Abstract
T cell subsets differ in their metabolic requirements, and further insight into such differences might be harnessed to selectively promote regulatory T cells (Tregs) for therapies in autoimmunity and transplantation. We found that Gln restriction during human T cell activation favored CD4 T cells with high expression of the Treg transcription factor FOXP3. This resulted from shrinking numbers and reduced proliferation of activated FOXP3lo/−CD4 T cells while FOXP3hiCD4 T cell numbers increased. This gain was abolished by blocking Gln synthetase, an enzyme that responds to Gln and purine/pyrimidine deficiencies. The shift toward FOXP3hiCD4 T cells under Gln restriction was recapitulated with inhibitors of Gln-dependent pyrimidine and purine syntheses that together closely mimicked declining cell numbers and cell cycles, and by small interfering RNA knockdown of the respective rate-limiting Gln-consuming enzymes CAD and PPAT. FOXP3hi-enriched CD25hiCD4 T cells from these cultures inhibited proliferation, but they also produced effector cytokines, including IL-17A. The latter was largely confined to CTLA-4hi-expressing FOXP3hi-enriched CD25hiCD4 T cells that suppressed proliferation more weakly than did CTLA-4lo/−CD25hiFOXP3hi–enriched T cells. A causal link between high IL-17A production and impaired suppression of proliferation could not be demonstrated, however. Collectively, these results reveal a Gln synthetase–dependent increase and resilience of FOXP3hi cells under Gln restriction, and they demonstrate that impaired Gln-dependent nucleotide synthesis promotes FOXP3hi cells with regulator properties. It remains to be investigated to what extent the concomitant retention of IL-17A–producing CD4 T cells may limit the therapeutic potential of Tregs enriched through targeting these pathways in vivo.
Introduction
Regulatory T cells (Tregs) expressing the transcription factor FOXP3 are pivotal for balanced immunity, as revealed by the severe immune dysregulation and autoimmune pathology caused by FOXP3 loss-of-function mutations (1). Evidence of Treg-mediated protection in numerous preclinical models of autoimmunity and transplantation has sparked efforts to exploit their therapeutic potential for human therapy (2, 3). FOXP3+ Tregs may be of thymic origin or develop from mature peripheral T cells (4). Clinical Treg applications have largely relied on cellular therapies using in vitro–expanded pre-existing Tregs rather than induced Tregs due to the superior stability and more clearly defined functionality of the former (5, 6). If Tregs were promoted in vivo rather than supplemented through cellular therapy, it might be possible to tap the potential of multiple types of Tregs through targeting pathways that selectively favor them over T effector cells.
T cell subsets, including Tregs and T effectors, differentially use metabolic pathways to meet specific nutritional needs according to their function and activation state (7, 8). Insight into T cell subtype-specific metabolic requirements might be harnessed, therefore, to selectively promote Tregs over T effectors. Tregs are also preferentially induced or selectively advantaged in the presence of oxidative stress, in situations of general low energy as recapitulated by loss of signals from the metabolic hormone leptin, and thanks to their superior capacity to use preformed fatty acids when de novo synthesis is blocked (9–11). Collectively, these findings appear to indicate that Tregs may also be more resilient under metabolic stress.
Besides glucose, Gln is a critical and versatile metabolite in multiple pathways that sustain cellular integrity and the synthesis of macromolecules required for cell growth and division, as well as antiapoptotic mechanisms under metabolic stress (12). As Gln requirements often exceed cellular capacities for endogenous synthesis, Gln has been termed a conditionally essential amino acid. Accordingly, Gln is by far the most highly concentrated amino acid in serum (∼0.6 mM) and standard cell culture media (2 mM). Although the importance of Gln for activated T cells is well established, it has not been defined whether T cell subsets differ in their Gln requirements (13–15).
In the present study we have analyzed the consequences of Gln restriction for the development of human FOXP3hi and FOXP3lo/−CD4 T cells during activation in vitro. Limiting Gln favored FOXP3hi cells by diminishing FOXP3lo/−CD4 T cell numbers and proliferation, and through an increase in FOXP3hi cell numbers. Low Gln-induced constraints on proliferation and cell numbers were more closely mimicked by targeting the purine and pyrimidine synthesis pathways, respectively, and the absolute increase in FOXP3hiCD4 T cell numbers between 0.2 and 0.4 mM Gln was abrogated by inhibiting Gln synthetase (GS), suggesting a superior capacity of activated FOXP3hi cells to produce endogenous Gln under limiting extracellular Gln.
CD25hiCD4 T cells enriched in FOXP3hi cells through activation under low Gln or purine/pyrimidine pathway inhibition impaired the proliferation of primary T cells, but they also produced inflammatory cytokines, including IL-17A. IL-17A producers were predominantly CTLA-4hi cells. Although IL-17A production coincided with weaker suppression by FOXP3hi-enriched CTLA-4hi over CTLA-4lo/− subpopulations, neutralizing IL-17A with the former and adding IL-17A to the latter did not change their inhibitory capacities with respect to in vitro proliferation. Taken together, these results indicate that restricting Gln or Gln-dependent nucleotide synthesis promotes functionally suppressive human FOXP3hi T cells, yet it also spares IL-17A–producing CD4 T cells.
Materials and Methods
Human PBMC
PBMC were isolated from buffy coats (blood donation foundation SRK Aargau-Solothurn Kantonsspital Aarau AG and blood donation service SRK Bern AG) by density gradient using Ficoll-Paque plus (GE Healthcare) in Leucosep tubes (Greiner Bio-One). Residual RBC were removed using RBC lysis buffer (Amimed). CD4+CD25− T cells were enriched from freshly isolated PBMC using EasySep CD4+ T cell enrichment and CD25 depletion kits (Stemcell Technologies). The purity of the enriched cells was >95%.
Cell culture media and reagents
For experiments on Gln restriction, culture medium consisted of Gln-free RPMI 1640 (Lonza, BioWhittaker) supplemented with varying concentrations of l-glutamine (Sigma-Aldrich), low TGF-β1 (0.2 ng/ml, R&D Systems), and 5 ng/ml IL-2 (R&D Systems), a concentration determined in pilot studies to be suboptimal for promoting FOXP3hi T cells in the absence of additional supporting conditions. For all other assay conditions, RPMI 1640 with GlutaMAX (Life Technologies) was used. Treg suppressor assays were not supplemented with TGF-β and IL-2. All media were supplemented with 10% heat-inactivated FCS (PAA Laboratories) that was dialyzed through 3.5-kDa size-exclusion membranes. This reduced the Gln in FCS from 782 to 46 μM, as determined by liquid chromatography–mass spectrometry, thereby adding another 4.6 μM Gln to the indicated Gln concentrations. Methionine sulfoximide (MSO), dimethyl α-ketoglutarate, glutamate, and mycophenolic acid were from Sigma-Aldrich. N-phosphonacetyl-l-aspartate (PALA) was from the Novartis compound archive. Abs included purified anti-human CD28 (clone CD28.2) and fluorochrome-labeled CD4 (clone RPA-T4) and CD25 (clone M-A251 and clone 2A3), which were from BD Biosciences. Fluorochrome-labeled anti-human FOXP3 (clone PCH101) and CTLA-4 (clone L3D10) were from eBioscience; fluorochrome-labeled anti–IL-17A (clone eBIO64DEC17) was from BioLegend. Purified anti-human CD3 (clone OKT3) was provided by Novartis. Cell tracer dye CFSE and anti-CD3/CD28–coated Dynabeads (type 11129D) were from Life Technologies.
Small interfering RNA nucleofection
PBMC or CD4+CD25− T cells were nucleofected with 300 nM small interfering RNA (siRNA) using P3 Primary Cell 4D-Nucleofector kit with an Amaxa 4D-Nucleofector device according to the manufacturer’s protocol (Lonza). siRNAs were from GE Dharmacon and included human carbamoyl phosphate synthetase/aspartate transcarbamylase/dihydroorotase (CAD; E-009471-00-0005), glutamine phosphoribosylpyrophosphate amidotransferase (PPAT; E-006003-00-0005), and nontargeting SMARTpool siRNA (D-001910-01-05, no. 1). Nucleofected cells were rested for 2 h at 37°C in culture medium. Expression knockdown was confirmed by real-time quantitative PCR (RT-qPCR).
Total RNA extraction and RT-qPCR
Freshly harvested cells were lysed and their RNA was extracted according to the manufacturer’s protocols using RNeasy Mini or Micro kits (Qiagen). cDNAs were generated from 200 ng isolated RNA using the high-capacity cDNA reverse transcription kit from Life Technologies. To analyze gene expression, RT-qPCR was performed in triplicate according to the TaqMan Master Mix protocol and run on the QuantStudio 12K Flex (Life Technologies). TaqMan primers and probes were from Life Technologies and included CAD (assay ID Hs00983188_m1), PPAT (assay ID Hs00601264_m1), and 18S rRNA (assay ID 4319413E). The amounts of mRNA expression were normalized to 18S rRNA and calculated according to the comparative cycle threshold (Ct) method (16).
Cell cultures
For Gln restriction experiments, freshly isolated PBMC were used without further treatment or stained with 1 μM CFSE according to the manufacturer’s protocol. Two to 2.4 × 106 cells per well were plated in 24-well flat-bottom plates in medium containing specified l-glutamine concentrations. In experiments with small molecule inhibitors, cells were preincubated with inhibitors at 37°C for 30 min before T cell activation was started with soluble 0.03 μg/ml anti-CD3 and 0.3 μg/ml anti-CD28. siRNA-transfected cells were activated in full 2 mM Gln-containing medium under the same culture conditions. After 4 d, cells were harvested and analyzed by flow cytometry for CD4, CD25, and FOXP3 expression. When cells were expanded to be tested as Tregs in functional assays, they were split after 3 d and grown for another 3 d.
Flow cytometry and cell sorting
Cells were surface stained with fluorochrome-labeled Abs using standard procedures (30 min at 4°C in PBS, 2% FCS, 2 mM EDTA, 0.1% NaN3). For intracellular staining of FOXP3, CTLA-4, and cytokines, buffers and reagents from eBioscience were used according to the manufacturer’s instructions. Data were acquired on an LSR II flow cytometer and analyzed using FlowJo analysis software (Tree Star). Total cell numbers were determined from acquired cell numbers within the live forward/side scatter gate and fixed numbers of beads (CompBeads, BD Biosciences) added to each sample prior to FACS acquisition. To enrich FOXP3hi cells for functional assays, cells were surface stained for CD4, CD25, and CTLA-4 and sorted on a FACSAria Fusion (BD Biosciences) using a 70-μm ceramic nozzle with a sheath pressure of 70 psi and an acquisition rate of 12,000 events/s. Gated CD4+ cells were sorted based on CD25hi or CD25hiCTLA-4hi or lo/− subgates as depicted in the figures.
Suppressor assay
CD4+CD25− CFSE-labeled autologous T responder cells (Tresp; 3 × 104) were cocultured with sorted cells obtained from activation cultures under FOXP3hi T cell–promoting conditions and activated with Dynabeads at a 1:2 bead/total cell ratio in round-bottom 96-well plates. After 4–5 d, supernatants and cells were harvested. Tresp were analyzed by flow cytometry for CFSE staining intensity, and in some experiments also for intracellular cytokines. To detect intracellular cytokines, cells were restimulated for 4 h with PMA and ionomycin in the presence of brefeldin A (Leukocyte Activation Cocktail, BD Biosciences). Cytokines in supernatants were measured by a multiplex electrochemoluminescence assay (Meso Scale Discovery) or ELISA (eBioscience) kit. To test the role of IL-17A during the suppressor assay, a neutralizing anti–IL-17A Ab (clone MAB317; R&D Systems) or isotype control (MAB004; R&D Systems) was added at the beginning of the assay at 1.5 μg/ml. Recombinant human IL-17A (Novartis Institutes for BioMedical Research) was added at 40 ng/ml.
Results
Gln restriction during T cell activation favors FOXP3hi cells
Limiting Gln during the activation of human PBMC with anti-CD3/CD28 consistently enriched high FOXP3-expressing CD4 T cells (Fig. 1A). Activated (CD25+) CD4 T cells from all donors gradually increased in FOXP3hi cells as Gln concentrations were reduced (Fig. 1B), although exact dose responses could vary between donors (Fig. 1C).
Gln restriction during T cell activation favors FOXP3hi cells. (A) Representative FOXP3 and CD25 expression of gated CD4+ cells following 4 d activation of PBMC with anti-CD3/CD28 in medium containing 2 or 0.02 mM Gln. (B) Scatter points represent individual data from different experiments depicting percentage FOXP3hi cell within CD25+CD4 T cells. Data are pooled from 13 independent experiments (13 donors). Horizontal bars and error bars indicate average values and SEM. Data were analyzed by one-way ANOVA. *p < 0.05, ****p < 0.0001. (C) Two representative dose responses.
Gln restriction during T cell activation favors FOXP3hi cells. (A) Representative FOXP3 and CD25 expression of gated CD4+ cells following 4 d activation of PBMC with anti-CD3/CD28 in medium containing 2 or 0.02 mM Gln. (B) Scatter points represent individual data from different experiments depicting percentage FOXP3hi cell within CD25+CD4 T cells. Data are pooled from 13 independent experiments (13 donors). Horizontal bars and error bars indicate average values and SEM. Data were analyzed by one-way ANOVA. *p < 0.05, ****p < 0.0001. (C) Two representative dose responses.
Glutamate or α-ketoglutarate does not reverse the FOXP3hi cell–enriching effect of Gln restriction
We subsequently asked what Gln-consuming pathways could account for the different Gln requirements of FOXP3lo/−CD25+ and FOXP3hi cells (Fig. 2). Gln, upon conversion to α-ketoglutarate via glutamate, is a key substrate to replenish TCA cycle metabolites and support NADPH generation for macromolecular synthesis during cell growth and proliferation (17, 18). If these pathways contributed to FOXP3hi cell enrichment through Gln restriction, then providing extra α-ketoglutarate or glutamate should at least partially compensate for Gln to reverse this effect. This was not the case, however. Supplementing media with α -ketoglutarate (in its cell-permeable form dimethyl-α-ketoglutarate) did not compromise FOXP3hi cell enrichment under limiting Gln (Fig. 3A). At concentrations >2 mM, excess dimethyl-α-ketoglutarate appeared to be generally cytoreductive or toxic in these cell cultures (not shown). Extra glutamate did not change overall FOXP3hi cell proportions (Fig. 3B). In some experiments, glutamate supplements moderately reduced total cell numbers of both FOXP3hi and FOXP3lo cells without changing their proportions (Fig. 3B). Taken together, these data suggest that Gln availability for these pathways does not substantially influence the balance between FOXP3hi and FOXP3lo/−CD25+ cells upon T cell activation in this model.
Gln-dependent metabolic pathways discussed in this report. Circled plus sign (+) indicates addition of metabolites, red bars indicate inhibition.
Gln-dependent metabolic pathways discussed in this report. Circled plus sign (+) indicates addition of metabolites, red bars indicate inhibition.
Excess glutamate or α-ketoglutarate do not reverse the selective advantage of FOXP3hi cells under Gln restriction. PBMC were activated with anti-CD3/CD28 in different concentrations of Gln and 2mM dimethyl α-ketoglutarate (A) or 2–4 mM extra glutamate (B). FOXP3 and CD25 expression on CD4 T cells was determined by flow cytometry after 4 d. Data are representative of three independent experiments.
Excess glutamate or α-ketoglutarate do not reverse the selective advantage of FOXP3hi cells under Gln restriction. PBMC were activated with anti-CD3/CD28 in different concentrations of Gln and 2mM dimethyl α-ketoglutarate (A) or 2–4 mM extra glutamate (B). FOXP3 and CD25 expression on CD4 T cells was determined by flow cytometry after 4 d. Data are representative of three independent experiments.
Gln restriction more strongly reduces the proliferation and cell numbers of activated FOXP3lo/− than FOXP3hiCD4 T cells
Earlier studies had demonstrated a nonredundant role of Gln for T cell proliferation that could not be replaced by glutamate or other amino acids (13, 15). We next asked how Gln restriction affected the proliferation of activated FOXP3lo/− and FOXP3hiCD4 T cells. The CD25+FOXP3lo/− population progressively lost cell numbers and cell cycles as Gln concentrations declined (Fig. 4, left). FOXP3hi cells usually underwent one cell cycle less than did FOXP3lo/− cells at high Gln; they were not constrained by Gln reductions down to 0.4 mM Gln and only weakly at 0.2 mM. Furthermore, FOXP3hi cell numbers were consistently higher at intermediate than at high Gln concentrations (Fig. 4, right).
Gln restriction more strongly reduces the proliferation and numbers of activated FOXP3lo/− than FOXP3hiCD4 T cells. Cell divisions were determined by loss of CFSE fluorescence intensity as illustrated in the histograms. Numbered markers within histograms indicate numbers of cell divisions. Data points in line graphs are means of replicates that differed by ∼10–15%. Data are representative of nine independent experiments.
Gln restriction more strongly reduces the proliferation and numbers of activated FOXP3lo/− than FOXP3hiCD4 T cells. Cell divisions were determined by loss of CFSE fluorescence intensity as illustrated in the histograms. Numbered markers within histograms indicate numbers of cell divisions. Data points in line graphs are means of replicates that differed by ∼10–15%. Data are representative of nine independent experiments.
Inhibition of pyrimidine and purine de novo synthesis favors FOXP3hi cells
Gln is the main nitrogen source for de novo nucleotide synthesis in animals (Fig. 2). If skewing toward FOXP3hi cells under low Gln depended on impaired pyrimidine and/or purine de novo synthesis, then inhibiting these pathways should promote FOXP3hi cells similarly to Gln reduction. We initially tested this possibility with small molecule inhibitors.
PALA selectively blocks the trifunctional Gln-consuming enzyme CAD that catalyzes the first three reactions of the de novo pyrimidine synthesis (19, 20). The first committed Gln-consuming rate-limiting step of the purine de novo synthesis is catalyzed by PPAT. Because there is no selective PPAT inhibitor we initially used mycophenolic acid (MPA), a selective inhibitor of inositol monophosphate dehydrogenases 1 and 2 (IMPDH1/2). Its short-lived product, XMP, is the substrate for, and determines the rate of, Gln-dependent GMP synthesis (Fig. 2).
PALA or MPA added to full 2 mM Gln during T cell activation increased FOXP3hi cells to levels otherwise achieved through Gln restriction (Fig. 5). The consequences for cell divisions and cell numbers, however, could differ between pyrimidine and purine inhibition, as well as between FOXP3lo/−CD25+ and FOXP3hi cells. In the experiment depicted in Fig. 5A, 100 nM MPA enriched FOXP3hi cells similar to 0.2–0.4 mM Gln; 100 nM MPA recapitulated FOXP3lo/−CD25+ cell numbers and divisions at 0.4 mM, whereas it more closely resembled FOXP3hi cell divisions at 0.2 mM and FOXP3hi cell numbers at 0.4 mM Gln. FOXP3hi cell enrichment with 100 μM PALA was comparable to 0.2 mM Gln, but proliferation was less strongly reduced with PALA for both FOXP3lo/−CD25+ and FOXP3hi cells.
Inhibiting pyrimidine and purine synthesis during T cell activation favors FOXP3hi T cells. PBMC were activated in different concentrations of Gln and the pyrimidine and purine synthesis inhibitors PALA and MPA, respectively. (A) Bar graphs show FOXP3hi cell enrichment with titrations of Gln, and with either PALA or MPA titrations in full 2 mM Gln. Line graphs depict CFSE-based cell division analyses for gated FOXP3lo/−CD25+CD4+ cells and FOXP3hiCD25+CD4+ cells for 0.4 and 0.2 mM Gln and those inhibitor concentrations that resulted in similar FOXP3hi T cell enrichment. (B) Bar graphs show FOXP3hi cell enrichment with 2, 0.4, and 0.2 mM Gln, compared with concentrations of PALA or MPA alone and in combinations that resulted in FOXP3hi cell enrichment closest to 0.4 and 0.2mM Gln, for T cells from two different donors and independent experiments. Line graphs below indicate the corresponding patterns of cell numbers against cell divisions for FOXP3lo/−CD25+CD4+ T cells (a, c, e, and g) and FOXP3hi cells (b, d, f, and h).
Inhibiting pyrimidine and purine synthesis during T cell activation favors FOXP3hi T cells. PBMC were activated in different concentrations of Gln and the pyrimidine and purine synthesis inhibitors PALA and MPA, respectively. (A) Bar graphs show FOXP3hi cell enrichment with titrations of Gln, and with either PALA or MPA titrations in full 2 mM Gln. Line graphs depict CFSE-based cell division analyses for gated FOXP3lo/−CD25+CD4+ cells and FOXP3hiCD25+CD4+ cells for 0.4 and 0.2 mM Gln and those inhibitor concentrations that resulted in similar FOXP3hi T cell enrichment. (B) Bar graphs show FOXP3hi cell enrichment with 2, 0.4, and 0.2 mM Gln, compared with concentrations of PALA or MPA alone and in combinations that resulted in FOXP3hi cell enrichment closest to 0.4 and 0.2mM Gln, for T cells from two different donors and independent experiments. Line graphs below indicate the corresponding patterns of cell numbers against cell divisions for FOXP3lo/−CD25+CD4+ T cells (a, c, e, and g) and FOXP3hi cells (b, d, f, and h).
As T cells from different donors could vary in their dose responses to Gln, MPA, and PALA, and because inhibition of each purine and pyrimidine synthesis appeared to better reflect Gln restriction–induced inhibition of proliferation and constraints on FOXP3lo/−CD25+ cell numbers, respectively, further experiments included titrations of MPA and PALA alone and in combination. For T cells from two different donors, the bar graphs of Fig. 5B depict concentrations and combinations of MPA and PALA that induced FOXP3hi cell enrichment closest to 0.4 and 0.2 mM Gln, the Gln range that consistently resulted in an absolute increase or stable FOXP3hi cell numbers compared with standard 2 mM Gln.
For FOXP3hi cells from both donors, the increase at 0.4 mM Gln was best mimicked by MPA (Fig. 5Bb, 5Bd). For donor 2 FOXP3lo/−CD25+ T cells, inhibition of proliferation and numbers at 0.4 mM Gln were similar to MPA or PALA (Fig. 5Bc). An MPA/PALA combination closer to 0.4 mM Gln was not identified in this experiment. In contrast, donor 1 FOXP3lo/−CD25+ cell numbers and proliferation at 0.4 mM Gln were most closely recapitulated by combined MPA and PALA (Fig. 5Ba).
Matching cell numbers and divisions at 0.2 mM Gln required combinations of MPA and PALA for FOXP3lo/−CD25+ T cells from both donors (Fig. 5Be, 5Bg) and for FOXP3hi cells from donor 2 (Fig. 5Bh). FOXP3hi T cell divisions and numbers from donor 1at 0.2 mM were more closely matched by PALA between no and four divisions and by combined PALA and MPA between four and six divisions (Fig. 5Bf).
To validate and compare directly the importance of the rate-limiting Gln-consuming steps of de novo purine and pyrimidine synthesis for the FOXP3hi/FOXP3lo/−CD25+ cell balance, we specifically knocked down CAD and PPAT using stabilized selective siRNA pools. Upon T cell activation, CAD and PPAT were transiently upregulated with similar kinetics, and siRNA targeting reduced their expression by >80% in CD4 T cells or PBMC (Fig. 6A). In addition to CAD knockdown, PPAT knockdown also resulted in increased FOXP3hi cells, thereby confirming the relevance of de novo purine synthesis for FOXP3hi cell enrichment through inhibiting the first and rate-limiting Gln-dependent step (Fig. 6B).
siRNA-mediated knockdown of CAD or PPAT promotes FOXP3hi cells. (A) Activation-dependent expression of CAD and PPAT mRNA following nucleofection alone (gray bars) or siRNA transfection with purified CD4 T cells and PBMC. Bar graphs are means of triplicate PCR reactions that differed by 10–20%. (B) CD25 and FOXP3 expression of gated CD4 T cells transfected with nontarget siRNA, CAD siRNA, or PPAT siRNA and activated for 4 d. Data are representative of three independent experiments.
siRNA-mediated knockdown of CAD or PPAT promotes FOXP3hi cells. (A) Activation-dependent expression of CAD and PPAT mRNA following nucleofection alone (gray bars) or siRNA transfection with purified CD4 T cells and PBMC. Bar graphs are means of triplicate PCR reactions that differed by 10–20%. (B) CD25 and FOXP3 expression of gated CD4 T cells transfected with nontarget siRNA, CAD siRNA, or PPAT siRNA and activated for 4 d. Data are representative of three independent experiments.
Inhibition of GS abolishes FOXP3hi cell maintenance under Gln restriction
Cells may generate Gln through GS, a tightly controlled enzyme that is positively regulated by low Gln and nucleotide starvation (Fig. 2) (21, 22). It seemed conceivable that the resilience of activated FOXP3hi cells to Gln restriction was due to a superior capacity to generate endogenous Gln. This possibility was tested with the selective GS inhibitior MSO (23, 24). As with dose responses to Gln and other inibitiors, the sensitivity to MSO at different Gln concentrations could vary between donors. Two general consistent observations with all donors were, however, that MSO at full 2 mM Gln already substantially reduced FOXP3lo/−CD25+ but not FOXP3hi cell numbers, and that MSO always abolished the characteristic increase and maintenance of FOXP3hi cells at intermediate–low Gln. With activated T cells from donor 1, MSO reversed the increase of FOXP3hi cells between 2 and 0.2 mM Gln and caused a decline comparable to FOXP3lo/−CD25+ cells in the presence of MSO (Fig. 7A, left). The analysis of cell numbers per cell division revealed that MSO caused an overall drop in cell numbers per cell cycle without reducing cell divisions (Fig. 7B, left). Donor 2 FOXP3hi T cells peaked at 0.4 mM Gln, and this was reduced by MSO to FOXP3hi baseline levels at 2 mM Gln (Fig. 7A, right and Fig. 7B, upper right).
Inhibition of GS abolishes the FOXP3hi cell increase and resilience under Gln restriction. (A) Human PBMC were activated with anti-CD3/CD28 in medium containing different Gln concentrations, with or without the selective GS inhibitor MSO, used at 10 mM as determined in pilot titration experiments. FOXP3 and CD25 expression on CD4 T cells was measured by flow cytometry after 4 d. (B) Analysis of cell numbers and cell divisions based on CFSE staining intensity. Data are from two independent experiments using PBMC from two different donors.
Inhibition of GS abolishes the FOXP3hi cell increase and resilience under Gln restriction. (A) Human PBMC were activated with anti-CD3/CD28 in medium containing different Gln concentrations, with or without the selective GS inhibitor MSO, used at 10 mM as determined in pilot titration experiments. FOXP3 and CD25 expression on CD4 T cells was measured by flow cytometry after 4 d. (B) Analysis of cell numbers and cell divisions based on CFSE staining intensity. Data are from two independent experiments using PBMC from two different donors.
FOXP3hi-enriched CD25hi cells are functionally suppressive but also produce effector cytokines
We next aimed to test the suppressor functions of FOXP3hi cells that emerged under low Gln and through inhibiting nucleotide synthesis. Because we did not identify surface markers or marker combinations that would clearly distinguish between CD25+FOXP3lo/− and FOXP3hi cells (e.g., CD30, GITR, GARP, CD39, CD73, CD127; data not shown), we initially enriched FOXP3hi cells further by sorting the 10% highest CD25-expressing CD4 T cells. This resulted in ∼60–70% FOXP3hi-enriched CD25hi cells that strongly suppressed the proliferation of CD4 T cells (Tresp) when added in equal numbers and more weakly at <1:1 ratios (Fig. 8A). FOXP3hi-enriched T cells alone or cultured together with Tresp did not consistently produce elevated IL-10 (not shown) but did reproducibly secrete effector cytokines, including IL-17A, IL-4, and IFN-γ (Fig. 8B).
FOXP3hi cell–enriched CD25hiCD4+ cells from low Gln or PPAT siRNA (siPPAT) cultures are functionally suppressive but also produce effector cytokines. PBMC were activated with anti-CD3/CD28 in 0.02 mM Gln. In a separate experiment, PPAT siRNA–transfected PBMC were activated in 2 mM standard Gln. After 6 d the 10% highest CD25-expressing CD4+ cells were sorted to be tested for their suppressor potential in functional assays. Sorted CD25hi populations were 70% (for low Gln) and 60% (for siPPAT) FOXP3hi. (A) Proliferation of primary autologous CD4 T cells (Tresp) after 4 d activation alone or in cocultures with FOXP3hi-enriched cells added at different ratios. Histograms show representative CFSE staining; line graphs summarize changes in Tresp divisions. Data are means of triplicate cultures that differed by ≤10%. (B) Cytokine concentrations in supernatants harvested from cultures after 4 d. Bar graphs are means of triplicates that differed by ∼5–15%. The low Gln experiment is one of two experiments with similar results; for FOXP3hi-enriched cells generated via activation upon PPAT knockdown, only one experiment was performed using a general CD25hi sort gate without further subgating.
FOXP3hi cell–enriched CD25hiCD4+ cells from low Gln or PPAT siRNA (siPPAT) cultures are functionally suppressive but also produce effector cytokines. PBMC were activated with anti-CD3/CD28 in 0.02 mM Gln. In a separate experiment, PPAT siRNA–transfected PBMC were activated in 2 mM standard Gln. After 6 d the 10% highest CD25-expressing CD4+ cells were sorted to be tested for their suppressor potential in functional assays. Sorted CD25hi populations were 70% (for low Gln) and 60% (for siPPAT) FOXP3hi. (A) Proliferation of primary autologous CD4 T cells (Tresp) after 4 d activation alone or in cocultures with FOXP3hi-enriched cells added at different ratios. Histograms show representative CFSE staining; line graphs summarize changes in Tresp divisions. Data are means of triplicate cultures that differed by ≤10%. (B) Cytokine concentrations in supernatants harvested from cultures after 4 d. Bar graphs are means of triplicates that differed by ∼5–15%. The low Gln experiment is one of two experiments with similar results; for FOXP3hi-enriched cells generated via activation upon PPAT knockdown, only one experiment was performed using a general CD25hi sort gate without further subgating.
High CTLA-4 expression identifies IL-17A and IL-4 producers among CD4CD25hi- enriched FOXP3hi cells
We next asked whether effector cytokine production could be separated from suppressor functions. The pivotal checkpoint inhibitor CTLA-4 was shown to be highly expressed by human Th17 cells and murine Th2 cells (25–27). Although CTLA-4 is more prominently known as a marker and functional mediator of Tregs, CTLA-4–mediated regulation does not depend on its expression by FOXP3+ Tregs, and CTLA-4–independent Treg-mediated suppression has been reported for in vitro models (28). When we sorted CD25hi cells as separate CTLA-4lo/− and CTLA-4hi subpopulations, the latter was more strongly enriched in FOXP3hi cells (Fig. 9A). CTLA-4hiCD25hi cells, despite larger proportions of FOXP3hi cells, inhibited more weakly than did CTLA-4lo/−FOXP3hi–enriched T cells, and not at all with cells from one donor (Fig. 9B). CTLA-4lo/−FOXP3hi–enriched T cells from CAD siRNA nucleofected cultures (siCAD) and PPAT siRNA nucleofected cultures (siPPAT) consistently shifted Tresp proliferation toward fewer cell cycles (Fig. 9B).
High CTLA-4 expression identifies IL-17A and IL-4 producers whereas CTLA-4loCD25hiFOXP3–enriched cells are also suppressive. PBMC were transfected with nontarget (NT), CAD, or PPAT siRNA and activated with anti-CD3/CD28. After 6 d, cells were surface stained for sorting based on CD4, CD25, and surface CTLA-4 expression. (A) Sort gates depict CTLA-4hi and CTLA-4lo/− gates for the highest CD25-expressing CD4+ cells. The table lists the postsort FOXP3hi-enriched cells used to generate the data in (B)–(D). (B) Proliferation of anti-CD3/CD28 bead–activated Tresp as analyzed by CFSE fluorescence intensities. Tresp were cultured alone or with equal numbers of CTLA-4hiCD25hi or CTLA-4lo/−CD25hiFOXP3hi–enriched CD4 T cells. Top, Representative histograms. Bottom, Cell division patterns, showing means of triplicate cultures that differed by ≤10%. (C) IL-17A, IL-4, and IFN-γ in supernatants from Tresp, FOXP3hi-enriched CD25hiCD4 T cells, and cocultures. Bar graphs are means of triplicates that differed by ∼5–20%. Data are from two different donors and independent experiments. (D) Dot plots depict FOXP3 and IL-17A expression of gated Tresp and FOXP3hi-enriched cells from a representative 4 d coculture of donor 2 Tresp with CTLA-4hi siPPAT FOXP3hi–enriched cells. (E) Data were generated as in (A)–(D), with the addition of including a neutralizing anti–IL-17A or isotype control (ctrl.) Ab in cocultures with CTLA-4hiCD25hiFOXP3hi–enriched cells (both at 1.5 μg/ml), or including recombinant human IL-17A at 40 ng/ml in cocultures with CTLA-4lo/–CD25hiFOXP3hi–enriched cells. IL-17A concentrations were measured in supernatants from cultures without added IL-17A reagents.
High CTLA-4 expression identifies IL-17A and IL-4 producers whereas CTLA-4loCD25hiFOXP3–enriched cells are also suppressive. PBMC were transfected with nontarget (NT), CAD, or PPAT siRNA and activated with anti-CD3/CD28. After 6 d, cells were surface stained for sorting based on CD4, CD25, and surface CTLA-4 expression. (A) Sort gates depict CTLA-4hi and CTLA-4lo/− gates for the highest CD25-expressing CD4+ cells. The table lists the postsort FOXP3hi-enriched cells used to generate the data in (B)–(D). (B) Proliferation of anti-CD3/CD28 bead–activated Tresp as analyzed by CFSE fluorescence intensities. Tresp were cultured alone or with equal numbers of CTLA-4hiCD25hi or CTLA-4lo/−CD25hiFOXP3hi–enriched CD4 T cells. Top, Representative histograms. Bottom, Cell division patterns, showing means of triplicate cultures that differed by ≤10%. (C) IL-17A, IL-4, and IFN-γ in supernatants from Tresp, FOXP3hi-enriched CD25hiCD4 T cells, and cocultures. Bar graphs are means of triplicates that differed by ∼5–20%. Data are from two different donors and independent experiments. (D) Dot plots depict FOXP3 and IL-17A expression of gated Tresp and FOXP3hi-enriched cells from a representative 4 d coculture of donor 2 Tresp with CTLA-4hi siPPAT FOXP3hi–enriched cells. (E) Data were generated as in (A)–(D), with the addition of including a neutralizing anti–IL-17A or isotype control (ctrl.) Ab in cocultures with CTLA-4hiCD25hiFOXP3hi–enriched cells (both at 1.5 μg/ml), or including recombinant human IL-17A at 40 ng/ml in cocultures with CTLA-4lo/–CD25hiFOXP3hi–enriched cells. IL-17A concentrations were measured in supernatants from cultures without added IL-17A reagents.
IL-17A and IL-4 were predominantly produced by CTLA-4hi FOXP3hi–enriched T cells cultured alone or in cocultures with Tresp, whereas IFN-γ and other cytokines such as TNF-α and IL-13 were more variably secreted by both CTLA-4lo/− and CTLA-4hi FOXP3hi-enriched T cells (Fig. 9C, Supplemental Fig. 1).
Cocultures with Tresp mostly contained higher cytokine concentrations than did CTLA-4hiFOXP3hi−–enriched T cells alone, even though cytokine production by Tresp alone is barely visible on the y-axis scales in Fig. 9C.
To identify cytokine producers in cocultures, intracellular cytokine staining was performed. This revealed prominent IL-17A+ populations among T cells originating from CTLA-4hiFOXP3hi–enriched T cells but few or none among Tresp from either single cultures or cocultures (Fig. 9D). Taken together, these data suggest that enhanced cytokine production in Tresp/FOXP3hi-enriched T cell cocultures over FOXP3hi-enriched T cells alone was attributable to Tresp-aided increased cytokine production by FOXP3hi-enriched T cells. IL-17A–producing cells were detected among FOXP3lo/− cells, but also among the small proportions of sorted FOXP3hi-enriched T cells that remained FOXP3hi at the end of the suppressor assay (Fig. 9D).
Given the correlation between high IL-17A production and reduced suppressor capacity of CTLA-4hi over CTLA-4lo/− FOXP3hi-enriched T cells, we interrogated the functional role of IL-17A for inhibition of Tresp proliferation in these assays. A neutralizing anti–IL-17A Ab was added to cocultures of Tresp with CTLA-4hiCD25hiFOXP3hi–enriched cells and rIL-17A was included at 40 ng/ml in cocultures of Tresp with CTLA-4lo/−CD25hiFOXP3hi–enriched cells (Fig. 9E). However, these reagents, although validated for their biological activity in other functional assays (data not shown), had no influence on the inhibition of Tresp proliferation (Fig. 9E).
Discussion
Gln is an abundant, versatile metabolic substrate in high demand by activated T cells. In this study, we show that limiting Gln during T cell activation induces a profound shift toward high FOXP3–expressing CD4 T cells. We have investigated the underlying cellular changes, identified corresponding Gln-dependent metabolic pathways, and demonstrated regulatory properties of FOXP3hi-enriched CD4 T cells emerging from T cell activation under Gln restriction and through pathway inhibition.
Cellular activation may enhance the flux of Gln through reactions that generate energy, metabolites, and NADPH for the biosynthesis of macromolecules required for cell growth and proliferation. These Gln functions may be replaced by glutamate or α-ketoglutarate (17, 18). In a recently published study using murine T cell cultures, a proportional increase in Foxp3+ T cells through Gln deprivation was reversed by α-ketoglutarate supplements (29). We had generated similar data with murine T cells (our unpublished data), but never with human T cells. It remains to be investigated whether these discrepancies reflect real differences in the T cell biology of mice and humans, or whether the different T cell sources, that is, spleen and lymph nodes versus blood, could account for these differences.
As an excess supply of the Gln-derived metabolites glutamate and α-ketoglutarate did not prevent the contraction of activated FOXP3lo/−CD4 T cells and the shift toward FOXP3hi cells under low Gln in our human T cell cultures, this implied a role for limiting Gln-derived amide rather than the Gln carbon scaffold. Furthermore, the tight correlation between gradually reduced Gln and CD25+FOXP3lo/− cell divisions recapitulated earlier evidence of a nonredundant role of Gln for T cell proliferation, suggesting insufficient Gln-derived amide for nucleotide biosynthesis (13, 15). This was supported by the ability to reproduce the effects of Gln restriction by inhibiting Gln-dependent de novo purine and pyrimidine synthesis. The purine synthesis inhibitor MPA often more closely matched Gln deprivation–induced inhibition of proliferation while leaving higher cell numbers, whereas the pyrimidine inhibitor PALA more strongly reduced T cell numbers.
Lower numbers of activated FOXP3lo/−CD4 T cells per cell division possibly resulted from apoptosis. Although not measured in our experiments, earlier studies identified apoptosis as a consequence of human T cell activation in the presence of pyrimidine inhibitors whereas purine inhibitors primarily impaired cell cycle progression (30). We found that the increase and resilience of FOXP3hiCD4 T cells under Gln restriction was abolished by blocking GS. GS inhibition did not reduce cell cycles but selectively affected cell numbers, similar to inhibiting pyrimidine synthesis. GS was previously shown to selectively support pyrimidine and not purine synthesis, and pyrimidine rather than purine synthesis was required to protect cells from inflammation-induced apoptosis (31, 32). Inflammation and tissue injury are physiological conditions of Gln reduction, and GS is de-repressed under low Gln, nucleotide starvation, and by glucocorticoid hormones (22, 33–35). Collectively, these findings suggest that Gln synthesized by GS in stressful situations is primarily used for pyrimidine synthesis, and is more important for cell survival than cell proliferation.
The different consequences of GS inhibition at full or suboptimal Gln for activated FOXP3lo/− and FOXP3hiCD4 T cells raise questions about the molecular mediators behind FOXP3-associated differential “wiring” between extracellular Gln and GS activity. Although entirely speculative at present, these might include FOXO transcription factors previously shown to support GS expression in skeletal muscle and cell lines, as well as FOXP3 expression in T cells (36–38). The extensive regulation of FOXOs includes feedback loops to control their own transcription and activation as well as FOXP3 within the PTEN/PI3K/Akt/mTOR pathway (38, 39). Inhibition of mTOR favors Tregs, and factors that control mTOR activity include Gln import through the transporter SLC1A5 (40, 41). Although it seemed possible that SLC1A5 inhibition would recapitulate FOXP3hi cell promotion through Gln restriction, we found that siRNA knockdown of SLC1A5 diminished activated T cells without favoring FOXP3hi T cells, and it generally reproduced the functional profile previously published for SLC1A5 knockout mice but not the consequences of Gln restriction in our model (our unpublished data) (42). More studies are needed to identify nodes that connect Gln, GS, FOXP3, and possibly FOXO and/or other factors with T cell fate and function.
Gln concentrations in vivo may drop transiently during exercise and inflammation, thus raising questions about the stability and function of FOXP3hiCD4 T cells emerging under such temporary metabolic stress. FOXP3hi cells enriched through Gln restriction or pathway inhibition reduced the proliferation of primary T cells, although high FOXP3 expression was largely lost during the 4-d suppressor assay. This demonstrated that high FOXP3 expression was stable only for as long as these metabolic constraints persisted and implied that a limited period of suppression following coculture of FOXP3hi-enriched T cells with Tresp was sufficient to reduce Tresp cycle numbers. This would be in line with previous studies demonstrating in a mouse model that the first 6–12 h of Treg/Tresp coculture were necessary and sufficient to inhibit Tresp proliferation (43).
Because CTLA-4hiCD25hiCD4 T cells were somewhat more strongly enriched in FOXP3hi cells, we might have expected them to be stronger suppressors for this reason but not due to their higher CTLA-4 expression, as CTLA-4–mediated inhibition was shown to operate via cell-extrinsic mechanisms through the shared CD28/CTLA-4 ligands CD80/CD86 that were not specifically provided in our assays (44). Despite their moderately larger FOXP3hi cell enrichment, CTLA-4hiCD25hi cells reduced Tresp cycles less strongly than did CTLA-4lo/−CD25hi cells. The former were the predominant producers of IL-17A and IL-4, whereas other cytokines, including IFN-γ, TNF-α, and IL-13, could be similarly produced by CTLA-4lo/−CD25hi cells.
Given the possible associations between inferior Treg functions during some autoimmune conditions, including those with IL-17A components, and in view of the high IL-17A concentrations produced by CTLA-4hiCD25hi T cells during the suppressor assays, we speculated that IL-17A might be associated with inferior inhibition of proliferation in these assays. This was not the case, however. A neutralizing anti–IL-17A Ab did not improve inhibition by CTLA-4hiCD25hi cells, nor did addition of IL-17A affect suppression by CTLA-4lo/−CD25hi cells. Although the latter could be explained by a failure to respond to IL-17A, the former would have been expected to remove a potential causal link between the presence of IL-17A and inferior inhibition of proliferation by CTLA-4hiCD25hi cells. Generally, our data seem to be in line with previous studies showing full suppression by a subset of IL-17A–producing human ex vivo isolated Tregs, although IL-17A concentrations in these studies were, at ∼0.5 ng/ml, ∼20- to 100-fold lower than those measured in our assays (45, 46). IL-17A producers in our suppression assays could have included FOXP3hi cells, former FOXP3hi cells that lost FOXP3 expression, or differentiated Th17 cells, with all possibilities having been demonstrated previously, including human and clinical settings (25, 47–50).
Unexpectedly, cytokine production was often higher in cocultures of CTLA-4hiCD25hiCD4 T cells with Tresp than in CTLA-4hiCD25hiCD4 single cultures, regardless of whether Tresp proliferation was inhibited, and even though Tresp did not substantially contribute to cytokine production. This suggests that primary and resting CD4 T cells enhanced cytokine production by preactivated T effector/memory cells, an interesting phenomenon that demands further analysis and mechanistic understanding to evaluate its physiological relevance.
Because Gln can be limiting under some physiological conditions, as outlined above, our studies may reflect aspects of immune modulation in vivo. A shift toward FOXP3hiCD4 T cells capable of curbing the proliferation of formerly naive or resting T cells may help to restrain productive (destructive) immunity in the absence of detrimental inflammation, help to avoid prolongation and spreading of an immune response, and prevent an acute immune response from turning chronic.
It is not entirely clear at present how (and if) CTLA-4hi IL-17A–producing T cells could interfere with Treg functions in vivo. However, observations of seemingly therapeutically ineffective Foxp3+CD4 T cells accumulating in inflamed tissues, also in association with IL-17A, suggest that further insight into the effects of IL-17A on Treg functions might help to harness the therapeutic potential of Tregs in IL-17A–driven diseases (49, 51).
FOXP3hiCD4 T cells maintained under or emerging from activation under low Gln or during impaired de novo purine/pyrimidine syntheses may presumably be suppressive in vivo, both under desirable and undesirable circumstances. Our data might help to understand, for example, why functional Tregs are well maintained in mycophenolate mofetil–treated transplant patients and might further hint at Treg-compatible immunosuppressive treatments (52). On the opposite side of the coin, cancer drugs targeting these pathways could possibly have a suboptimal therapeutic effect in some cases (53, 54). Furthermore, our results might help to explain why Tregs may thrive well within tumors if these are addicted to Gln or, similar to activated FOXP3hiCD4 T cells in our studies, are also resistant to Gln deprivation through utilizing GS (55).
Acknowledgements
We thank Franco Di Padova, Friedrich Raulf, and Eva D’Hennezel for critically commenting on the manuscript. We are also grateful to Juan Zhang for analyzing FCS, to Marie-Jo Duriatti for advice on IL-17A assay reagents, and to Friedrich Raulf’s group and Eva D’Hennezel for technical advice on the siRNA technology.
Footnotes
The online version of this article contains supplemental material.
Abbreviations used in this article:
- CAD
carbamoyl phosphate synthetase/aspartate transcarbamylase/dihydroorotase
- GS
glutamine synthetase
- MPA
mycophenolic acid
- MSO
methionine sulfoximide
- PALA
N-phosphonacetyl-l-aspartate
- PPAT
glutamine phosphoribosylpyrophosphate amidotransferase
- RT-qPCR
real-time quantitative PCR
- siRNA
small interfering RNA
- Treg
regulatory T cell
- Tresp
T responder cell.
References
Disclosures
All authors are employees of Novartis, Switzerland.