It is now well established that interaction of PGD2 with chemoattractant receptor- homologous molecule expressed on Th2 cells (CRTH2) promotes chemotaxis and proinflammatory cytokine production by Th2 lymphocytes. In this study we show a novel function of CRTH2 in mediating an inhibitory effect of PGD2 on the apoptosis of human Th2 cells induced by cytokine deprivation. This effect was mimicked by the selective CRTH2 agonist 13,14-dihydro-15-keto-PGD2, inhibited by the CRTH2 antagonists ramatroban and TM30089, and not observed in CRTH2-negative T cells. D prostanoid receptor 1 (DP1) or the thromboxane-like prostanoid (TP) receptor did not play a role in mediating the effects of PGD2 on the apoptosis of Th2 cells because neither the DP1 antagonist BW868C nor the TP antagonist SQ29548 had any effect on the antiapoptotic effect of PGD2. Apoptosis of Th2 cells induced by Fas ligation was not suppressed by treatment with PGD2, illustrating that activation of CRTH2 only inhibits apoptosis induced by cytokine deprivation. Treatment with PGD2 induced phosphorylation of Akt and BAD, prevented release of cytochrome c from mitochondria, and suppressed cleavage of caspase-3 and poly(ADP-ribose) polymerase in Th2 cells deprived of IL-2. The PI3K inhibitor LY294002 blocked the effect of PGD2 both on the signaling events and on the apoptotic death of Th2 cells. These data suggest that in addition to promoting the recruitment and activation of Th2 cells, PGD2 may also impede the resolution of allergic inflammation through inhibiting apoptosis of Th2 cells.

Prostaglandin D2 is the major prostanoid released from mast cells (1) and is also produced in substantial quantities by Th2 lymphocytes (2, 3). It has been detected in high concentrations at sites of allergic inflammation and mediates its effects through high affinity interactions with D prostanoid receptor (DP)2 1 (2) and chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2; also named DP2). DP1 is expressed by vascular smooth muscle and some leukocyte populations, and activation of this receptor leads to increased blood flow, consequent congestion in allergic tissue (4, 5), and modulation of both eosinophil and basophil functions (6, 7, 8). Through activation of CRTH2, PGD2 promotes the migration of eosinophils, basophils, and Th2 cells (6, 9, 10) and has the unusual ability to stimulate Th2 cytokine production (11) even in the absence of allergen recognition or costimulation (12). Initial studies with CRTH2 knockout mice (13) suggested that CRTH2 might constrain Th2 cytokine production and promote Th1 cytokine production, but this is likely to be due to CRTH2 expression by Th1 cells in mice and the particular immunization procedures used. More recent studies with selective CRTH2 antagonists and genetically deficient mice have highlighted an important role for CRTH2 in promoting the accumulation of lymphocytes and eosinophils (14, 15) and the production of Th2 cytokines (16) and IgE (14, 16) at sites of allergic inflammation. The PI3K and calcineurin pathways are involved in mediating the responses of Th2 cells to CRTH2 activation (17). It is well known that, in addition to its roles in cell motility and activation (18), the PI3K pathway is critical in regulating T cell survival and proliferation (19, 20). T cell survival and death, especially apoptosis, are believed to be important for lymphocyte homeostasis and self-tolerance in the immune system (21, 22). Too little cell death of activated lymphocytes can result in autoimmune or allergic disorders, whereas too much apoptosis can lead to immunodeficiency. We have therefore investigated the role of CRTH2 in the apoptosis of Th2 cells that may be relevant to the pathogenesis of asthma where a defect in T cell apoptosis has been proposed as a central feature resulting in chronic airway inflammation (23, 24, 25).

We found that PGD2 significantly suppressed the apoptosis of Th2 cells induced by IL-2 deprivation. This effect was mediated by CRTH2-dependent activation of the PI3K pathway.

PGD2 was purchased from Biomol. 13,14-Dihydro-15-keto-PGD2 (DK-PGD2), BW245C, BW868C, SQ29548, and ramatroban were from Cayman Chemical. TM30089 was supplied by ChemieTek. LY294002, recombinant human Fas ligand (FasL), t-butoxycarbonyl-Asp-fluoromethyl ketone (BAF), N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (Z-VAD), N-benzyloxycarbonyl-Ile-Glu-Thr-Asp-fluoromethyl ketone (Z-IETD), and a cytochrome c (cyt c) release apoptosis assay kit were obtained from Calbiochem. The human CD4+ T cell isolation kit II, an anti-human CRTH2 microbead kit, a human CD4+CD25+ regulatory T cell isolation kit, a T cell activation/expansion kit, anti-CD4-PE, anti-CD25-allophycocyanin, anti-CRTH2-PE, anti-FoxP3-PE, and a FoxP3 staining buffer set were from Miltenyi Biotec. X-VIVO 15 medium was purchased from Lonza. Recombinant human IL-2 and recombinant human IL-4 were from eBioscience. PE-annexin V, propidium iodide (PI), and annexin-binding buffer were obtained from Invitrogen. All Abs were supplied by New England Biolabs except for the mAb to CD95 Fas (clone 7C11), which was from Immunotech, the Abs to p-BAD (Ser136) and β-tubulin, which were from Santa Cruz Biotechnology, and Alexa Fluor 647 mouse anti-cyt c and rabbit anti-caspase-8, which were from BD Pharmingen. Other chemicals were from Sigma-Aldrich.

Human CRTH2+CD4+ Th2 cells were prepared using a modified method described previously (3). Briefly, PBMC were isolated from buffy coats from healthy blood donors whose atopic status was unknown (National Blood Service, Bristol, U.K.) by Ficoll Hypaque (Amersham Biosciences) density gradient centrifugation, followed by CD4+ cell purification using MACS CD4+ T cell isolation kit II (Miltenyi Biotec). After 7 days of culture in X-VIVO 15 medium containing 10% human serum, 50 U/ml IL-2, and 100 ng/ml IL-4, CRTH2-positive cells were isolated from the CD4+ cultures by positive selection using an anti-human CRTH2 microbead kit. The harvested CD4+ CRTH2+ cells were treated as Th2 cells and were further amplified by stimulation with a T cell activation/expansion kit (Miltenyi Biotec) and grown in X-VIVO 15 medium containing 10% human serum and 50 U/ml IL-2 before use.

Human CD4+CD25+ regulatory T cells (Treg) were isolated with buffy coats by Ficoll Hypaque density gradient centrifugation, followed by Treg cell separation using a human CD4+CD25+ regulatory T cell isolation kit (Miltenyi Biotec). The cells were cultured in X-VIVO 15 medium containing 10% human serum and 50 U/ml IL-2 before use.

The purity of the Th2 and Treg cells was confirmed by fluorescent staining with anti-CD4-PE, anti-CD25-allophycocyanin, anti-CRTH2-PE, or anti-FoxP3-PE according to the supplier’s instructions (Miltenyi Biotec) followed by analysis using a FACSArray flow cytometer (BD Bioscience).

Th2 or Treg cells were harvested after different treatments and transferred to annexin-binding buffer followed by incubation with PE-annexin V/PI at room temperature for 15 min according to the manufacturer’s instructions (Invitrogen). The stained cells were analyzed by a FACSArray flow cytometer. The cells with annexin V positive and PI negative were counted as apoptotic cells.

Th2 cells, after treatment, were washed with PBS and loaded into a poly-l-lysine-coated, 8-well chamber slide (Lab-Tek). After a short spin to attach the cells, the cells were fixed with 5% formaldehyde in PBS, permeabilized with precold (−20°C) 90% methanol, blocked with 3% BSA in PBS and then probed with Alexa Fluor 647 mouse anti-cyt c followed by staining with 2.5 μg/ml Hoechst 33342 in PBS. The slides were observed on a Leica DMRXA fluorescence microscope and images were recorded with Openlab 3.0.9 software (Improvision).

The assay was performed using a cyt c release apoptosis assay kit according to the manufacturer’s protocol (Calbiochem). Briefly, Th2 cells, after treatment as indicated in the results, were washed and resuspended in a cytosol extraction buffer. After 10 min of incubation in ice, the cells were homogenized using a tissue grinder. Supernatants were collected after 10 min of centrifugation at 700 × g for a further centrifugation at 10,000 × g for 30 min. The supernatants after the second centrifugation were treated as cytosolic fractions, and the pellets resuspended in a mitochondrial extraction buffer were treated as mitochondrial fractions. The fractions were analyzed with Western blotting for cyt c.

Human Th2 cells were treated under different conditions as indicated in Results for 20 min (for Akt), 30 min (for BAD), 7 h (for caspase 8), or 16 h (for caspase 3 or poly (ADP-ribose) polymerase (PARP)). The cells were washed once with PBS and then solubilized in lysis buffer (20 mM Tris-HCl (pH 7.4), 250 mM sucrose, 1 mM EDTA, 1 mM EGTA, 1 mM sodium orthovanadate, 10 mM sodium glycerophosphate, 50 mM sodium fluoride, 5 mM sodium pyrophosphate, 0.1% 2-ME, protease inhibitor mixture, and 1% Triton X-100). The samples from the above treatments or from cyt c release apoptosis assay were fractionated by SDS-PAGE and then electrophoretically transferred to a nitrocellulose membrane, and probed with Abs as indicated in the results. Immunocomplexes were detected using ECL, and immunopositive bands were recorded using x-ray film.

Data were analyzed using one-way ANOVA followed by the Newman-Keuls test. Probability values of p < 0.05 were considered as statistically significant.

Th2 cells grown in normal medium containing 50U/ml IL-2 consistently demonstrated normal nuclear morphology, intact mitochondria filled with cyt c, and a low degree of annexin V binding (<7%) (Fig. 1, AC). Removal of IL-2 led to an increase in the number of cells exhibiting chromatin condensation, nuclear shrinkage, DNA degradation, and cyt c release from mitochondria into cytoplasm and a decrease in the number of cells with normal morphology (Fig. 1, A and B). The proportion of annexin V-positive cells was increased to ∼31% at 24 h (Fig. 1,C). The increase in cellular morphological changes and annexin V binding was suppressed markedly by PGD2 (Fig. 1, AC). The addition of 100 nM PGD2 reduced the number of annexin V-positive cells from ∼26.2 to ∼11% at 16 h and from ∼28.7 to ∼12.8% at 20 h. The reduction was decreased thereafter, suggesting that the inhibition of apoptosis diminished after 20 h. However, due to the degradation of early apoptotic cells and the instability of PGD2 over prolonged periods of time, the precise potency of PGD2 on Th2 apoptosis after 24 h could not be determined (data not shown). The inhibitory effect of PGD2 was concentration dependent (IC50 = 11.4 ± 3.1 nM at 16 h) (Fig. 1 D).

FIGURE 1.

PGD2 suppresses apoptosis of human Th2 cells induced by IL-2 deprivation. A and B, Th2 cells were incubated in medium with or without 50 U/ml IL-2 or a medium deprived of IL-2 but containing 100 nM PGD2 for 16 h. A, The cells were fixed for immunocytochemical analysis of cyt c (green) and nuclei (blue). Arrows indicate chromatin condensation and nuclear shrinkage, and arrowheads indicate DNA degradation. B, The cells were homogenized and isolated into mitochondrial fractions (top panel) and cytosolic fractions (bottom panel) for immunoblotting of cyt c. C and D, For annexin V binding assays the cells were incubated in medium with (▵) or without 50 U/ml IL-2 (□) or in a medium without IL-2 but containing 100 nM PGD2 (•) for various times (C) or were treated with various concentrations of PGD2 in the absence of IL-2 (○) for 16 h (D). The cells were harvested and stained with PE-annexin V/PI, followed by flow cytometric analysis. The background for annexin V binding in control cultures containing 50 U/ml IL-2 in D was 6.3 ± 0.8% and is indicated by the dashed line.

FIGURE 1.

PGD2 suppresses apoptosis of human Th2 cells induced by IL-2 deprivation. A and B, Th2 cells were incubated in medium with or without 50 U/ml IL-2 or a medium deprived of IL-2 but containing 100 nM PGD2 for 16 h. A, The cells were fixed for immunocytochemical analysis of cyt c (green) and nuclei (blue). Arrows indicate chromatin condensation and nuclear shrinkage, and arrowheads indicate DNA degradation. B, The cells were homogenized and isolated into mitochondrial fractions (top panel) and cytosolic fractions (bottom panel) for immunoblotting of cyt c. C and D, For annexin V binding assays the cells were incubated in medium with (▵) or without 50 U/ml IL-2 (□) or in a medium without IL-2 but containing 100 nM PGD2 (•) for various times (C) or were treated with various concentrations of PGD2 in the absence of IL-2 (○) for 16 h (D). The cells were harvested and stained with PE-annexin V/PI, followed by flow cytometric analysis. The background for annexin V binding in control cultures containing 50 U/ml IL-2 in D was 6.3 ± 0.8% and is indicated by the dashed line.

Close modal

To confirm that the annexin V binding was indicative of the apoptotic process in human Th2 cells, we examined the caspase activities in the same samples (Fig. 2). IL-2 withdrawal promoted the cleavage of caspase 3 and PARP in the Th2 cell cultures. Cleaved caspase 3 (∼17 kDa) and cleaved PARP (∼89 kDa) fragments increased significantly after IL-2 deprivation (Fig. 2,A). An extra band smeared below the major bands and in proportion with the major bands in the blot to PARP is likely caused by incomplete denaturation of the protein during sample preparation. The cleavages of caspase 3 and PARP were almost completely abolished by 100 μM BAF, a pan-caspase inhibitor (26). PGD2 (100 nM) also inhibited cleavage of caspase 3 and PARP. Addition of the PI3K inhibitor LY294002 (100 μM) inhibited the antiapoptotic effect of PGD2 and also significantly enhanced the cleavage of caspase 3 and PARP (Fig. 2,A). Furthermore, the pan-caspase inhibitors, BAF and Z-VAD also suppressed the increase of annexin V binding induced by IL-2 deprivation (Fig. 2 B).

FIGURE 2.

Apoptosis in Th2 cells induced by IL-2 deprivation is caspase dependent. Th2 cells were treated for 16 h in the presence or absence of 50 U/ml IL-2, 100 nM PGD2, 100 μM BAF, 100 μM Z-VAD, or 100 μM LY294002 and then were lysed for immunoblotting of caspase 3 (top panel) or PARP (bottom panel) (A) or stained with PE-annexin V/PI for quantitative flow cytometric analysis (B). The molecular size of the proteins in the immunoblot is indicated on the left side of each blot panel. For B, p < 0.001 by ANOVA; p > 0.05 by Newman-Keuls test for 50U/ml IL-2 vs 100 nM PGD2/100 μM BAF/100 μM Z-VAD, 100 nM PGD2 vs 100 μM BAF/100 μM Z-VAD, and 100 μM BAF vs 100 μM Z-VAD; n = 3.

FIGURE 2.

Apoptosis in Th2 cells induced by IL-2 deprivation is caspase dependent. Th2 cells were treated for 16 h in the presence or absence of 50 U/ml IL-2, 100 nM PGD2, 100 μM BAF, 100 μM Z-VAD, or 100 μM LY294002 and then were lysed for immunoblotting of caspase 3 (top panel) or PARP (bottom panel) (A) or stained with PE-annexin V/PI for quantitative flow cytometric analysis (B). The molecular size of the proteins in the immunoblot is indicated on the left side of each blot panel. For B, p < 0.001 by ANOVA; p > 0.05 by Newman-Keuls test for 50U/ml IL-2 vs 100 nM PGD2/100 μM BAF/100 μM Z-VAD, 100 nM PGD2 vs 100 μM BAF/100 μM Z-VAD, and 100 μM BAF vs 100 μM Z-VAD; n = 3.

Close modal

To elucidate the receptor mediating the above effects of PGD2, we compared effects of BW245C, a specific DP1 agonist, and DK-PGD2, a specific CRTH2 agonist, with that of PGD2 on the apoptosis of Th2 cells (Fig. 3,A). DK-PGD2 exhibited a similar potency (IC50 = 10.2 ± 0.7 nM) to that of PGD2 in mediating this effect. In contrast, BW245C was without effect. The inhibitory effect of PGD2 on Th2 apoptosis was completely inhibited by the CRTH2 antagonists ramatroban and TM30089, but not by the DP1 antagonist BW868C or by the thromboxane-like prostanoid receptor (TP) antagonist SQ29548 (Fig. 3 B).

FIGURE 3.

The inhibitory effect of PGD2 on apoptosis of IL-2-deprived Th2 cells is mediated by CRTH2. Th2 cells were incubated in medium without IL-2 but containing various concentrations of PGD2 (•), BW245C (⋄), or DK-PGD2 (□) for 16 h (A) or treated in the presence or absence of 50 U/ml IL-2, 100 nM PGD2, 1 μM ramatroban, 1 μM TM30089, 1 μM BW868C or 1 μM SQ29548 for 16 h (B); or Treg cells were incubated with or without 50 U/ml IL-2 or 100 nM PGD2 for 16 h (C). The cells were stained with PE-annexin V/PI, followed by quantitative flow cytometric analysis. For A, the background annexin V binding in control cultures with 50 U/ml IL-2 was 7.32 ± 0.55% and is indicated with a dashed line. For B, p < 0.0001 by ANOVA; p > 0.05 by Newman-Keuls test for 50U/ml IL-2 vs 100 nM PGD2/100 nM PGD2 plus 1 μM BW868C/100 nM PGD2 plus 1 μM SQ29548; 100 nM PGD2 vs 100 nM PGD2 plus 1 μM BW868C/100 nM PGD2 plus 1 μM SQ29548; 100 nM PGD2 plus 1 μM BW868C vs 100 nM PGD2 plus 1 μM SQ29548; no additive vs 100 nM PGD2 plus 1 μM ramatroban/100 nM PGD2 plus 1 μM TM30089; 100 nM PGD2 plus 1 μM ramatroban vs 100 nM PGD2 plus 1 μM TM30089; n = 3. For C, p < 0.005 by ANOVA; p > 0.05 by Newman-Keuls test for no additive vs 100 nM PGD2; n = 3.

FIGURE 3.

The inhibitory effect of PGD2 on apoptosis of IL-2-deprived Th2 cells is mediated by CRTH2. Th2 cells were incubated in medium without IL-2 but containing various concentrations of PGD2 (•), BW245C (⋄), or DK-PGD2 (□) for 16 h (A) or treated in the presence or absence of 50 U/ml IL-2, 100 nM PGD2, 1 μM ramatroban, 1 μM TM30089, 1 μM BW868C or 1 μM SQ29548 for 16 h (B); or Treg cells were incubated with or without 50 U/ml IL-2 or 100 nM PGD2 for 16 h (C). The cells were stained with PE-annexin V/PI, followed by quantitative flow cytometric analysis. For A, the background annexin V binding in control cultures with 50 U/ml IL-2 was 7.32 ± 0.55% and is indicated with a dashed line. For B, p < 0.0001 by ANOVA; p > 0.05 by Newman-Keuls test for 50U/ml IL-2 vs 100 nM PGD2/100 nM PGD2 plus 1 μM BW868C/100 nM PGD2 plus 1 μM SQ29548; 100 nM PGD2 vs 100 nM PGD2 plus 1 μM BW868C/100 nM PGD2 plus 1 μM SQ29548; 100 nM PGD2 plus 1 μM BW868C vs 100 nM PGD2 plus 1 μM SQ29548; no additive vs 100 nM PGD2 plus 1 μM ramatroban/100 nM PGD2 plus 1 μM TM30089; 100 nM PGD2 plus 1 μM ramatroban vs 100 nM PGD2 plus 1 μM TM30089; n = 3. For C, p < 0.005 by ANOVA; p > 0.05 by Newman-Keuls test for no additive vs 100 nM PGD2; n = 3.

Close modal

To further confirm the receptor mediating the effect of PGD2 on Th2 cell apoptosis, we also investigated the effect of PGD2 on the apoptosis of human CD4+CD25+ Treg cells. Although, like Th2 cells, Treg cells express both CD4 and CD25 and their growth is IL-2-dependent in culture, Treg cells are CRTH2 negative (Table I) and PGD2 did not prevent their apoptosis induced by IL-2 deprivation (Fig. 3 C).

Table I.

Comparison of human CD4+/CRTH2+ Th2 cells and CD4+/CD25+ Treg cellsa

CD4 (%)bCD25 (%)bCRTH2 (%)bFoxP3 (%)b
Th2 97.7 ± 8.9 98.2 ± 0.5 85 ± 5 17.9 ± 2 
Treg 95.1 ± 0.45 83.9 ± 1 4.4 ± 0.6 52.8 ± 4 
CD4 (%)bCD25 (%)bCRTH2 (%)bFoxP3 (%)b
Th2 97.7 ± 8.9 98.2 ± 0.5 85 ± 5 17.9 ± 2 
Treg 95.1 ± 0.45 83.9 ± 1 4.4 ± 0.6 52.8 ± 4 
a

The cells were stained with fluorescence-conjugated Abs followed by analysis using a flow cytometer as described in Materials and Methods. Data are from pooled samples and are representative of two experiments.

b

Percentage of positive cells within the purified cell population.

Reduced phosphorylation of Akt, a substrate for PI3K, and phosphorylation of BAD at Ser136, a substrate for Akt, was detected in Th2 cells shortly after IL-2 withdrawal (1–2 h) and was restored by the addition of IL-2 or PGD2 (Fig. 4,A). PGD2 stimulated a dose-dependent increase in phospho-Akt (Fig. 4,B), and the intensity of Akt phosphorylation was highly correlated with the antiapoptotic effect of PGD2 (Fig. 1,C). In contrast, BAF did not enhance the levels of phospho-Akt and phospho-BAD (Fig. 4,A). The effect of PGD2 on both Akt and BAD phosphorylation was completely blocked by 100 μM LY294002 (Fig. 4,A). Furthermore, inhibition of PI3K with LY294002 prevented the inhibitory effect of PGD2 on annexin V binding of Th2 cells and increased annexin V binding even in the presence of IL-2 (Fig. 4 C).

FIGURE 4.

PI3K signaling is involved in the antiapoptotic effect of PGD2. Th2 cells were (for A and C) pretreated with 100 μM LY294002 or control buffer for 30 min and then treated with medium with or without 50 U/ml IL-2 in the presence or absence of 100 nM PGD2, 100 μM BAF, or 100 μM LY294002 for 20 min (A, top two panels), 30 min (A, middle two panels), or 16 h (A, bottom panel, and C) or (for B) stimulated with various concentration of PGD2 for 20 min. The cells were lysed for immunoblotting of phosphorylated Akt (p-Akt), Akt (A and B), phosphorylated BAD (p-BAD; Ser136), BAD, or β-tubulin (A) or stained with PE-annexin V/PI for quantitative flow cytometric analysis (C). The molecular size of the protein in the immunoblot is indicated on the left side of each panel. For C, p < 0.0001 by ANOVA; p > 0.05 by Newman-Keuls test for no additive vs 50U/ml IL-2 plus 100 μM LY294002; n = 3.

FIGURE 4.

PI3K signaling is involved in the antiapoptotic effect of PGD2. Th2 cells were (for A and C) pretreated with 100 μM LY294002 or control buffer for 30 min and then treated with medium with or without 50 U/ml IL-2 in the presence or absence of 100 nM PGD2, 100 μM BAF, or 100 μM LY294002 for 20 min (A, top two panels), 30 min (A, middle two panels), or 16 h (A, bottom panel, and C) or (for B) stimulated with various concentration of PGD2 for 20 min. The cells were lysed for immunoblotting of phosphorylated Akt (p-Akt), Akt (A and B), phosphorylated BAD (p-BAD; Ser136), BAD, or β-tubulin (A) or stained with PE-annexin V/PI for quantitative flow cytometric analysis (C). The molecular size of the protein in the immunoblot is indicated on the left side of each panel. For C, p < 0.0001 by ANOVA; p > 0.05 by Newman-Keuls test for no additive vs 50U/ml IL-2 plus 100 μM LY294002; n = 3.

Close modal

Treatment of Th2 cells with increasing concentrations of FasL (Fig. 5,A) or 7C11, an Ab against human Fas (Fig. 5,B), for 16 h increased annexin V-positive cells in a concentration-dependent manner, and this response was not affected by addition of PGD2 (100 nM). To confirm the apoptotic mechanism induced by the Fas reaction in Th2 cells, caspase activities in the cells were analyzed using Western blotting. Treatment with 7C11 induced significant cleavage of caspases 3 and 8 in Th2 cells (Fig. 5 C). Z-IETD, a caspase 8 inhibitor, blocked the cleavage of caspase 3 but not caspase 8 cleavage. However, PGD2 had no effect on the cleavage of either caspase induced by Fas activation.

FIGURE 5.

PGD2 has no effect on apoptosis of Th2 cells induced by Fas ligation. A and B, Th2 cells were treated with various concentrations of FasL (A) or anti-Fas Ab 7C11 (B) in the absence (□) or presence (•) of 100 nM PGD2 for 16 h. The cells were stained with PE-annexin V/PI, followed by quantitative flow cytometric analysis. C, Th2 cells were incubated with 1 μg/ml 7C11 Ab in the absence or presence of 100 nM PGD2 or Z-IETD for 7 h and then lysed for immunoblotting of caspase 8 (top panel) or caspase 3 (bottom panel).

FIGURE 5.

PGD2 has no effect on apoptosis of Th2 cells induced by Fas ligation. A and B, Th2 cells were treated with various concentrations of FasL (A) or anti-Fas Ab 7C11 (B) in the absence (□) or presence (•) of 100 nM PGD2 for 16 h. The cells were stained with PE-annexin V/PI, followed by quantitative flow cytometric analysis. C, Th2 cells were incubated with 1 μg/ml 7C11 Ab in the absence or presence of 100 nM PGD2 or Z-IETD for 7 h and then lysed for immunoblotting of caspase 8 (top panel) or caspase 3 (bottom panel).

Close modal

PGD2 is able to promote recruitment and activation of Th2 cells through a high affinity interaction with its receptor, CRTH2 (9, 11, 12). However, in addition to enhanced recruitment, the inflammatory burden of Th2 cells within inflamed tissues is likely to be also influenced by the clearance of these cells (27). Apoptosis is critically required for the clearance of Th2 cells, and a defect in apoptosis has been proposed as a feature of asthma pathology (23, 24, 25, 28). Indeed, induction of T cell apoptosis has been proposed as an important component of the action of both inhaled corticosteroids and immunotherapy in asthma (29, 30, 31). We have therefore investigated whether the apoptosis of Th2 cells can be influenced by PGD2 and the signaling mechanisms involved. It was found that PGD2 suppressed the cellular morphological changes, annexin V binding, cyt c release, and caspase activities in Th2 cells undergoing apoptosis as a result of IL-2 deprivation. This effect is likely to be due to a direct effect of PGD2 rather than secondary effect on the production of IL-2, because IL-2 levels were not increased by PGD2 treatment (data not shown). The inhibitory effect of PGD2 was concentration dependent and mimicked by the selective CRTH2 agonist DK-PGD2, but not by the selective DP1 agonist BW245C. Ramatroban, a dual TP/CRTH2 antagonist, and TM30089, a selective CRTH2 antagonist, blocked the antiapoptotic effect of PGD2 whereas the selective TP antagonist SQ29548 was without effect. Further evidence for the involvement of CRTH2 in this response is provided by the absence of effect of PGD2 on the apoptosis of CD4+CD25+ Treg cells that do not express CRTH2. PI3K activity is clearly involved in the inhibition of apoptosis induced by PGD2, because IL-2 deprivation resulted in the dephosphorylation of Akt and its downstream substrate BAD and induced cyt c release from mitochondria into cytoplasm, whereas PGD2 restored phosphorylation of these proteins and prevented the release of cyt c. The PI3K inhibitor LY294002 prevented the effect of PGD2 on both the signaling events and the apoptosis of Th2 cells. Therefore, PGD2/CRTH2 interaction prevents the apoptosis of Th2 cells induced by cytokine deprivation through stimulation of the PI3K signal pathway.

T cell homeostasis can be controlled by several processes including two key apoptotic mechanisms (27, 32): 1) activation-induced cell death triggered by the restimulation of TCR involving death receptors, e.g., Fas (Apo-1/CD95) (33, 34, 35); and 2) activated T cell autonomous death induced by the disappearance of survival signals involving Bcl-2 family members in the mitochondria (36, 37). Both mechanisms are thought to be important in avoiding the overexpansion of activated cell clones after Ag stimulation and to avoid attack by autoreactive lymphocytes (21, 22, 38, 39). It has also been reported recently that Treg cells can suppress immune responses by inducing IL-2 deprivation-mediated apoptosis of effector CD4+ T cells (40). Pathophysiological concentrations of PGD2 prevented the apoptosis of Th2 cells induced by IL-2 deprivation, a form of activated T cell autonomous death, suggesting that this may contribute to the survival of Th2 cells at sites of allergic inflammation and impede the resolution of disease because PGD2 is found in high concentrations in a number of allergic diseases (41, 42, 43). We did not test the effect of PGD2 on activation-induced cell death responses, but interestingly PGD2 has no effect on the apoptosis of Th2 cells induced by Fas ligation. Considering that Bcl-2 regulation and Fas regulation are two different apoptotic mechanisms but share the same downstream effector pathway involving caspase 3 activation (27, 36), it is likely that PGD2 does not interfere directly with the cellular apoptotic machinery but rather acts as a survival factor through the activation of PI3K.

In T cells, PI3K can be activated by a number of different receptors, including the TCR, costimulatory receptors, cytokine receptors, and chemokine receptors (44). We have reported that PI3Kγ, a class IB PI3K isoform, is involved in CRTH2-mediated proinflammatory responses of Th2 cells to PGD2 (17). Considerable evidence suggests that activation of the PI3K pathway is required for T cell survival and proliferation (19, 45). Overactivity of PI3K activity leads to lymphoproliferative disorders and autoimmune disease (46), and inhibition of PI3K increases cell apoptosis (20, 47). It also has been reported that activation of class IA PI3K (p85) by the ligation of CD3 was able to protect activated Th2 clones from Fas-mediated apoptosis through blockade of Fas aggregation (48, 49). However, no such an effect was observed in our system when the PI3K pathway was activated by PGD2/CRTH2, suggesting that the signaling via PI3Kγ induced by G protein-coupled receptors is probably not sufficient to interfere with Fas-dependent apoptosis (50).

One of the mechanisms used by PI3K to protect cells from apoptosis is regulation of the activity of proteins of the Bcl-2 family. It promotes Bcl-xL expression in response to CD28 costimulation (51) and catalyzes BAD phosphorylation at Ser136 through its downstream kinase Akt (52, 53, 54). Bcl-xL is a prosurvival member of Bcl-2 family that can heterodimerize with proapoptotic members of the family to neutralize their activity (55), while phospho-BAD can be sequestered in the cytosol by 14-3-3 proteins, thereby preventing BAD-mediated inhibition of Bcl-xL/Bcl-2 (56). Both of these effects will result in the protection of mitochondria from membrane permeabilization and the release of apoptotic factors, including cyt c. The effect of PGD2/CRTH2 on phospho-BAD and cyt c in Th2 cells suggests that PI3K-mediated protection of mitochondria plays a central role in the antiapoptotic effects observed.

In conclusion, this study highlights an antiapoptotic role for PGD2 mediated by CRTH2 in human Th2 cells. PGD2 inhibits Th2 cell apoptosis in response to the disappearance of growth factor but not in response to Fas ligation. The summary scheme in Fig. 6 shows the proposed survival signals induced by PGD2/CRTH2 interaction that leads Th2 cells to become resistant to apoptosis. Antagonism of CRTH2 has been proposed as an attractive approach to treat allergic diseases (57). Other mediators in addition to PGD2 may contribute to Th2 cell survival through the activation of PI3K and possibly other pathways. IL-2 itself has been reported to be present in asthmatic airways (58) and may prevent the apoptosis of Th2 cells. However, the high levels of PGD2 produced in a chronic allergic setting combined with its known dominant role in driving other aspects of Th2 cell activation (59, 60) suggest that it may contribute to survival signaling in Th2 cells. Further studies are required to define the precise pathophysiological role of PGD2 in promoting Th2 cell survival. The identification of an inhibitory effect of CRTH2 activation on Th2 apoptosis suggests that the activation of CRTH2 may impede the resolution of allergic inflammation in addition to promoting the recruitment and activation of Th2 cells. These findings lend further support to the view that antagonism of CRTH2 may find utility in the treatment of allergic disease.

FIGURE 6.

Scheme summarizing the proposed signaling pathways used by PGD2/CRTH2 to suppress the apoptosis of human Th2 cells induced by IL-2 deprivation. Acting in a manner to similar to that of cytokine growth factors, activation of CRTH2 by PGD2 stimulates the PI3K signaling pathway leading to phosphorylation of Akt, which catalyzes BAD phosphorylation. Phospho-BAD is sequestered in the cytosol by 14-3-3, precluding its inhibition of Bcl-xL. Consequently, the downstream apoptotic signaling events, including mitochondrial membrane permeabilization, cyt c efflux from mitochondria, caspase9/3 cleavage, and DNA fragmentation, are blocked. Inhibition of CRTH2 by antagonists or inhibition of PI3K by LY294002 can abolish the antiapoptotic effects of PGD2/CRTH2, whereas blockade of caspase activities with BAF only inhibits the late stages of the apoptotic process and exerts no effect on upstream Akt, BAD, and cyt c. Activation of CRTH2 does not interfere with the Fas-dependent apoptotic pathway.

FIGURE 6.

Scheme summarizing the proposed signaling pathways used by PGD2/CRTH2 to suppress the apoptosis of human Th2 cells induced by IL-2 deprivation. Acting in a manner to similar to that of cytokine growth factors, activation of CRTH2 by PGD2 stimulates the PI3K signaling pathway leading to phosphorylation of Akt, which catalyzes BAD phosphorylation. Phospho-BAD is sequestered in the cytosol by 14-3-3, precluding its inhibition of Bcl-xL. Consequently, the downstream apoptotic signaling events, including mitochondrial membrane permeabilization, cyt c efflux from mitochondria, caspase9/3 cleavage, and DNA fragmentation, are blocked. Inhibition of CRTH2 by antagonists or inhibition of PI3K by LY294002 can abolish the antiapoptotic effects of PGD2/CRTH2, whereas blockade of caspase activities with BAF only inhibits the late stages of the apoptotic process and exerts no effect on upstream Akt, BAD, and cyt c. Activation of CRTH2 does not interfere with the Fas-dependent apoptotic pathway.

Close modal

The authors have no financial conflict of interest.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

2

Abbreviations used in this paper: DP, D prostanoid receptor; BAF, t-butoxycarbonyl-Asp-fluoromethyl ketone; CRTH2, chemoattractant receptor-homologous molecule expressed on Th2 cells; cyt c, cytochrome c; DK-PGD2, 13,14-dihydro-15-keto-PGD2; FasL, Fas ligand; PARP, poly(ADP-ribose) polymerase; PI, propidium iodide; TP, thromboxane-like prostanoid receptor; Treg, regulatory T cell; Z-IETD, N-benzyloxycarbonyl-Ile-Glu-Thr-Asp-fluoromethyl ketone; Z-VAD, N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone.

1
Lewis, R. A., N. A. Soter, P. T. Diamond, K. F. Austen, J. A. Oates, L. J. Roberts, II.
1982
. Prostaglandin D2 generation after activation of rat and human mast cells with anti-IgE.
J. Immunol.
129
:
1627
-1631.
2
Tanaka, K., K. Ogawa, K. Sugamura, M. Nakamura, S. Takano, K. Nagata.
2000
. Cutting edge: differential production of prostaglandin D2 by human helper T cell subsets.
J. Immunol.
164
:
2277
-2280.
3
Vinall, S. L., E. R. Townsend, R. Pettipher.
2007
. A paracrine role for chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2) in mediating chemotactic activation of CRTH2+CD4+ T helper type 2 lymphocytes.
Immunology
121
:
577
-584.
4
Heavey, D. J., P. Lumley, S. E. Barrow, M. B. Murphy, P. P. Humphrey, C. T. Dollery.
1984
. Effects of intravenous infusions of prostaglandin D2 in man.
Prostaglandins
28
:
755
-767.
5
Van Hecken, A., M. Depré, I. De Lepeleire, C. Thach, M. Oeyen, J. Van Effen, T. Laethem, K. Mazina, T. Crumley, L. Wenning, et al
2007
. The effect of MK-0524, a prostaglandin D2 receptor antagonist, on prostaglandin D2-induced nasal airway obstruction in healthy volunteers.
Eur. J. Clin. Pharmacol.
63
:
135
-141.
6
Gervais, F. G., R. P. Cruz, A. Chateauneuf, S. Gale, N. Sawyer, F. Nantel, K. M. Metters, G. P. O'neill.
2001
. Selective modulation of chemokinesis, degranulation, and apoptosis in eosinophils through the PGD2 receptors CRTH2 and DP.
J. Allergy Clin. Immunol.
108
:
982
-988.
7
Yoshimura-Uchiyama, C., M. Iikura, M. Yamaguchi, H. Nagase, A. Ishii, K. Matsushima, K. Yamamoto, M. Shichijo, K. B. Bacon, K. Hirai.
2004
. Differential modulation of human basophil functions through prostaglandin D2 receptors DP and chemoattractant receptor-homologous molecule expressed on Th2 cells/DP2.
Clin. Exp. Allergy
34
:
1283
-1290.
8
Schratl, P., J. F. Royer, E. Kostenis, T. Ulven, E. M. Sturm, M. Waldhoer, G. Hoefler, R. Schuligoi, I. T. Lippe, B. A. Peskar, A. Heinemann.
2007
. The role of the prostaglandin D2 receptor, DP, in eosinophil trafficking.
J. Immunol.
179
:
4792
-4799.
9
Hirai, H., K. Tanaka, O. Yoshie, K. Ogawa, K. Kenmotsu, Y. Takamori, M. Ichimasa, K. Sugamura, M. Nakamura, S. Takano, K. Nagata.
2001
. Prostaglandin D2 selectively induces chemotaxis in T helper type 2 cells, eosinophils, and basophils via seven-transmembrane receptor CRTH2.
J. Exp. Med.
193
:
255
-261.
10
Nagata, K., H. Hirai.
2003
. The second PGD2 receptor CRTH2: structure, properties, and functions in leukocytes.
Prostaglandins Leukot. Essent. Fatty Acids
69
:
169
-177.
11
Tanaka, K., H. Hirai, S. Takano, M. Nakamura, K. Nagata.
2004
. Effects of prostaglandin D2 on helper T cell functions.
Biochem. Biophys. Res. Commun.
316
:
1009
-1014.
12
Xue, L., S. L. Gyles, F. R. Wettey, L. Gazi, E. Townsend, M. G. Hunter, R. Pettipher.
2005
. Prostaglandin D2 causes preferential induction of proinflammatory Th2 cytokine production through an action on chemoattractant receptor-like molecule expressed on Th2 cells.
J. Immunol.
175
:
6531
-6536.
13
Chevalier, E., J. Stock, T. Fisher, M. Dupont, M. Fric, H. Fargeau, M. Leport, S. Soler, S. Fabien, M. P. Pruniaux, et al
2005
. Cutting edge: chemoattractant receptor-homologous molecule expressed on Th2 cells plays a restricting role on IL-5 production and eosinophil recruitment.
J. Immunol.
175
:
2056
-2060.
14
Satoh, T., R. Moroi, K. Aritake, Y. Urade, Y. Kanai, K. Sumi, H. Yokozeki, H. Hirai, K. Nagata, T. Hara, et al
2006
. Prostaglandin D2 plays an essential role in chronic allergic inflammation of the skin via CRTH2 receptor.
J. Immunol.
177
:
2621
-2629.
15
Uller, L., J. M. Mathiesen, L. Alenmyr, M. Korsgren, T. Ulven, T. Hogberg, G. Andersson, C. G. Persson, E. Kostenis.
2007
. Antagonism of the prostaglandin D2 receptor CRTH2 attenuates asthma pathology in mouse eosinophilic airway inflammation.
Respir. Res.
8
:
16
16
Nomiya, R., M. Okano, T. Fujiwara, M. Maeda, Y. Kimura, K. Kino, M. Yokoyama, H. Hirai, K. Nagata, T. Hara, et al
2008
. CRTH2 plays an essential role in the pathophysiology of Cry j 1-induced pollinosis in mice.
J. Immunol.
180
:
5680
-5688.
17
Xue, L., S. L. Gyles, A. Barrow, R. Pettipher.
2007
. Inhibition of PI3K and calcineurin suppresses chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2)-dependent responses of Th2 lymphocytes to prostaglandin D2.
Biochem. Pharmacol.
73
:
843
-853.
18
Medina-Tato, D. A., S. G. Ward, M. L. Watson.
2007
. Phosphoinositide 3-kinase signalling in lung disease: leucocytes and beyond.
Immunology
121
:
448
-461.
19
Okkenhaug, K., A. Bilancio, J. L. Emery, B. Vanhaesebroeck.
2004
. Phosphoinositide 3-kinase in T cell activation and survival.
Biochem. Soc. Trans.
32
:
332
-335.
20
Schade, A. E., J. J. Powers, M. W. Wlodarski, J. P. Maciejewski.
2006
. Phosphatidylinositol-3-phosphate kinase pathway activation protects leukemic large granular lymphocytes from undergoing homeostatic apoptosis.
Blood
107
:
4834
-4840.
21
Cohen, J. J., R. C. Duke, V. A. Fadok, K. S. Sellins.
1992
. Apoptosis and programmed cell death in immunity.
Annu. Rev. Immunol.
10
:
267
-293.
22
Goodnow, C. C..
1996
. Balancing immunity and tolerance: deleting and tuning lymphocyte repertoires.
Proc. Natl. Acad. Sci. USA
93
:
2264
-2271.
23
Vignola, A. M., P. Chanez, G. Chiappara, L. Siena, A. Merendino, C. Reina, R. Gagliardo, M. Profita, J. Bousquet, G. Bonsignore.
1999
. Evaluation of apoptosis of eosinophils, macrophages, and T lymphocytes in mucosal biopsy specimens of patients with asthma and chronic bronchitis.
J. Allergy Clin. Immunol.
103
:
563
-573.
24
Lamb, J. P., A. James, N. Carroll, L. Siena, J. Elliot, A. M. Vignola.
2005
. Reduced apoptosis of memory T-cells in the inner airway wall of mild and severe asthma.
Eur. Respir. J.
26
:
265
-270.
25
Todo-Bom, A., A. Mota Pinto, V. Alves, S. Vale Pereira, M. Santos Rosa.
2007
. Apoptosis and asthma in the elderly.
J. Investig. Allergol. Clin. Immunol.
17
:
107
-112.
26
Xue, L., G. C. Fletcher, A. M. Tolkovsky.
2001
. Mitochondria are selectively eliminated from eukaryotic cells after blockade of caspases during apoptosis.
Curr. Biol.
11
:
361
-365.
27
Krammer, P. H., R. Arnold, I. N. Lavrik.
2007
. Life and death in peripheral T cells.
Nat. Rev. Immunol.
7
:
532
-542.
28
Tong, J., H. S. Bandulwala, B. S. Clay, R. A. Anders, R. A. Shilling, D. D. Balachandran, B. Chen, J. V. Weinstock, J. Solway, K. J. Hamann, A. I. Sperling.
2006
. Fas-positive T cells regulate the resolution of airway inflammation in a murine model of asthma.
J. Exp. Med.
203
:
1173
-1184.
29
Melis, M., L. Siena, E. Pace, M. Gjomarkaj, M. Profita, A. Pirazzoli, M. Todaro, G. Stassi, G. Bonsignore, A. M. Vignola.
2002
. Fluticasone induces apoptosis in peripheral T-lymphocytes: a comparison between asthmatic and normal subjects.
Eur. Respir. J.
19
:
257
-266.
30
Pace, E., R. Gagliardo, M. Melis, S. La Grutta, M. Ferraro, L. Siena, G. Bonsignore, M. Gjomarkaj, J. Bousquet, A. M. Vignola.
2004
. Synergistic effects of fluticasone propionate and salmeterol on in vitro T-cell activation and apoptosis in asthma.
J. Allergy Clin. Immunol.
114
:
1216
-1223.
31
Tsai, Y. G., J. W. Chien, W. L. Chen, J. J. Shieh, C. Y. Lin.
2005
. Induced apoptosis of Th2 lymphocytes in asthmatic children treated with Dermatophagoides pteronyssinus immunotherapy.
Pediatr. Allergy Immunol.
16
:
602
-608.
32
Arnold, R., D. Brenner, M. Becker, C. R. Frey, P. H. Krammer.
2006
. How T lymphocytes switch between life and death.
Eur. J. Immunol.
36
:
1654
-1658.
33
Ju, S. T., D. J. Panka, H. Cui, R. Ettinger, M. el-Khatib, D. H. Sherr, B. Z. Stanger, A. Marshak-Rothstein.
1995
. Fas(CD95)/FasL interactions required for programmed cell death after T-cell activation.
Nature
373
:
444
-448.
34
Krammer, P. H..
2000
. CD95’s deadly mission in the immune system.
Nature
407
:
789
-795.
35
Green, D. R., N. Droin, M. Pinkoski.
2003
. Activation-induced cell death in T cells.
Immunol. Rev.
193
:
70
-81.
36
Strasser, A., M. Pellegrini.
2004
. T-lymphocyte death during shutdown of an immune response.
Trends Immunol.
25
:
610
-615.
37
Dooms, H., A. K. Abbas. Control of CD4+ T-cell memory by cytokines and costimulators.
Immunol. Rev.
211
:
23
-38.
38
Webb, S., C. Morris, J. Sprent.
1990
. Extrathymic tolerance of mature T cells: clonal elimination as a consequence of immunity.
Cell
63
:
1249
-1256.
39
Kawabe, Y., A. Ochi.
1991
. Programmed cell death and extrathymic reduction of Vβ8+CD4+ T cells in mice tolerant to Staphylococcus aureus enterotoxin B.
Nature
349
:
245
-248.
40
Pandiyan, P., L. Zheng, S. Ishihara, J. Reed, M. J. Lenardo.
2007
. CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation-mediated apoptosis of effector CD4+ T cells.
Nat. Immunol.
812
:
1353
-1362.
41
Naclerio, R. M., H. L. Meier, A. Kagey-Sobotka, N. F. Adkinson, D. A. Meyers, P. S. Norman, L. M. Lichtenstein.
1983
. Mediator release after nasal airway challenge with allergen.
Am. Rev. Respir. Dis.
128
:
597
-602.
42
Murray, J. J., A. B. Tonnel, A. R. Brash, L. J. Roberts, P. Gosset, R. Workman, A. Capron, J. A. Oates.
1986
. Release of prostaglandin D2 into human airways during acute antigen challenge.
N. Engl. J. Med.
315
:
800
-804.
43
Charlesworth, E. N., A. Kagey-Sobotka, R. P. Schleimer, P. S. Norman, L. M. Lichtenstein.
1991
. Prednisone inhibits the appearance of inflammatory mediators and the influx of eosinophils and basophils associated with the cutaneous late-phase response to allergen.
J. Immunol.
146
:
671
-676.
44
Okkenhaug, K., B. Vanhaesebroeck.
2003
. PI3K in lymphocyte development, differentiation and activation.
Nat. Rev. Immunol.
3
:
317
-330.
45
Webb, L. M., E. Vigorito, M. P. Wymann, E. Hirsch, M. Turner.
2005
. Cutting edge: T cell development requires the combined activities of the p110γ and p110δ catalytic isoforms of phosphatidylinositol 3-kinase.
J. Immunol.
175
:
2783
-2787.
46
Borlado, L. R., C. Redondo, B. Alvarez, C. Jimenez, L. M. Criado, J. Flores, M. A. Marcos, C. Martinez-A, D. Balomenos, A. C. Carrera.
2000
. Increased phosphoinositide 3-kinase activity induces a lymphoproliferative disorder and contributes to tumor generation in vivo.
FASEB J.
14
:
895
-903.
47
Breslin, E. M., P. C. White, A. M. Shore, M. Clement, P. Brennan.
2005
. LY294002 and rapamycin co-operate to inhibit T-cell proliferation.
Br. J. Pharmacol.
144
:
791
-800.
48
Varadhachary, A. S., M. E. Peter, S. N. Perdow, P. H. Krammer, P. Salgame.
1999
. Selective up-regulation of phosphatidylinositol 3′-kinase activity in Th2 cells inhibits caspase-8 cleavage at the death-inducing complex: a mechanism for Th2 resistance from Fas-mediated apoptosis.
J. Immunol.
163
:
4772
-4779.
49
Varadhachary, A. S., M. Edidin, A. M. Hanlon, M. E. Peter, P. H. Krammer, P. Salgame.
2001
. Phosphatidylinositol 3′-kinase blocks CD95 aggregation and caspase-8 cleavage at the death-inducing signaling complex by modulating lateral diffusion of CD95.
J. Immunol.
166
:
6564
-6569.
50
Stephens, L., A. Smrcka, F. T. Cooke, T. R. Jackson, P. C. Sternweis, P. T. Hawkins.
1994
. A novel phosphoinositide 3 kinase activity in myeloid-derived cells is activated by G protein βγ subunits.
Cell
77
:
83
-93.
51
Wu, L. X., J. La Rose, L. Chen, C. Neale, T. Mak, K. Okkenhaug, R. Wange, R. Rottapel.
2005
. CD28 regulates the translation of Bcl-xL via the phosphatidylinositol 3-kinase/mammalian target of rapamycin pathway.
J. Immunol.
174
:
180
-194.
52
Datta, S. R., H. Dudek, X. Tao, S. Masters, H. Fu, Y. Gotoh, M. E. Greenberg.
1997
. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery.
Cell
91
:
231
-241.
53
del Peso, L., M. González-García, C. Page, R. Herrera, G. Nuñez.
1997
. Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt.
Science
278
:
687
-689.
54
Mok, C. L., G. Gil-Gómez, O. Williams, M. Coles, S. Taga, M. Tolaini, T. Norton, D. Kioussis, H. J. Brady.
1999
. Bad can act as a key regulator of T cell apoptosis and T cell development.
J. Exp. Med.
189
:
575
-586.
55
Adams, J. M., S. Cory.
1998
. The Bcl-2 protein family: arbiters of cell survival.
Science
281
:
1322
-1326.
56
Zha, J., H. Harada, E. Yang, J. Jockel, S. J. Korsmeyer.
1996
. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-XL.
Cell
87
:
619
-628.
57
Pettipher, R., T. T. Hansel, R. Armer.
2007
. Antagonism of the prostaglandin D2 receptors DP1 and CRTH2 as an approach to treat allergic diseases.
Nat. Rev. Drug Discov.
6
:
313
-325.
58
Park, C. S., S. M. Lee, S. T. Uh, H. T. Kim, Y. T. Chung, Y. H. Kim, B. W. Choi, S. H. Hue, H. B. Lee.
1993
. Soluble interleukin-2 receptor and cellular profiles in bronchoalveolar lavage fluid from patients with bronchial asthma.
J. Allergy Clin. Immunol.
91
:
623
-633.
59
Gyles, S. L., L. Xue, E. R. Townsend, F. Wettey, R. Pettipher.
2006
. A dominant role for chemoattractant receptor-homologous molecule expressed on T helper type 2 (Th2) cells (CRTH2) in mediating chemotaxis of CRTH2+CD4+ Th2 lymphocytes in response to mast cell supernatants.
Immunology
119
:
362
-368.
60
Xue, L., A. Barrow, R. Pettipher.
2009
. Interaction between prostaglandin D2 and chemoattractant receptor-homologous molecule expressed on Th2 cells mediates cytokine production by Th2 lymphocytes in response to activated mast cells.
Clin. Exp. Immunol.
156
:
126
-133.