in CD4 cells. n≥5 for each genotype. D) Total numbers of B cells and CD4 and CD8 T cells. Student’s t test was used to calculate p values.
Pten regulates lymphocyte homeostasis
Pten deletion led to enhanced CD4 T cell activation in non-challenged animals. Ultimately, this led to enhanced activation of CD4 T cells as evidenced by increased proportion of cells expressing high levels of CD44 and lower levels of CD62L, and CD45RB in LNs, spleen and blood of OX40CrePtenf mice (Fig. 1C). However, unlike mice with Pten deletion in thymocytes (30), auto-antibodies were not detected in OX40CrePtenf mice even at an advanced age (Fig. S2). Furthermore, histology of 22 organs from 19 week old OX40CrePtenf mice (n=5) and controls (n=5) showed no signs of inflammation or autoimmunity. More than 80% of mice with a germ-line heterozygous deletion in Pten form lymphomas by 24 weeks of age and 100% of mice with a thymocyte deletion of Pten develop lymphomas by 12 weeks of age (30, 45, 46). These lymphomas derive from the thymus (47-49), but this did not exclude the possibility of lymphomas also arising in mice with post-thymic deletion of Pten. However, none of 31 OX40CrePtenf mice culled between 25-37 weeks of age showed evidence of lymphomas. In addition, none of five mice aged over 52 weeks had lymphomas. Therefore the pathology associated with Pten deletion in all T cells was not evident when using OX40Cre. Although OX40CrePtenf mice did not develop lymphomas, their LNs –but not spleens –were enlarged from five weeks after birth with 2-3-fold more T and B cells in the LNs compared to littermates. In contrast, lymphocyte numbers in the spleen and blood were similar (Fig. 1D).
Since OX40cre is expressed by regulatory T cells in the thymus (35), and because Pten can affect Treg expansion in vitro (50), we enumerated Tregs in OX40CrePtenf mice. There were similar numbers of Tregs in the thymi of OX40CrePtenf mice, but more in the LNs (Fig. S3A-D). Consistent with previous results (50, 51), there were no significant differences in suppression of wild type (WT) responders by OX40CrePtenf Tregs or in the ability of Tregs of either genotype to suppress OX40CrePtenf responders (Fig. S3E). Hence, deficiencies in Tregs or susceptibility of Th cells to suppression are unlikely to have caused LN hyperplasia in OX40CrePtenf mice.
Lymphocyte expansion is controlled in trans by Th cells
We next considered whether Pten-deficient Th cells altered homeostasis of Pten-sufficient bystander lymphocytes. To test this hypothesis, we generated mixed bone marrow chimeras in lethally irradiated Rag−/− mice, in which 50% of the donor bone marrow derived from CD45.1+ WT SJL mice and 50% derived from either CD45.2+ OX40CrePtenf or CD45.2+ WT mice. Eight weeks after reconstitution, the number of lymphocytes in spleens of hosts receiving WT:SJL or OX40CrePtenf:SJL bone marrow was similar (Fig. 2A). By contrast, there was 3-4-fold more B and T cells in LNs of mice receiving SJL:OX40CrePtenf bone marrow than controls (Fig. 2B). When the ratio of CD45.1 to CD45.2 cells was analyzed, however, we found both donors contributed equally to cells re-populating the LNs and spleens, suggesting that OX40CrePtenf CD4 T cells support general accumulation of LN cells independently of their Pten status.
Figure 2. OX40CrePtenf CD4 T cells support expansion of lymphocytes.
Lethally irradiated Rag2−/− were reconstituted with a 50:50 mix of CD45.1+ WT SJL BM and either CD45.2+ WT (n=7) or OX40CrePtenf (n=9) bone marrow. A) Spleen and B) LN were analyzed eight weeks later for the number of lymphocytes and the relative contribution (ratio) of CD45.1 and CD45.2 bone marrow. Students t test was used to calculate p values. C) Activated CD4 cells from CD45.2+ OT2 Ptenf (n=3) and OT2 OX40CrePtenf (n=3) mice were co-cultured at various ratios with CD45.1+ splenocytes while in direct contact (top panels) or while separated by a Transwell filter (bottom panels). After three days, the number of CD45.1+ lymphocytes was calculated. Data shows mean+SD. Three-way ANOVA was used to calculate statistical significance in number of responder cells recovered. OT2 OX40CrePtenf vs.WT OT2 without Transwell: CD4 and CD8 T cells 0.05≥p≥0.01; B cells p≤0.001. No Transwell vs. Transwell: No significant difference in either OT2 OX40CrePtenf or WT OT2. Results of significant Bonferroni post-tests for each titration point are indicated on the graph.
To test if a soluble factor was needed to expand lymphocytes, Ptenf or OT2 OX40CrePtenf CD4 OT2 T cells were activated using the OVA-derived peptide recognized by the OT2 TCR. The activated OT2 T cells were then purified and co-cultured with unfractionated WT lymphocytes. After three days, the number of WT lymphocytes was counted (Fig. 2C). OT2 OX40CrePtenf CD4 T cells maintained WT lymphocytes better than controls when effectors were in direct contact with responders . This difference between genotypes was unaffected by inclusion of a Transwell filter, demonstrating that expansion of responders was due to secretion of a soluble factor by OX40CrePtenf effector cells.
Pten regulates cytokine production by activated CD4 T cells
In an effort to determine which cytokines might be responsible for the hyperplasia, LN cells were stimulated with PdBu and ionomycin to stimulate cytokine production. Next, lysates made from equal numbers of cells were used to probe a cytokine array testing 40 cytokines and chemokines and the signal from OX40CrePtenf cells was divided by the signal from control cells (Fig. S4A). This chemiluminescent array is a relative measure of cytokine production, where a small difference in pixel density between genotypes can indicate a large difference in the absolute amount of cytokine. Although we could not identify a single factor responsible for hyperplasia, we found a limited range of factors were over-produced in OX40CrePtenf LN, and these included cytokines made by different Th subsets and those that could be induced indirectly by Th cells, such as the chemokine CXCL9 (also known as MIG: Monokine-induced by IFNγ). Hence, we postulate that an environment rich in multiple helper T cell-dependent cytokines resulted in LN hyperplasia.
OT2+OX40CrePtenf showed enhanced proliferation and produced greater amounts of IL-2, IL-4 and, IFNγ when stimulated with peptide (Fig. S4C-E). The enhanced proliferation and cytokine production could be blocked by inhibitors against p110δ, Akt and Erk added 24h after activation (Fig. S4B-E). The results from figure S4 could be affected the increased number of CD4+ T cells with an activated phenotype (32). We therefore purified naïve CD62L+ and antigen-experienced CD62L− CD4 T cells from OX40CrePtenf mice. Both populations showed a 2-3 fold increase in proliferation and cytokine production when stimulated with anti-CD3 (Fig. 3A-D). High concentrations of the pan-PI3K inhibitor LY294002 (LY) or the p110δ-selective inhibitor IC87114 (IC) reduced proliferation and cytokine production in OX40CrePtenf and WT cells to near background levels. At lower concentrations of IC, OX40CrePtenf T cells were less sensitive than WT T cells, suggesting that the activity of PI3K isoforms other than p110δ are also normally restrained by Pten (Fig. 3A-D). To examine Th cell differentiation, CD4 T cells were activated in vitro in presence of IL-12 to produce IFNγ-secreting Th1 cells. A greater proportion of OT2 OX40CrePtenf T cells had divided and produced IFNγ than controls (Fig 3E-G). In addition, IFNγ+ OX40CrePtenf T cells stained more brightly for IFNγ than did WT cells (Fig. 3E,H). Together these data demonstrate that Pten acts to restrain the number of cells dividing, the number of those cells producing cytokine, and the amount of cytokine produced by individual cells.
Figure 3. Pten antagonizes p110δ-regulated proliferation and cytokine production.
A-D) CD4 T cells from Ptenf (n=3) and OX40CrePtenf (n=3) were sorted into A,C) CD62L+ and B,D) CD62L− populations and stimulated with anti-CD3 and either DMSO, 10μM LY, 10μM, 1μM or 0.1μM IC (doses represented by triangle). Two days later, A, B) proliferation and C,D) IFNγ production were analyzed. Data shows mean± SEM. Data represents three independent experiments. E-H) CFSE-labelled OT2 Ptenf and OT2 OX40CrePtenf were stimulated with peptide in Th1-skewed conditions, and cells were stained after three days for intracellular IFNγ. Data is representative of three independent experiments. E) Representative plots. F) CFSE dilution was used to determine the number of mitoses/generations each cell had gone through. G) The proportion of IFNγ+ cells was determined for each generation. H) The MFI in IFNγ+ cells was determined for each generation to quantify the amount of IFNγ each cell was making.
Pten regulates the magnitude and kinetics of immune responses
To understand if altered homeostasis caused by Pten-deficient Th cells affected immune responses, we used a CHS model where increased ear thickness can be used as a measure of T cell-dependent inflammation. Mice were sensitized by application of TNCB on their abdomens and then re-challenged six and 32 days later on their ear to elicit primary and secondary hypersensitivity responses, respectively. Mice were dosed orally with either IC or vehicle control during re-challenges (Fig. 4A). The magnitude and duration of the primary response was greater in OX40CrePtenf than in WT mice. The enhanced inflammation was partially p110δ dependent because IC reduced ear swelling in WT and OX40CrePtenf mice. The magnitude and duration of the secondary responses was also greater in OX40CrePtenf than WT mice (Fig. 4B). IC again reduced ear swelling in both groups. We conclude that OX40CrePtenfl mice experience a greater inflammatory immune response which nonetheless can be resolved and which can be attenuated by oral administration of p110δ inhibitors.
Figure 4. Pten limits CHS responses by a p110δ-dependent mechanism.
A) TNCB-sensitized WT and OX40CrePtenf mice were challenged with TNCB on an ear. Mice were dosed with 30mg/kg IC or vehicle control one hr before challenge and twice daily for two days. Ear size was measured before challenge and at 24 hr intervals. B) TNCB-sensitized WT and OX40CrePtenf mice were challenged with TNCB on an ear and then re-challenged on the same ear 24 days later. Mice were also dosed with 30mg/kg or vehicle control and then twice daily for two days during the secondary response only. Data shows mean±SD of ≥6 mice for each group, and represents two experiments for un-drugged mice and one experiment for IC-drugged mice. C) Statistics were measured by repeated measures ANOVA. Table shows results of Bonferroni post-tests on each day of ear measurement.
In order to follow the activation kinetics of OX40CrePtenf T cells in vivo, we adoptively transferred OT2 Ptenf or OX40CrePtenf CD4 OT2 T cells into WT hosts. Following immunization with LPS and OVA protein, there were significantly more OT2 OX40CrePtenf donor cells than controls after six days of activation. However, nine days after activation both donor cell types had returned to baseline levels (Fig. 5A). This shows that Pten can control the magnitude of T cell response to Ag after initial TCR activation events, yet the lack of Pten expression does not interfere with the contraction of the T cell response. Consistent with this, we found no difference in apoptosis of OT2 OX40CrePtenf cells compared to controls during the first 72 h following activation in vitro (Fig. 5B). Activated OT2 OX40CrePtenf had a mild survival advantage in response to anti-Fas-induced death, but behaved similarly to controls in response to cytokine deprivation, anti-CD3 stimulation, and γ-irradiation (Fig. 5C). Hence, while activation is enhanced, the absence of Pten does not necessarily interfere with apoptotic signaling in OX40CrePtenf T cells.
Figure 5. OX40CrePtenf Th cells can undergo apoptosis in vitro and contract in vivo normally.
A) OT2 Ptenf or OT2 OX40CrePtenf CD4 T cells were injected into SJL (CD45.1). The number of CD45.2+ donor cells in the draining LNs was counted at various time points after s.c. injection with either PBS or LPS+OVA. Data show mean±SD of a minimum of four mice from three independent experiments. Student’s t test was used to calculate p values. B) Death was assayed in CD4 T cells at 24hr intervals after in vitro activation. Data represents 2 independent experiments. C) CD4 T cells were activated in vitro for three days then cultured with no IL-2, 20ng/mL IL-2, 20ng/mL IL-2 +activation-induced death stimuli (10μg anti-CD3, 1μg/mL anti-Fas) or 20ng/mL IL-2 +DNA damage (400 rads γ-irradiation). Death was assayed after 24hrs. Data show mean ± SD for n ≥ 6 for each genotype. Student’s t test was used to calculate p values.
To test how OX40CrePtenf T cells respond to pathogens, we transferred naïve OT2 OX40CrePtenfRag2−/− or OT2 Rag2−/− control T cells into WT hosts and infected these with attenuated Lm expressing OVA (ActA-Lm-OVA). The numbers of adoptively transferred OT2 OX40CrePtenfRag2−/− cells appeared to be enhanced compared with controls in response to infection, though this difference was not statistically significant (Fig. 6A). However, twice as many of these cells produced IL-2 and IFNγ compared to controls three days after infection (Fig. 6B,C). We also measured the magnitude of the endogenous CD8 T cell response to ActA-Lm-OVA using MHC tetramers. More OVA-specific CD8 T cells expanded in infected mice that had received OT2 OX40CrePtenfRag2−/− compared to those receiving OT2 Rag2−/− cells (Fig. 6D). In fact, WT OT2 Rag2−/− cells failed to increase the proportion of Lm-responsive CD8 T cells, consistent with published results (52).
Figure 6. OX40CrePtenf Th cells can enhance bacterial and tumor responses.
A-C) Mice injected with cells from OT2 Rag−/− (n=5) or OT2-OX40CrePtenfRag2−/− (n=5) mice were immunized with 107 CFU ActA-Lm-Ova. After 3 days, spleens were analyzed for A) the number of transferred cells and the proportion that produced B,C) IL-2 or IFNγ. D) Mice injected with PBS (n=6), OT2 Rag2−/− (n=6) or OT2 OX40CrePtenfRag2−/− (n=6) were immunized with 107 CFU ActA-Lm-Ova. The percent of endogenous OVA-tetramer positive CD8 T cells from blood taken on day 8 after infection is shown. Repeated measures ANOVA was used to calculate p values. E) Mice injected i.v. with OT2 PtenfRag2−/− (n=10), OT2 OX40CrePtenfRag2−/− (n=10) or PBS (n=8) were inoculated s.c. with EG7 cells. Excised tumors were weighed on day 14. Data show median values. p values were calculated using Mann-Whitney tests. Data represent two independent experiments.
These results raised the possibility that OX40CrePtenf CD4 T cells could promote cytotoxic immune responses in vivo under conditions where WT CD4 T cells fail to make a difference. To test this possibility further, mice were inoculated with an OVA-expressing thymoma cell line (EG7) and the ability of transferred OT2 cells to limit the growth of the tumors was determined. Indeed, mice with adoptively transferred OT2 OX40CrePtenfRag2−/− but not OT2 PtenfRag2−/− T cells rejected the tumors (Fig. 6E,F). We conclude that OX40CrePtenf CD4 T cells can promote cytotoxic immune responses potentially by activating CD8 T cells under conditions where WT CD4 T cells fail to do so.
Discussion
We have shown here that Pten is not an essential tumor suppressor in peripheral CD4 Th cells. Instead, Pten plays an important role in regulating lymphocyte homeostasis. When Pten was lost after activation, CD4 Th cells hyper-proliferated and produced higher concentrations of cytokines. We postulate that cytokine over-production turns Pten-deficient Th cells into ‘super-helpers’ that enhance inflammatory, anti-bacterial and anti-tumor responses. However, the enhanced responses could still be resolved and spontaneous disease did not develop, in contrast to pathology in many other mouse KO models of negative regulatory proteins such as CTLA-4 and Cbl (53). In addition, bone marrow chimera and Transwell studies suggest that a soluble factor led to a cell non-CD4 T cell autonomous expansion of lymphocytes in LNs. Recently it was shown that cytokines permeate the LN and induce signaling in distant bystander lymphocytes (54). Hence, excess cytokine produced by Pten-deficient Th cells might affect other naïve lymphocytes, and significantly alter lymph node homeostasis.
We confirmed in vitro and in a CHS model that Pten acts at least in part by antagonizing signaling by the PI3K p110δ. However, while p110δ inhibitors completely block activation in WT cells, they were only partially effective in Pten-deficient cells. This suggests that other isoforms can participate in a sub-dominant manner after T cell activation, but secondary to p110δ. When negative regulatory pathways are impaired, the effects of other PI3K isoforms can accrue and become more readily detectable. Possible candidates are p110α, which we have recently shown contributes to tonic Ag receptor signaling in B cells (55), or chemokine-dependent p110γ activity (19, 20, 56, 57).
Previous studies have shown that although lymphomas are found in the lymph nodes and spleens of Pten-deficient mice, they are actually derived from the thymus (48, 49). Mechanistically, this has been linked to c-myc translocation and overexpression (49, 58). Using converse experiments, we show here that Pten does not act as a tumor suppressor in mature T cells. In humans, mature T cell lymphomas are rare compared to thymic-derived lymphomas, which may reflect increased genome stability in more mature T cells. Indeed, DNA damage checkpoint regulators were dysregulated in Pten-deficient thymocytes but not peripheral T cells (48). Alternatively, there may be additional backup mechanisms to prevent lymphomagenesis in peripheral T cells. Consistent with this, leukemic oncogenes only caused transformation when ectopically expressed in hematopietic stem cells and not mature T cells (59).
Pten has previously been shown to deter autoimmunity because when it was deleted from thymocytes, autoantibodies, autoreactive T cells and lymphoid interstitial pneumonia developed (30). Autoantibodies and tissue infilitration still arose when young mice were thymectomized, which the authors suggested was caused by defective activation-induced cell death in peripheral T cells (49). However, autoreactivity in those studies could still have been caused by failed thymocyte negative selection in young animals (30, 32). We detected no evidence of spontaneous autoantibody production, autoimmunity or inflammation in our model, possibly because there were no defects in Treg suppression or because apoptosis was only mildly affected. We thus conclude Pten is not an essential repressor of apoptosis or autoimmunity stemming from mature T cells.
Although spontaneous immunopathologies did not develop in OX40CrePtenf mice, heightened immune responses did. We previously showed that T-dependent humoral immune responses were enhanced in OX40CrePtenf mice due to increased number and cytokine production of TFH cells (24). Here we show that OX40CrePtenf Th cells enhanced cellular immune responses to Lm and prevent tumour growth. Since Pten deficiency revealed a previously unrecognized potential for Th in promoting primary CD8 T cell responses (52), we propose that OX40CrePtenf T cells are ‘super-helpers’. The concept of super-helpers has potential clinical implications because adoptive immunotherapy and prophylactic vaccination is currently aimed at modulating responding CTLs, although interest is growing in exploiting CD4 T cells as well (4). Pten-deficient super-helper Th could potentially improve clinical outcomes. Ag-specific CD4 cells have previously been shown to promote partial or complete regression in lymphopenic environments or when high numbers were infused at regular intervals alongside Ag-specific CTLs (60-62). Here we demonstrate that similar results can be achieved using a single injection of Pten-deficient Ag-specific CD4 T cells in normal hosts. Reduction of Pten activity in mature T cells using genetic or chemical means may therefore be both safe and desirable in some therapeutic settings such as tumor immunotherapy.
Supplementary Material
Acknowledgements
We thank Nigel Killeen, Pier Paolo Pandolfi and Hans Jorg Fehling for mouse strains and Hou Shen for the attenuated Listeria expressing ovalbumin. We thank Jonathan Clark for the synthesis of IC87114, staff at the Biological Services Unit and Flow Cytometry facility for expert help and Anne Segonds-Pichon for statistical advice.
Abbreviations
- CHS
contact hypersensitivity
- Treg
Regulatory T cell
- IC
IC87114
- TNCB
trinitrochlorobenzene
- Lm
Listeria monocytogenes
- LY
LY294002
Footnotes
This research was funded by a core strategic grant (KO and MT) and project grant BBF0154611 (KO), both from the Biotechnology and Biological Sciences Research Council; a Wellcome Trust New Investigator Award (KO); and a Senior Non Clinical Fellowship from the Medical Research Council (MT).
References
- 1.Castellino F, Germain RN. Cooperation between CD4+ and CD8+ T cells: when, where, and how. Annu Rev Immunol. 2006;24:519–540. doi: 10.1146/annurev.immunol.23.021704.115825. [DOI] [PubMed] [Google Scholar]
- 2.Crotty S. Follicular helper CD4 T cells (TFH) Annu Rev Immunol. 2011;29:621–663. doi: 10.1146/annurev-immunol-031210-101400. [DOI] [PubMed] [Google Scholar]
- 3.Wang B, Fujisawa H, Zhuang L, Freed I, Howell BG, Shahid S, Shivji GM, Mak TW, Sauder DN. CD4+ Th1 and CD8+ type 1 cytotoxic T cells both play a crucial role in the full development of contact hypersensitivity. J Immunol. 2000;165:6783–6790. doi: 10.4049/jimmunol.165.12.6783. [DOI] [PubMed] [Google Scholar]
- 4.Muranski P, Restifo NP. Adoptive immunotherapy of cancer using CD4(+) T cells. Curr Opin Immunol. 2009;21:200–208. doi: 10.1016/j.coi.2009.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Henrickson SE, von Andrian UH. Single-cell dynamics of T-cell priming. Curr Opin Immunol. 2007;19:249–258. doi: 10.1016/j.coi.2007.04.013. [DOI] [PubMed] [Google Scholar]
- 6.Pepper M, Jenkins MK. Origins of CD4(+) effector and central memory T cells. Nat Immunol. 2011;131:467–471. doi: 10.1038/ni.2038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Williams MA, Bevan MJ. Effector and memory CTL differentiation. Annu Rev Immunol. 2007;25:171–192. doi: 10.1146/annurev.immunol.25.022106.141548. [DOI] [PubMed] [Google Scholar]
- 8.Okkenhaug K, Fruman DA. PI3Ks in lymphocyte signaling and development. Curr Top Microbiol Immunol. 2010;346:57–85. doi: 10.1007/82_2010_45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fruman DA, Bismuth G. Fine tuning the immune response with PI3K. Immunol Rev. 2009;228:253–272. doi: 10.1111/j.1600-065X.2008.00750.x. [DOI] [PubMed] [Google Scholar]
- 10.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 mTOR pathways control T lymphocyte trafficking. Nat Immunol. 2008;9:513–521. doi: 10.1038/ni.1603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Okkenhaug K, Bilancio A, Farjot G, Priddle H, Sancho S, Peskett E, Pearce W, Meek SE, Salpekar A, Waterfield MD, Smith AJ, Vanhaesebroeck B. Impaired B and T cell antigen receptor signaling in p110delta PI 3-kinase mutant mice. Science. 2002;297:1031–1034. doi: 10.1126/science.1073560. [DOI] [PubMed] [Google Scholar]
- 12.Okkenhaug K, Patton DT, Bilancio A, Garcon F, Rowan WC, Vanhaesebroeck B. The p110delta isoform of phosphoinositide 3-kinase controls clonal expansion and differentiation of Th cells. J Immunol. 2006;177:5122–5128. doi: 10.4049/jimmunol.177.8.5122. [DOI] [PubMed] [Google Scholar]
- 13.Okkenhaug K, Ali K, Vanhaesebroeck B. Antigen receptor signalling: a distinctive role for the p110delta isoform of PI3K. Trends Immunol. 2007;28:80–87. doi: 10.1016/j.it.2006.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Patton DT, Garcon F, Okkenhaug K. The PI3K p110delta controls T-cell development, differentiation and regulation. Biochem Soc Trans. 2007;35:167–171. doi: 10.1042/BST0350167. [DOI] [PubMed] [Google Scholar]
- 15.Salmond RJ, Emery J, Okkenhaug K, Zamoyska R. MAPK, phosphatidylinositol 3-kinase, and mammalian target of rapamycin pathways converge at the level of ribosomal protein S6 phosphorylation to control metabolic signaling in CD8 T cells. J Immunol. 2009;183:7388–7397. doi: 10.4049/jimmunol.0902294. [DOI] [PubMed] [Google Scholar]
- 16.Soond DR, Bjorgo E, Moltu K, Dale VQ, Patton DT, Torgersen KM, Galleway F, Twomey B, Clark J, Gaston JS, Tasken K, Bunyard P, Okkenhaug K. PI3K p110delta regulates T-cell cytokine production during primary and secondary immune responses in mice and humans. Blood. 2010;115:2203–2213. doi: 10.1182/blood-2009-07-232330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nombela-Arrieta C, Lacalle RA, Montoya MC, Kunisaki Y, Megias D, Marques M, Carrera AC, Manes S, Fukui Y, Martinez AC, Stein JV. Differential requirements for DOCK2 and phosphoinositide-3-kinase gamma during T and B lymphocyte homing. Immunity. 2004;21:429–441. doi: 10.1016/j.immuni.2004.07.012. [DOI] [PubMed] [Google Scholar]
- 18.Nombela-Arrieta C, Mempel TR, Soriano SF, Mazo I, Wymann MP, Hirsch E, Martinez AC, Fukui Y, von Andrian UH, Stein JV. A central role for DOCK2 during interstitial lymphocyte motility and sphingosine-1-phosphate-mediated egress. J Exp Med. 2007;204:497–510. doi: 10.1084/jem.20061780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Martin AL, Schwartz MD, Jameson SC, Shimizu Y. Selective regulation of CD8 effector T cell migration by the p110 gamma isoform of phosphatidylinositol 3-kinase. J Immunol. 2008;180:2081–2088. doi: 10.4049/jimmunol.180.4.2081. [DOI] [PubMed] [Google Scholar]
- 20.Thomas MS, Mitchell JS, DeNucci CC, Martin AL, Shimizu Y. The p110gamma isoform of phosphatidylinositol 3-kinase regulates migration of effector CD4 T lymphocytes into peripheral inflammatory sites. J Leukoc Biol. 2008;84:814–823. doi: 10.1189/jlb.0807561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Barber DF, Bartolome A, Hernandez C, Flores JM, Redondo C, Fernandez-Arias C, Camps M, Ruckle T, Schwarz MK, Rodriguez S, Martinez AC, Balomenos D, Rommel C, Carrera AC. PI3Kgamma inhibition blocks glomerulonephritis and extends lifespan in a mouse model of systemic lupus. Nat Med. 2005;11:933–935. doi: 10.1038/nm1291. [DOI] [PubMed] [Google Scholar]
- 22.Smith LD, Hickman ES, Parry RV, Westwick J, Ward SG. PI3Kgamma is the dominant isoform involved in migratory responses of human T lymphocytes: effects of ex vivo maintenance and limitations of non-viral delivery of siRNA. Cell Signal. 2007;19:2528–2539. doi: 10.1016/j.cellsig.2007.08.006. [DOI] [PubMed] [Google Scholar]
- 23.Garcon F, Patton DT, Emery JL, Hirsch E, Rottapel R, Sasaki T, Okkenhaug K. CD28 provides T-cell costimulation and enhances PI3K activity at the immune synapse independently of its capacity to interact with the p85/p110 heterodimer. Blood. 2008;111:1464–1471. doi: 10.1182/blood-2007-08-108050. [DOI] [PubMed] [Google Scholar]
- 24.Rolf J, Bell SE, Kovesdi D, Janas ML, Soond DR, Webb LM, Santinelli S, Saunders T, Hebeis B, Killeen N, Okkenhaug K, Turner M. Phosphoinositide 3-kinase activity in T cells regulates the magnitude of the germinal center reaction. J Immunol. 2010;185:4042–4052. doi: 10.4049/jimmunol.1001730. [DOI] [PubMed] [Google Scholar]
- 25.Macintyre AN, Finlay D, Preston G, Sinclair LV, Waugh CM, Tamas P, Feijoo C, Okkenhaug K, Cantrell DA. Protein kinase B controls transcriptional programs that direct cytotoxic T cell fate but is dispensable for T cell metabolism. Immunity. 2011;34:224–236. doi: 10.1016/j.immuni.2011.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Song J, Salek-Ardakani S, Rogers PR, Cheng M, Van Parijs L, Croft M. The costimulation-regulated duration of PKB activation controls T cell longevity. Nat Immunol. 2004;5:150–158. doi: 10.1038/ni1030. [DOI] [PubMed] [Google Scholar]
- 27.Palomero T, Sulis ML, Cortina M, Real PJ, Barnes K, Ciofani M, Caparros E, Buteau J, Brown K, Perkins SL, Bhagat G, Agarwal AM, Basso G, Castillo M, Nagase S, Cordon-Cardo C, Parsons R, Zuniga-Pflucker JC, Dominguez M, Ferrando AA. Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat Med. 2007;13:1203–1210. doi: 10.1038/nm1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Astoul E, Edmunds C, Cantrell DA, Ward SG. PI 3-K and T-cell activation: limitations of T-leukemic cell lines as signaling models. Trends Immunol. 2001;22:490–496. doi: 10.1016/s1471-4906(01)01973-1. [DOI] [PubMed] [Google Scholar]
- 29.Abraham RT, Weiss A. Jurkat T cells and development of the T-cell receptor signalling paradigm. Nat Rev Immunol. 2004;4:301–308. doi: 10.1038/nri1330. [DOI] [PubMed] [Google Scholar]
- 30.Suzuki A, Yamaguchi MT, Ohteki T, Sasaki T, Kaisho T, Kimura Y, Yoshida R, Wakeham A, Higuchi T, Fukumoto M, Tsubata T, Ohashi PS, Koyasu S, Penninger JM, Nakano T, Mak TW. T cell-specific loss of Pten leads to defects in central and peripheral tolerance. Immunity. 2001;14:523–534. doi: 10.1016/s1074-7613(01)00134-0. [DOI] [PubMed] [Google Scholar]
- 31.Hagenbeek TJ, Naspetti M, Malergue F, Garcon F, Nunes JA, Cleutjens KB, Trapman J, Krimpenfort P, Spits H. The loss of PTEN allows TCR alphabeta lineage thymocytes to bypass IL-7 and Pre-TCR-mediated signaling. J Exp Med. 2004;200:883–894. doi: 10.1084/jem.20040495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Buckler JL, Walsh PT, Porrett PM, Choi Y, Turka LA. Cutting edge: T cell requirement for CD28 costimulation is due to negative regulation of TCR signals by PTEN. J Immunol. 2006;177:4262–4266. doi: 10.4049/jimmunol.177.7.4262. [DOI] [PubMed] [Google Scholar]
- 33.Xiao C, Srinivasan L, Calado DP, Patterson HC, Zhang B, Wang J, Henderson JM, Kutok JL, Rajewsky K. Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes. Nat Immunol. 2008;9:405–414. doi: 10.1038/ni1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.You H, Pellegrini M, Tsuchihara K, Yamamoto K, Hacker G, Erlacher M, Villunger A, Mak TW. FOXO3a-dependent regulation of Puma in response to cytokine/growth factor withdrawal. J Exp Med. 2006;203:1657–1663. doi: 10.1084/jem.20060353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Klinger M, Kim JK, Chmura SA, Barczak A, Erle DJ, Killeen N. Thymic OX40 expression discriminates cells undergoing strong responses to selection ligands. J Immunol. 2009;182:4581–4589. doi: 10.4049/jimmunol.0900010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Croft M. Control of immunity by the TNFR-related molecule OX40 (CD134) Annu Rev Immunol. 2010;28:57–78. doi: 10.1146/annurev-immunol-030409-101243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Barnden MJ, Allison J, Heath WR, Carbone FR. Defective TCR expression in transgenic mice constructed using cDNA-based alpha- and beta-chain genes under the control of heterologous regulatory elements. Immunol Cell Biol. 1998;76:34–40. doi: 10.1046/j.1440-1711.1998.00709.x. [DOI] [PubMed] [Google Scholar]
- 38.Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68:869–877. doi: 10.1016/0092-8674(92)90030-g. [DOI] [PubMed] [Google Scholar]
- 39.Shinkai Y, Rathbun G, Lam KP, Oltz EM, Stewart V, Mendelsohn M, Charron J, Datta M, Young F, Stall AM, Alt FW. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell. 1992;68:855–867. doi: 10.1016/0092-8674(92)90029-c. [DOI] [PubMed] [Google Scholar]
- 40.Trotman LC, Niki M, Dotan ZA, Koutcher JA, Di Cristofano A, Xiao A, Khoo AS, Roy-Burman P, Greenberg NM, Van Dyke T, Cordon-Cardo C, Pandolfi PP. Pten dose dictates cancer progression in the prostate. PLoS Biol. 2003;1:E59. doi: 10.1371/journal.pbio.0000059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Srinivas S, Watanabe T, Lin CS, William CM, Tanabe Y, Jessell TM, Costantini F. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol. 2001;1:4. doi: 10.1186/1471-213X-1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Luche H, Weber O, Nageswara Rao T, Blum C, Fehling HJ. Faithful activation of an extra-bright red fluorescent protein in “knock-in” Cre-reporter mice ideally suited for lineage tracing studies. Eur J Immunol. 2007;37:43–53. doi: 10.1002/eji.200636745. [DOI] [PubMed] [Google Scholar]
- 43.Patton DT, Wilson MD, Rowan WC, Soond DR, Okkenhaug K. The PI3K p110delta Regulates Expression of CD38 on Regulatory T Cells. PLoS One. 2011;6:e17359. doi: 10.1371/journal.pone.0017359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Pearce EL, Shen H. Generation of CD8 T cell memory is regulated by IL-12. J Immunol. 2007;179:2074–2081. doi: 10.4049/jimmunol.179.4.2074. [DOI] [PubMed] [Google Scholar]
- 45.Suzuki A, de la Pompa JL, Stambolic V, Elia AJ, Sasaki T, del Barco Barrantes I, Ho A, Wakeham A, Itie A, Khoo W, Fukumoto M, Mak TW. High cancer susceptibility and embryonic lethality associated with mutation of the PTEN tumor suppressor gene in mice. Curr Biol. 1998;8:1169–1178. doi: 10.1016/s0960-9822(07)00488-5. [DOI] [PubMed] [Google Scholar]
- 46.Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nat Genet. 1998;19:348–355. doi: 10.1038/1235. [DOI] [PubMed] [Google Scholar]
- 47.Hagenbeek TJ, Spits H. T-cell lymphomas in T-cell-specific Pten-deficient mice originate in the thymus. Leukemia. 2008;22:608–619. doi: 10.1038/sj.leu.2405056. [DOI] [PubMed] [Google Scholar]
- 48.Xue L, Nolla H, Suzuki A, Mak TW, Winoto A. Normal development is an integral part of tumorigenesis in T cell-specific PTEN-deficient mice. Proc Natl Acad Sci U S A. 2008;105:2022–2027. doi: 10.1073/pnas.0712059105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Liu X, Karnell JL, Yin B, Zhang R, Zhang J, Li P, Choi Y, Maltzman JS, Pear WS, Bassing CH, Turka LA. Distinct roles for PTEN in prevention of T cell lymphoma and autoimmunity in mice. J Clin Invest. 2010;120:2497–2507. doi: 10.1172/JCI42382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Walsh PT, Buckler JL, Zhang J, Gelman AE, Dalton NM, Taylor DK, Bensinger SJ, Hancock WW, Turka LA. PTEN inhibits IL-2 receptor-mediated expansion of CD4+ CD25+ Tregs. J Clin Invest. 2006;116:2521–2531. doi: 10.1172/JCI28057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Buckler JL, Liu X, Turka LA. Regulation of T-cell responses by PTEN. Immunol Rev. 2008;224:239–248. doi: 10.1111/j.1600-065X.2008.00650.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sun JC, Bevan MJ. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science. 2003;300:339–342. doi: 10.1126/science.1083317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Acuto O, Di Bartolo V, Michel F. Tailoring T-cell receptor signals by proximal negative feedback mechanisms. Nat Rev Immunol. 2008;8:699–712. doi: 10.1038/nri2397. [DOI] [PubMed] [Google Scholar]
- 54.Perona-Wright G, Mohrs K, Mohrs M. Sustained signaling by canonical helper T cell cytokines throughout the reactive lymph node. Nat Immunol. 2010;11:520–526. doi: 10.1038/ni.1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ramadani F, Bolland DJ, Garcon F, Emery JL, Vanhaesebroeck B, Corcoran AE, Okkenhaug K. The PI3K isoforms p110alpha and p110delta are essential for pre-B cell receptor signaling and B cell development. Sci Signal. 2010;3:ra60. doi: 10.1126/scisignal.2001104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Barber DF, Bartolome A, Hernandez C, Flores JM, Fernandez-Arias C, Rodriguez-Borlado L, Hirsch E, Wymann M, Balomenos D, Carrera AC. Class IB-phosphatidylinositol 3-kinase (PI3K) deficiency ameliorates IA-PI3K-induced systemic lupus but not T cell invasion. J Immunol. 2006;176:589–593. doi: 10.4049/jimmunol.176.1.589. [DOI] [PubMed] [Google Scholar]
- 57.Berod L, Heinemann C, Heink S, Escher A, Stadelmann C, Drube S, Wetzker R, Norgauer J, Kamradt T. PI3Kgamma deficiency delays the onset of experimental autoimmune encephalomyelitis and ameliorates its clinical outcome. Eur J Immunol. 2011;41:833–844. doi: 10.1002/eji.201040504. [DOI] [PubMed] [Google Scholar]
- 58.Guo W, Schubbert S, Chen JY, Valamehr B, Mosessian S, Shi H, Dang NH, Garcia C, Theodoro MF, Varella-Garcia M, Wu H. Suppression of leukemia development caused by PTEN loss. Proc Natl Acad Sci U S A. 2011 doi: 10.1073/pnas.1006937108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Newrzela S, Cornils K, Li Z, Baum C, Brugman MH, Hartmann M, Meyer J, Hartmann S, Hansmann ML, Fehse B, von Laer D. Resistance of mature T cells to oncogene transformation. Blood. 2008;112:2278–2286. doi: 10.1182/blood-2007-12-128751. [DOI] [PubMed] [Google Scholar]
- 60.Antony PA, Piccirillo CA, Akpinarli A, Finkelstein SE, Speiss PJ, Surman DR, Palmer DC, Chan CC, Klebanoff CA, Overwijk WW, Rosenberg SA, Restifo NP. CD8+ T cell immunity against a tumor/self-antigen is augmented by CD4+ T helper cells and hindered by naturally occurring T regulatory cells. J Immunol. 2005;174:2591–2601. doi: 10.4049/jimmunol.174.5.2591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Xie Y, Akpinarli A, Maris C, Hipkiss EL, Lane M, Kwon EK, Muranski P, Restifo NP, Antony PA. Naive tumor-specific CD4(+) T cells differentiated in vivo eradicate established melanoma. J Exp Med. 2010;207:651–667. doi: 10.1084/jem.20091921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Shafer-Weaver KA, Watkins SK, Anderson MJ, Draper LJ, Malyguine A, Alvord WG, Greenberg NM, Hurwitz AA. Immunity to murine prostatic tumors: continuous provision of T-cell help prevents CD8 T-cell tolerance and activates tumor-infiltrating dendritic cells. Cancer Res. 2009;69:6256–6264. doi: 10.1158/0008-5472.CAN-08-4516. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






