Activation of PI3K is among the earliest signaling events observed in T cells after conjugate formation with antigen-presenting cells (APCs). The relevant PI3K catalytic isoform and relative contribution of the TcR and CD28 to PI3K activity at the immune synapse have not been determined unequivocally. Using a quantitative imaging-based assay, we show that the PI3K activity at the T cell–APC contact area is dependent on the p110δ, but not the p110γ, isoform of PI3K. CD28 enhanced PIP3 production at the T-cell synapse independently of its YMNM PI3K-recruitment motif that instead was required for efficient PKCθ recruitment. CD28 could partially compensate for the lack of p110δ activity during T-cell activation, which indicates that CD28 and p110δ act in parallel and complementary pathways to activate T cells. Consistent with this, CD28 and p110δ double-deficient mice were severely immune compromised. We therefore suggest that combined pharmaceutic targeting of p110δ activity and CD28 costimulation has potent therapeutic potential.

Full activation of T cells by antigen requires the integration of signals from several receptors on the T-cell surface that bind ligands on antigen-presenting cells (APCs). The T-cell receptor for antigen (TcR) is associated with CD3 and ζ chains that act as substrates and docking sites for nonreceptor tyrosine kinases. In CD4+ T helper cells (Th), CD4-bound Lck is brought in proximity with the TcR and phosphorylates the CD3 and ζ cytoplasmic immunoreceptor tyrosine-based activation motifs, thus providing docking sites for the ZAP-70 SH2 domains.1  ZAP-70 subsequently phosphorylates the transmembrane adapter protein LAT.2  When phosphorylated, LAT binds directly to the SH2 domains of PLCγ, Grb2, and Gads.3  Gads recruits SLP-76 to the complex and along with it Vav and Itk, each of which have the potential to recruit additional signaling proteins.3,4  These signaling events result in the activation of 2 key lipid-based second messenger signaling pathways catalyzed by PLCγ and PI3K, respectively.5 

PLCγ hydrolyzes PtdIns(4,5)P2 (PIP2) to yield the second messenger signaling molecules diacylglycerol (DAG) and inositol(1,4,5)P3 (IP3). DAG recruits both RasGRP and PKCθ to the plasma membrane, and these initiate the Ras-Erk and NF-κB signaling pathways, respectively.6  IP3 is released into the cytosol where it stimulates the release of Ca2+ into the cytoplasm from intracellular stores which stimulates the nuclear translocation of NFAT transcription factors.7 

PI3Ks catalyze the conversion of PIP2 to PtdIns(3,4,5)P3 (PIP3), which is resistant to PLC-hydrolysis and acts as a secondary signaling molecule, recruiting PH domain–containing proteins to the plasma membrane.8,9  T cells express 4 different catalytic isoforms of PI3K that have the capacity to generate PIP3.10  The class IA PI3Ks p110α, p110β, and p110δ associate with p85 regulatory subunits and are activated by tyrosine kinases. The class IB p110γ isoform is bound by a p101 or p84 regulatory subunit and is activated by G-protein–coupled receptors. Generation of PIP3 is among the earliest signals that can be observed when T cells respond to stimulation with antigen.11-15  It is still not known how the TcR activates PI3K, although a number of mechanisms are possible, including p85-SH2–mediated binding to YxxM motifs in the transmembrane adapter protein TRIM or to noncanonical phosphotyrosines in LAT, SLP-76, or ZAP-70.2,16-18  p85 may also associate by its SH3 domains with Src kinases.19,20  Finally, the p110 subunit may be activated directly by Ras.21-25 

CD28 can bind directly to PI3K by a well-characterized YMNM binding motif in its cytoplasmic domain.26-30  The CD28 ligands CD80 and CD86 can stimulate the association between CD28 and PI3K as well as PIP3 production in T-cell lines.27,31-33  However, both in Jurkat T cells and in primary mouse T cells, CD28 mutants that interfere with the capacity of CD28 to recruit PI3K retain costimulatory capacity in vitro and in vivo.29,34-36  In addition, CD28 costimulation is able to compensate for the lack of PI3K activity under conditions in which CD3-dependent proliferation is impaired.37,38  However, other studies had suggested a key role of PI3K in the context of CD28 costimulation.35,39,40  Therefore, the question of the relative contributions of the TcR and CD28 to PI3K activity at the immune synapse remains unresolved.10,41 

We have previously identified p110δ as a key class IA PI3K isoform in T cells.5,37,42  Genetic inactivation of p110δ in the p110δD910A/D910A mice abrogated the capacity of anti-CD3 and anti-CD28 antibodies to stimulate Akt phosphorylation.37,42  Moreover, proliferation of naive T cells, Th1 and Th2 differentiation, and Treg functions were attenuated in p110δD910A/D910A mice.5,37,42-44  However, p110α and p110β are also expressed in primary T cells and have the potential to generate PIP3, at least in vitro.37,42,45  The G-protein–activated PI3K isoform p110γ could potentially also contribute to PIP3 accumulation at the immune synapse. Indeed, p110γ−/− mice have been documented to have impaired T-cell function, and deficiency of both p110γ and p110δ leads to a block in T-cell development at the CD4CD8 DN3 stage.46-48  In this context it is worth considering that chemokine receptors can form complexes with the TcR.49,50  To date, the activation of p110δ and p110γ in T cells has only been determined indirectly in cells stimulated with antibodies and using the phosphorylation of Akt and other downstream proteins as surrogate markers for PI3K activity.

Here, we use a transgenic model system in which an AktPH-GFP fusion protein51  allows visualization and relative quantification of PIP3 generation in primary live CD4+ mouse T cells responding to stimulation with antigen presented by APCs. We also determined the relative contribution of CD28 and p110δ in the regulation of PKCθ and NF-κB. Finally, we have investigated the consequences of inhibiting both p110δ activity and CD28 costimulation on T-cell immunity in vitro and in vivo.

Mice

The p110δD910A/D910A (37), p110γ−/− (52), CD28−/− (53), CD28Y170 (29), OT-II TcR (54), and AktPH-GFP (51) mice have all been described previously and had been backcrossed to the C57BL/6 background for 5 to 10 generations. The CD28Y170F mice express a CD28Y170F transgene on the CD28−/− background.29  Compound mutant mice were generated by intercrossing. Mice were maintained under specific pathogen-free conditions. All protocols involving live animals were approved by the United Kingdom Home Office and institutional ethical review committees.

Reagents

IC87114 (55) was a kind gift from Joel Hayflick (ICOS, Seattle, WA). OVA323-339 was synthesized by Southampton Polypeptide, Southampton, United Kingdom. Other chemicals were purchased from Sigma-Aldrich (St Louis, MO) unless otherwise specified.

Antibodies

Antibodies used for fluorescence-activated cell sorting (FACS) and cell sorting were from BD PharMingen (San Diego, CA) or eBioscience (San Diego, CA), unless otherwise noted. The following clones were used: anti-CD3 (2C11), anti-CD28 (37.51), anti-CD4 (L3T4), anti-CD8 (YTS105.18.10), anti-CD69 (H1.2F3), anti-B220 (RA3-6B2), anti-CD49b (DX5), anti-CD80 (16-10A1), anti-CD86 (GL1), anti-MHC class II (M5/114.15.2), anti-Thy1.2 (TS; Sigma-Aldrich), anti-Vα2 (B20.1), and anti-Vβ5 (MR9-4). Antibodies for biochemical studies were as follows: anti-pAkt Ser473, anti-Akt (total) were from Cell Signaling Technology (Danvers, MA). For confocal analysis, anti-TcRβ (H57-597) was from BD Biosciences PharMingen (San Diego, CA), anti-p65/RelA, and anti-PKCθ polyclonal antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA), anti–rabbit Alexa Fluor 405 and anti–rabbit Alexa Fluor 488 were purchased from Invitrogen, Carlsbad, CA.

Cell purification

Naive T cells were purified from lymph nodes by magnetic sorting (Miltenyi Biotec, Auburn, CA). For total CD4+ T-cell purification, non-CD4+ T cells were labeled with FITC-conjugated anti-CD8, anti-CD69, anti-B220, anti-CD49b, and anti-MHC class II, followed by negative selection with anti-FITC magnetic cell sorting beads (Miltenyi Biotec). The resulting cells were more than 95% CD4+. OT-II TcR transgene expression in WT and p110δD910A/D910A backgrounds was similar (as determined by FACS analysis of cells stained anti–TcR-Vα2 and anti–TcR-Vβ5).

OT-II T-cell blasts were obtained by stimulation of cells from spleens with 1μM OVA323-339 peptide. After 6 days of stimulation, CD4+ T cells were purified by magnetic sorting and cultured with 20 ng/mL IL-2 and 1 ng/mL IL-7. Before any experiments, cells were washed to remove IL-2 and IL-7 and rested for 2 hours.

APCs used in this study were B cells purified from spleen cells after T-cell depletion with anti-mouse Thy1.2 (Sigma-Aldrich) and rabbit complement (Cedarlane, Burlington, ON), followed by purification of viable cells using Lympholyte-M (Cedarlane). APCs used in imaging experiments had been incubated overnight in LPS.

Fluorescence microscopy and image analysis

For live cell images, purified OT-II CD4+ T cells were added at a 1:1 ratio to LPS-activated APCs pulsed overnight with 10 μm OVA323-339 peptide. These interactions were imaged in RPMI supplemented with 0.5% FCS and 20mM HEPES pH 7.5 using an objective heater. For PI3K inhibition experiments, T cells were preincubated at 37°C for 15 minutes with 10 μM LY294002 before being added to the APCs. For p110δ-specific inhibition, the drug (IC87114 at 5 μM) was added during the acquisition. Images were taken every 15 seconds. Background noise was minimized by applying the ImageJ fast Fourier transform bandpass filter to the GFP images.

For fixed CD4+ T cell–APC conjugates analysis, APCs were plated on glass coverslips coated with poly(L-lysine) and were incubated at 37°C for 20 minutes, before naive CD4+ T cells were added to the APCs and incubated for 30 minutes. Conjugates were washed once in PBS and fixed in PBS plus 3.7% paraformaldehyde. Cells were permeabilized with 0.2% Triton X-100 in PBS for 4 minutes, blocked with 5% BSA-PBS for 30 minutes, and then stained with the appropriate antibodies. For the analysis of the NF-κB nuclear translocation, nuclei were additionally stained with 7-AAD (Invitrogen). The slides were mounted with fluorescent mounting medium and stored at 4°C before analysis. For 3-dimensional visualization of the PKCθ recruitment, image stacks of 0.3-μM intervals were acquired.

Images were taken with a Zeiss LSM510 META system consisting of a Zeiss Axiovert 200 microscope fitted with a Zeiss Plan-Achromat 63×/1.4 NA oil objective (Carl Zeiss, Jena, Germany). Images were analyzed using the ImageJ software (http://rsb.info.nih.gov/ij/). For the quantification of the fluorescence of the GFP at the membrane in fixed cells, we used a 2-color approach: in addition to the GFP, the CD4+ T cells were stained for TcR expression. The TcR staining was then used to create a mask corresponding to the membrane of the CD4+ T cells. The rest of the cell was considered as the “nonmembrane” region. The mask created was then used to determine the average intensity of fluorescence of GFP at the membrane, and this was compared with the average intensity of fluorescence of the nonmembrane region.

Proliferation assay

Proliferation assays were done in RPMI with 10% FCS, 5 × 10−5M β-mercaptoethanol, and antibiotics.

For anti-CD3 + anti-CD28, lymph node cells were stimulated at 2 × 105 per well with anti-CD3 (0.1 μg/mL) and anti-CD28 (1/100 supernatant from 37.51 hybridoma cultures). After 3 days, the cells were pulsed with [3H]-Thymidine for 6 hours.

For OT-II, purified CD4+ OT-II T cells were incubated with irradiated T cell–depleted spleen cells (both at 5 × 104 per well) and OVA323-339 peptide at the indicated concentrations. After 3 days, the cells were pulsed with [3H]-Thymidine for 6 hours.

Immunization

Each mouse was immunized with 100 μg DNP-KLH (Calbiochem, San Diego, CA) absorbed onto alum (Serva, Garden City Park, NY) on days 0 and 78. Blood was taken 11 and 12 days after the first and second immunizations, respectively, and serum was obtained. Antibody titers directed against DNP-BSA were determined using the SBA clonotyping system-HRP (Southern Biotechnology Associates, Birmingham, AL). The EC50 values were calculated from serial dilutions of serum using the GraphPad Prism program (GraphPad Software, San Diego, CA).

In vivo killing assay

Donor lymphocytes from C57BL/6 (B6) or (C57BL/6 × Cba/1) F1 mice were differentially labeled with 1.5 μM or 0.15 μM CFSE, respectively, and injected into recipient mice on the C57BL/6 background (5 × 106 cells per recipient). The proportion of F1 to B6 donors were analyzed in blood taken from the tail vein on days 1 to 4 after transfer. The relative killing efficiency in each mouse was normalized so that the ratio on day 1 was 1:1.

Statistical analysis

The data included in the analysis of the fluorescence at the plasma membrane of AktPH-GFP start 3 time windows before the contact between the cells up to 406 seconds. The time at which the plateau was reached was determined graphically for each individual mouse, and the values included in the calculation of the mean fluorescence for each mouse correspond to the 15 subsequent frames. This number was chosen because it corresponds to the minimum plateau length for CD28Y170F mice which took the longest to stabilize. The choice was also made to provide balanced samples more suitable for statistical analysis. On the basis of the mean fluorescence values for each animal, data were tested for normality, and 2 cells (WT + LY294002) were consistently identified as outliers. The natural log of the fluorescent ratios obtained from 15 plateau measurements (shown in Figure 1C) was calculated. Statistical analysis of the ratios between membrane and cytosol fluorescence in Figure 1C and Figure 2B and C was analyzed by one-way ANOVA followed by Tukey multiple comparison test using GraphPad Prism software (GraphPad Software).

Regulation of PIP3 production at the immune synapse

The phosphorylation of PIP2 at the plasma membrane by PI3K produces PIP3 that acts as a docking site for the PH domain of Akt. Thus, the translocation of the Akt PH domain, fused to GFP, allows real-time visualization of PIP3 generated at the immune synapse. Therefore, to monitor the relative contributions of p110δ, p110γ, and CD28 to PIP3 metabolism at the immune synapse, we crossed p110δD910A/D910A, p110γ−/−, CD28−/−, and CD28Y170 mice to mice expressing OVA323-339–specific OT-II TcR and AktPH-GFP transgenes. The AktPH-GFP transgene did not interfere with T-cell development, Akt phosphorylation, or T-cell proliferation56  (Figure S1, available on the Blood website; see the Supplemental Materials link at the top of the online article). CD4+ T-cell blasts from each of these mouse lines were incubated with WT APCs expressing MHC II-Ab bearing the OVA323-339 peptide. AktPH-GFP accumulation in the T cells at the contact areas with the APCs was monitored in real time, and the ratio of AktPH-GFP at the contact area relative to the rest of the cell was determined.

PIP3 accumulated at the contact area between the T cells and the APCs within seconds of conjugate formation and remained concentrated at the contact area for the duration of the recordings (usually 10 minutes; Figure 1A,B). Figure 1B shows the average intensity of PIP3 for the different conditions over time, whereas Figure 1C shows the average of 15 measurements for each cell once they had reached a plateau level of activation (1-2 minutes after conjugate formation). Pretreatment with LY294002, a pan-PI3K inhibitor, reduced AktPH-GFP translocation nearly to base level (ie, where the ratio = 1) in the majority of T cells.

Figure 1

p110δ is essential for PIP3 accumulation to the plasma membrane. (A) Images showing fluorescence and transmitted light from a movie of AktPH-GFP+ WT, p110δD910A/D910A, p110γ−/−, CD28−/−, and CD28Y170F CD4+ T-cell blasts interacting with LPS-activated APCs presenting OVA323-339 peptide. In some experiments, WT cells were pretreated with PI3K inhibitor (LY294002) for 15 minutes before stimulation. (B) Quantification of AktPH-GFP redistribution in WT (n = 11), LY294002 pretreated WT (n = 8), p110δD910A/D910A (n = 9), p110γ−/− (n = 12), CD28−/− (n = 9), and CD28Y170F (n = 7) CD4+ T cells. A few LY294002-treated T cells showed apparently normal AktPH-GFP accumulation (Figure S3). It is possible that a subset of T cells express sufficient levels of multidrug resistance protein transporter and could potentially thus evade inhibition.81,82  Therefore, to establish a realistic baseline for minimal PI3K activity, these outliers were excluded from the analysis, but they are shown in Figure S3. The recruitment of AktPH-GFP at the plasma membrane is expressed as a ratio between the fluorescence measured at the contact area and the average fluorescence within the cell. Fluorescence was measured on at least 2 independent days. (C) Average ratio of PIP3 at membrane during the plateau phase of the response (approximately 40-400 seconds). The mean fluorescence values were determined as described in “Methods” (**P < .01, ***P < .001). (D) Acute inhibition of PI3K activity with the p110δ-selective inhibitor IC87114. Two T cell–B cell conjugates are shown after the addition of IC87114 as indicated by arrows. PIP3 depletion from the membrane occurred rapidly in both cases.

Figure 1

p110δ is essential for PIP3 accumulation to the plasma membrane. (A) Images showing fluorescence and transmitted light from a movie of AktPH-GFP+ WT, p110δD910A/D910A, p110γ−/−, CD28−/−, and CD28Y170F CD4+ T-cell blasts interacting with LPS-activated APCs presenting OVA323-339 peptide. In some experiments, WT cells were pretreated with PI3K inhibitor (LY294002) for 15 minutes before stimulation. (B) Quantification of AktPH-GFP redistribution in WT (n = 11), LY294002 pretreated WT (n = 8), p110δD910A/D910A (n = 9), p110γ−/− (n = 12), CD28−/− (n = 9), and CD28Y170F (n = 7) CD4+ T cells. A few LY294002-treated T cells showed apparently normal AktPH-GFP accumulation (Figure S3). It is possible that a subset of T cells express sufficient levels of multidrug resistance protein transporter and could potentially thus evade inhibition.81,82  Therefore, to establish a realistic baseline for minimal PI3K activity, these outliers were excluded from the analysis, but they are shown in Figure S3. The recruitment of AktPH-GFP at the plasma membrane is expressed as a ratio between the fluorescence measured at the contact area and the average fluorescence within the cell. Fluorescence was measured on at least 2 independent days. (C) Average ratio of PIP3 at membrane during the plateau phase of the response (approximately 40-400 seconds). The mean fluorescence values were determined as described in “Methods” (**P < .01, ***P < .001). (D) Acute inhibition of PI3K activity with the p110δ-selective inhibitor IC87114. Two T cell–B cell conjugates are shown after the addition of IC87114 as indicated by arrows. PIP3 depletion from the membrane occurred rapidly in both cases.

Close modal

p110δD910A/D910A T cells showed greatly reduced AktPH-GFP recruitment to the T-B contact area, suggesting that p110δ is the main PI3K isoform that produces PIP3 at the immune synapse (Figure 1A,B). Although moderate recruitment of PIP3 could be observed in some p110δD910A/D910A T cells, the difference between T cells treated with LY294002 and p110δD910A/D910A T cells did not reach statistical significance in these experiments (Figure 1C). Addition of IC87114, a small molecule inhibitor with high selectivity for p110δ, (55 ) after conjugate formation returned PIP3 levels to baseline, showing that continuous p110δ activity at the contact area is required to sustain elevated PIP3 levels (Figure 1D). p110γ−/− T cells showed a more variable but not significantly reduced AktPH-GFP recruitment (Figure 1A-C). By contrast, p110γ but not p110δ was critical for AktPH-GFP recruitment to the plasma membrane induced by the chemokine CXCL-12 (Figure 2A,B).

Figure 2

CXCL12-induced (500 ng/mL) accumulation of PIP3 at the plasma membrane of CD4+ T-cell blasts is dependent on p110γ but not p110δ. (A) Representative images showing the translocation of the AktPH-GFP probe to the plasma membrane. (B) Graphical representation of the percentage of total fluorescence at the plasma membrane as function of time. Data are representative of 2 independent experiments (WT, n = 6; p110δD910A/D910A, n = 4; p110γ−/−, n = 5).

Figure 2

CXCL12-induced (500 ng/mL) accumulation of PIP3 at the plasma membrane of CD4+ T-cell blasts is dependent on p110γ but not p110δ. (A) Representative images showing the translocation of the AktPH-GFP probe to the plasma membrane. (B) Graphical representation of the percentage of total fluorescence at the plasma membrane as function of time. Data are representative of 2 independent experiments (WT, n = 6; p110δD910A/D910A, n = 4; p110γ−/−, n = 5).

Close modal

In CD28−/− T cells, PIP3 accumulation was observed early in the response, but was not sustained, nor did it reach WT levels (Figure 1A,B). Surprisingly, CD28Y170F T cells reached a plateau level of PIP3 accumulation equivalent to that of WT T cells, albeit with somewhat delayed kinetics (Figure 1A-C). These results indicate that CD28 can promote sustained PIP3 generation at the immune synapse independently of its capacity to bind p85/p110δ directly. The imaging-based experiments were corroborated by biochemical analysis because we found that Akt was robustly phosphorylated in CD28−/− and CD28Y170F T cells using anti-CD3 and anti-CD28, whereas, as previously reported,37,42  virtually no Akt phosphorylation was detected in p110δD910A/D910A T cells (Figure S1C,D). We conclude from these experiments that p110δ is the main PI3K isoform responsible for generating PIP3 at the immune synapse and that p110δ can be activated without being recruited to Y170 in the CD28 cytoplasmic domain.

Regulation of PKCθ and NF-κB

We next investigated how reduced PIP3 production at the synapse might contribute to the regulation of PLCγ-dependent signaling. During the formation of an immune synapse, production of DAG by PLCγ leads to the recruitment of PKCθ that localizes in the center of the contact area between the T cell and the APC.57  CD28 and PI3K have been suggested to influence the recruitment of PKCθ to the immune synapse.58  We therefore examined PKCθ localization in the various mutant T cells to determine the involvement of CD28 and p110δ during the formation of the immune synapse. At the same time we observed PIP3 recruitment using the AktPH-GFP probe. In contrast to the experiments presented in Figure 1, these experiments were done using naive T cells that had been incubated with peptide-loaded APCs for 30 minutes and then fixed. After fixation, the cells were stained with antibodies to the TcR and PKCθ (Figure 3A). These experiments were consistent with those presented in Figure 1 by revealing that p110δ, but not p110γ, was required for membrane accumulation of PIP3 and by showing that PIP3 recruitment was defective in CD28−/−, but not CD28Y170F T cells (Figure 3A,B). Thus, the lack of requirement for the CD28 YMNM motif to activate PI3K is true both in naive and in previously activated T cells. In addition, these experiments showed that neither of the PI3K mutants showed defect in PKCθ accumulation at the plasma membrane (Figure 3C). However, the recruitment of PKCθ was attenuated in CD28−/− T cells and to a lesser extent in CD28Y170F T cells (Figure 3C). Moreover, CD28−/− and CD28Y170F T cells showed a more diffuse localization of PKCθ within the immune synapse than in conjugates with WT or p110δD910A/D910A T cells (Figure 3D). Thus, CD28 contributes to PKCθ localization by a mechanism that is at least partially dependent on the YMNM motif, but which is independent of p110δ activity.

Figure 3

CD28, but not p110δ, regulates PKCθ localization at the synapse. (A) Conjugates between naive WT, WT + LY294002, p110δD910A/D910A, p110γ−/−, CD28−/−, or CD28Y170F CD4+ T cells and OVA323-339–pulsed APCs were stained with antibodies against PKCθ and the TcRβ. Representative images of the distribution of AktPH-GFP, TcR, and PKCθ during conjugate formation are shown. (B) Quantification of AktPH-GFP recruitment at the plasma membrane (WT, n = 72; WT + LY294002, n = 31; p110δD910A/D910A, n = 53; p110γ−/−, n = 23; CD28−/−, n = 51; CD28Y170F, n = 62), determined as described in “Methods” (**P < .01, ***P < .001). (C) Recruitment of PKCθ to the contact area (WT, n = 76; WT + LY294002, n = 24; p110δD910A/D910A, n = 28; p110γ−/−, n = 28; CD28−/−, n = 33; CD28Y170F, n = 67), expressed as the ratio between the fluorescence at the contact area and the fluorescence within the cell. Only the means of the values for the CD28−/− and CD28Y170F values were significantly different from the WT values (***P < .001). (D) A 3-dimensional reconstruction of the contact area was done and representative pictures of the interface projection and the mean area (± SEM, *P < .05, **P < .001, Student 2-tailed t test; WT, n = 38; p110δD910A/D910A, n = 18; CD28−/−, n = 23; CD28Y170F, n = 21) occupied by PKCθ at the immune synapse are shown. Data are representative of 2 independent experiments. (E) The YMNM motif of CD28 is not required for NF-κB nuclear translocation. Conjugates between WT, CD28−/−, or CD28Y170F CD4+ T cells and OVA323-339–pulsed APCs were stained for p65 NF-κ, and nuclei were stained with 7-AAD. The conjugates were scored for the frequency of nuclear localization of NF-κB. The p110δ inhibitor IC87114 blocked NF-κB nuclear translocation in CD28−/− and CD28Y170F T cells. Data represent mean (± SEM) with more than 30 cells analyzed in each of 3 experiments.

Figure 3

CD28, but not p110δ, regulates PKCθ localization at the synapse. (A) Conjugates between naive WT, WT + LY294002, p110δD910A/D910A, p110γ−/−, CD28−/−, or CD28Y170F CD4+ T cells and OVA323-339–pulsed APCs were stained with antibodies against PKCθ and the TcRβ. Representative images of the distribution of AktPH-GFP, TcR, and PKCθ during conjugate formation are shown. (B) Quantification of AktPH-GFP recruitment at the plasma membrane (WT, n = 72; WT + LY294002, n = 31; p110δD910A/D910A, n = 53; p110γ−/−, n = 23; CD28−/−, n = 51; CD28Y170F, n = 62), determined as described in “Methods” (**P < .01, ***P < .001). (C) Recruitment of PKCθ to the contact area (WT, n = 76; WT + LY294002, n = 24; p110δD910A/D910A, n = 28; p110γ−/−, n = 28; CD28−/−, n = 33; CD28Y170F, n = 67), expressed as the ratio between the fluorescence at the contact area and the fluorescence within the cell. Only the means of the values for the CD28−/− and CD28Y170F values were significantly different from the WT values (***P < .001). (D) A 3-dimensional reconstruction of the contact area was done and representative pictures of the interface projection and the mean area (± SEM, *P < .05, **P < .001, Student 2-tailed t test; WT, n = 38; p110δD910A/D910A, n = 18; CD28−/−, n = 23; CD28Y170F, n = 21) occupied by PKCθ at the immune synapse are shown. Data are representative of 2 independent experiments. (E) The YMNM motif of CD28 is not required for NF-κB nuclear translocation. Conjugates between WT, CD28−/−, or CD28Y170F CD4+ T cells and OVA323-339–pulsed APCs were stained for p65 NF-κ, and nuclei were stained with 7-AAD. The conjugates were scored for the frequency of nuclear localization of NF-κB. The p110δ inhibitor IC87114 blocked NF-κB nuclear translocation in CD28−/− and CD28Y170F T cells. Data represent mean (± SEM) with more than 30 cells analyzed in each of 3 experiments.

Close modal

PKCθ is essential for the nuclear translocation of NF-κB.59  We have previously shown that the nuclear translocation of p65 NF-κB is normal in p110δD910A/D910A T cells stimulated with peptide antigen.42  To our surprise, p65 NF-κB translocation was also readily observed in the absence of CD28 signaling (Figure 2E). However, when CD28−/− or CD28Y170F T cells were incubated with the p110δ-specific inhibitor IC87114, p65 NF-κB nuclear translocation was attenuated by approximately 50%. The latter is consistent with our previously observed defect in anti-CD3 but not anti-CD3 plus anti-CD28–stimulated NF-κB translocation in p110δD910A/D910A T cells.42  These results show that p110δ and CD28 cooperatively regulate p65 NF-κB translocation to some extent, but neither input signal is essential for p65 NF-κB nuclear translocation.

CD28 and p110δ complement each other to provide full T-cell stimulation

Our results indicate that CD28 is not essential for p110δ activation and that CD28 and p110δ, at least in part, control parallel signaling pathways. A prediction from these observations is that dual inhibition of CD28 and p110δ should act synergistically. However, if CD28 were a key regulator of p110δ activation, then the prediction would be that the inhibition of p110δ would not potentiate inhibition of CD28 signaling alone. To test these predictions, we generated p110δD910A/D910ACD28−/− and p110δD910A/D910ACD28Y170F double mutant mice. In response to anti-CD3 and anti-CD28 stimulation, the p110δD910A/D910A, CD28Y170F, or p110δD910A/D910ACD28Y170F mutant T cells showed intact proliferative responses (Figure 4A). These results exclude the possibility that p110α or p110β could be recruited to CD28 Y170 to compensate for the lack of p110δ activity and promote proliferation in p110δD910A/D910A T cells. Consistent with previous results,37  in response to anti-CD3 alone, p110δ was required for optimal proliferation (compare the response of CD28−/− T cells with p110δD910A/D910ACD28−/− T cells). We have previously shown that proliferation of p110δD910A/D910A T cells in response to peptide presented by APCs is impaired, suggesting that the natural ligand for CD28, in contrast to antibodies directed against CD28, is insufficient to rescue proliferation to WT levels in the absence of p110δ activity.37,42  By contrast, CD28Y170F T cells proliferate normally regardless of the stimulus.29,60  In p110δD910A/D910ACD28−/− double-deficient T cells, the proliferative response was abolished (Figure 4B). However, the expression of the CD28Y170F transgene partially rescued proliferation of p110δD910A/D910ACD28−/− T cells stimulated with 1 μm, but not 0.1 μm, peptide (Figure 4B). Thus, the CD28 YMNM motif may compensate in part for the lack of p110δ activity under conditions of weak TcR stimulation, but it is not required in p110δ-sufficient T cells.

Figure 4

Proliferation of CD28 and p110δ double mutant T cells. (A) Anti-CD3– and anti-CD28–dependent proliferation of WT, p110δD910A/D910A, CD28−/−, CD28Y170F, p110δD910A/D910ACD28−/−, and p110δD910A/D910ACD28Y170F lymph node T cells stimulated with 0.1 μg/mL anti-CD3 and with anti-CD28 37.51 hybridoma supernatant (1/100). (B) Proliferation of T cells from the mutants OT-II+ transgenic WT, p110δD910A/D910A, CD28−/−, CD28Y170F, p110δD910A/D910ACD28−/−, and p110δD910A/D910ACD28Y170F CD4+ T cells in response to 0.01μM, 0.1μM, or 1.0μM OVA323-339 peptide.

Figure 4

Proliferation of CD28 and p110δ double mutant T cells. (A) Anti-CD3– and anti-CD28–dependent proliferation of WT, p110δD910A/D910A, CD28−/−, CD28Y170F, p110δD910A/D910ACD28−/−, and p110δD910A/D910ACD28Y170F lymph node T cells stimulated with 0.1 μg/mL anti-CD3 and with anti-CD28 37.51 hybridoma supernatant (1/100). (B) Proliferation of T cells from the mutants OT-II+ transgenic WT, p110δD910A/D910A, CD28−/−, CD28Y170F, p110δD910A/D910ACD28−/−, and p110δD910A/D910ACD28Y170F CD4+ T cells in response to 0.01μM, 0.1μM, or 1.0μM OVA323-339 peptide.

Close modal

We next investigated the genetic interactions between CD28, CD28Y170F, and p110δ in vivo. Each of the 6 lines described in Figure 4A were immunized with KLH-DNP adsorbed onto alum and rechallenged 78 days later. DNP-specific antibody titers were measured 11 or 12 days after each immunization. This experiment confirmed our previous reports,29,37  showing defective primary immune humoral immune responses in p110δD910A/D910A mice and in CD28−/− mice, with antibody responses in CD28Y170F mice being intact (Figure 5A). The antibody responses of CD28−/−p110δD910A/D910A mice were dramatically reduced, whereas p110δD910A/D910ACD28Y170F mice showed equivalent immune responses to p110δD910A/D910A mice. During recall responses, the antibody titers from WT, CD28−/−, and CD28Y170F mice were enhanced by 2 orders of magnitude, whereas p110δD910A/D910A mice did not show enhanced secondary responses. Although the impaired humoral immune response is caused by defective antigen receptor signaling in p110δD910A/D910A B cells, humoral immune responses are also attenuated in mice in which PI3K signaling has been selectively eliminated in the T-cell lineage.38  We conclude that p110δ and CD28 complement each other during the regulation of humoral immune responses and that p110δ, but not CD28, is essential for enhanced recall responses.

Figure 5

Impaired humoral immune responses in p110δD910A/D910A CD28−/− mice. (A) WT, p110δD910A/D910A, CD28−/−, CD28Y170F, p110δD910A/D910A CD28−/−, and p110δD910A/D910ACD28Y170F mice were immunized with DNP-coupled KLH adsorbed onto alum. The mice were rechallenged with the same dose of antigen 78 days later. Anti-DNP–specific antibodies were detected by enzyme-linked immunoabsorbent assay. Each dot represents one mouse. (B) CD28 and p110δ are indispensable for efficient killing of allogeneically mismatched lymphocytes in vivo. Recipient mice on the B6 background (H-2b) were injected with a mixture of 5 × 106 CFSEhigh B6 lymphocytes and CFSElow CB6 F1 lymphocytes (H-2b/k). The ratio of CFSElow to CFSEhigh was normalized to 1 on day 1, and the average from 3 recipients from each genotype is shown (± SD).

Figure 5

Impaired humoral immune responses in p110δD910A/D910A CD28−/− mice. (A) WT, p110δD910A/D910A, CD28−/−, CD28Y170F, p110δD910A/D910A CD28−/−, and p110δD910A/D910ACD28Y170F mice were immunized with DNP-coupled KLH adsorbed onto alum. The mice were rechallenged with the same dose of antigen 78 days later. Anti-DNP–specific antibodies were detected by enzyme-linked immunoabsorbent assay. Each dot represents one mouse. (B) CD28 and p110δ are indispensable for efficient killing of allogeneically mismatched lymphocytes in vivo. Recipient mice on the B6 background (H-2b) were injected with a mixture of 5 × 106 CFSEhigh B6 lymphocytes and CFSElow CB6 F1 lymphocytes (H-2b/k). The ratio of CFSElow to CFSEhigh was normalized to 1 on day 1, and the average from 3 recipients from each genotype is shown (± SD).

Close modal

We next examined the role of CD28 and p110δ in cytotoxic immune responses. To this end, recipient mice on the C57BL/6 (B6) background (H-2b) were injected with differentially labeled lymph node cells from a C57BL/6 or a C57BL6 × Cba/1 (F1) donor (H-2b/k). The proportion of circulating F1 cells to B6 cells was monitored during the next 4 days. p110δD910A/D910A mice rejected the F1 cells more slowly than did WT hosts, whereas CD28−/− mice showed accelerated rejection consistent with a nonessential role for CD28 in CD8 T cell–dependent killing of allogeneic grafts61,62  (Figure 5B). p110δD910A/D910ACD28−/− mice rejected the F1 cells at a rate that was indistinguishable from WT mice, which also could be considered intermediate between that of p110δD910A/D910A and CD28−/− mice. We conclude that CD28 and p110δ are less important for CD8 T cell–dependent cytotoxic immune responses than they are for humoral immune responses.

In this study, we have used an imaging approach to visualize PI3K signaling in single T cells in response to antigen stimulation. This approach circumvents some of the potential artifacts associated with antibody stimulation of T cells and presents a more dynamic view of PI3K activity in real time. Moreover, by measuring the ratio of the fluorescence near the membrane to that in the cytosol, we were able to assess the relative PI3K signaling strength in different PI3K or CD28 mutant T cells. Using this approach, we show that p110δ is the main PI3K isoform responsible for accumulation of PIP3 at the immune synapse and that p110γ is required for chemokine- but not antigen-dependent PI3K activity. In contrast to a previous report,46  we did not observe defects in proliferation or cytokine secretion in p110γ−/− CD4+ T cells (data not shown). Moreover, pharmacologic inhibition of p110δ, but not of p110γ, led to reduced proliferation and cytokine secretion by CD4+ T cells.63  The previously reported defective activation of p110γ−/− T cells may have been secondary to an undefined genetic lesion that also lead to colorectal carcinomas, a phenotype that has not been observed in other p110γ−/− strains (including the one used in this study).64,65  In addition, dendritic cell migration is impaired in p110γ−/− mice which may have contributed to impaired T-cell responses in vivo.66  Nonetheless, T-cell development is largely normal in p110δD910A/D910A mice and p110γ−/− mice, whereas p110δp110γ double-knockout mice have a pronounced defect in TcRβ-dependent selection of double-negative thymocytes.47,48  These results suggest that the functional redundancy between p110δ and p110γ is more pronounced during thymocyte selection than during activation of mature T cells and implicates the contribution of an undefined G-protein–coupled receptor at this thymic selection stage.

Although CD28 also contributes to PI3K activity and PIP3 accumulation at the synapse, direct interaction between PI3K and CD28 via the YMNM motif was not required. One possibility could be that CD28 could interact with PI3K by a distinct motif. Indeed, the CD28 cytoplasmic domain contains 4 tyrosines, and it has been suggested the most C-terminal tyrosine (with a YRS motif) contributes to the CD28-p85 association.67  However, this tyrosine is thought to stabilize the interaction between CD28 and p85 bound to the YMNM motif rather than binding independently to PI3K. Thus, a Y to F mutation of the YMNM motif completely abrogated CD28-dependent PI3K activity, whereas a mutation of the YRS delayed and attenuated the PI3K response.27,67 

In addition to the tyrosine-based motifs, the cytoplasmic domain of CD28 contains 2 PxxP motifs that can interact with SH3 domains.68-72  In particular, CD28 costimulation has been shown to sustain Lck activity in the immune synapse by an interaction involving the C-terminal PxxP motif in the cytoplasmic domain of CD28.73  In CD4+ T cells, Lck is recruited to the immune synapse by CD4; however, the association of CD4 with the immune synapse is transient, and, in the absence of CD28, Lck activity is not sustained.73  Consistent with these results, mice with mutations in the C-terminal PxxP motif in CD28 show impaired costimulation in vitro and in vivo.34,74  Because the CD28Y170F T cells showed intact accumulation of PIP3 at the immune synapse, the capacity of CD28 to sustain Lck activity is a plausible mechanism for how CD28 maintains PI3K signaling, the assumption being that Lck phosphorylates other docking sites for p85-p110δ at the immune synapse. CD28−/− T cells have been shown capable of initiating but not sustaining intracellular Ca2+ flux after activation, which is similar to what we observed for PIP3 accumulation.75  Thus, the reduced PIP3 accumulation at the immune synapse in CD28−/− T cells probably reflects a more general role for CD28 in enhancing TcR signaling than a specific role for CD28 in regulating PI3K activation.76 

Our results do not exclude a role for CD28 in the direct recruitment of p85-p110δ or for Grb2 of Gads under different circumstances. Indeed, we found a requirement for the YMNM motif in concentrating the accumulation of PKCθ at the immune synapse. Inactivation of p110δ did not affect PKCθ localization, suggesting that PKCθ localization may be regulated by Grb2 or Gads that binds to the same tyrosine. Superagonistic antibodies against CD28 stimulated T-cell responses that were dependent on the YMNM motif but not on PI3K77  (data not shown). We therefore propose that the CD28-Grb2/Gads association can regulate PKCθ and other pathways normally regulated by the TcR, but that this capacity of CD28 only becomes functionally relevant when CD28 signaling occurs in the absence of agonist-induced TcR signaling.77  The CD28 YMNM motif can also influence T-cell trafficking to nonlymphoid organs in vivo.60 

Our results showing synergy between CD28 and p110δ have potential clinical relevance, because inhibition of CD28 and PI3K signaling is achievable pharmaceutically. CTLA4Ig (abatacept) has recently received FDA approval for the treatment of rheumatoid arthritis, and the related inhibitor belatacept is in clinical trials for transplantation.78  However, CD28 blockade alone is unlikely to be sufficient to prevent autoimmunity or graft rejection in most cases. More recently, p110δ-selective small molecule inhibitors have been developed as candidate drugs to treat autoimmunity and inflammation.55,79,80  Our data suggest that dual inhibition of CD28 and p110δ may achieve more potent alleviation of pathologic immune responses than can be achieved with either inhibitor alone.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Anne Segonds-Pichon for expert statistical analysis, Simon Walker for assistance with microscopy, and members of the small animal barrier unit for animal husbandry and immunizations. We thank Dalya Soond, Martin Turner, Jacques Nunès, and Bart Vanhaesebroeck for their constructive comments on the manuscript.

This work was supported by grants from the Biotechnology and Biological Sciences Research Council (BBSRC; JF19128 and BBC5098901) and from the Juvenile Diabetes Research Foundation (5-2006-229).

Contribution: F.G. designed and performed research and analyzed and interpreted data; D.T.P. and J.L.E. performed research; E.H., R.R., and T.S. contributed vital reagents; K.O. designed and performed research, analyzed and interpreted data, and drafted the manuscript.

Conflict-of-interest disclosure: One of the authors (K.O.) is employed as consultant by PIramed. The other authors declare no competing financial interests.

Correspondence: Klaus Okkenhaug, Laboratory of Lymphocyte Signaling and Development, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom; e-mail: klaus.okkenhaug@bbsrc.ac.uk.

1
Iwashima
 
M
Irving
 
BA
van Oers
 
NS
Chan
 
AC
Weiss
 
A
Sequential interactions of the TcR with two distinct cytoplasmic tyrosine kinases.
Science
1994
, vol. 
263
 (pg. 
1136
-
1139
)
2
Zhang
 
W
Sloan
 
LJ
Kitchen
 
J
Trible
 
RP
Samelson
 
LE
LAT: the ZAP-70 tyrosine kinase substrate that links T cell receptor to cellular activation.
Cell
1998
, vol. 
92
 (pg. 
83
-
92
)
3
Samelson
 
LE
Signal transduction mediated by the T cell antigen receptor: the role of adapter proteins.
Annu Rev Immunol
2002
, vol. 
20
 (pg. 
371
-
394
)
4
Koretzky
 
GA
Abtahian
 
F
Silverman
 
MA
SLP76 and SLP65: complex regulation of signalling in lymphocytes and beyond.
Nat Rev Immunol
2006
, vol. 
6
 (pg. 
67
-
78
)
5
Okkenhaug
 
K
Ali
 
K
Vanhaesebroeck
 
B
Antigen receptor signalling: a distinctive role for the p110δ isoform of PI3K.
Trends Immunol
2007
, vol. 
28
 (pg. 
80
-
87
)
6
Spitaler
 
M
Cantrell
 
DA
Protein kinase C and beyond.
Nat Immunol
2004
, vol. 
5
 (pg. 
785
-
790
)
7
Gallo
 
EM
Cante-Barrett
 
K
Crabtree
 
GR
Lymphocyte calcium signaling from membrane to nucleus.
Nat Immunol
2006
, vol. 
7
 (pg. 
25
-
32
)
8
Hawkins
 
PT
Anderson
 
KE
Davidson
 
K
Stephens
 
LR
Signalling through Class I PI3Ks in mammalian cells.
Biochem Soc Trans
2006
, vol. 
34
 (pg. 
647
-
662
)
9
Vanhaesebroeck
 
B
Leevers
 
SJ
Ahmadi
 
K
, et al. 
Synthesis and function of 3-phosphorylated inositol lipids.
Annu Rev Biochem
2001
, vol. 
70
 (pg. 
535
-
602
)
10
Okkenhaug
 
K
Vanhaesebroeck
 
B
PI3K in lymphocyte development, differentiation and activation.
Nat Rev Immunol
2003
, vol. 
3
 (pg. 
317
-
330
)
11
Fabre
 
S
Lang
 
V
Harriague
 
J
, et al. 
Stable activation of phosphatidylinositol 3-kinase in the T cell immunological synapse stimulates Akt signaling to FoxO1 nuclear exclusion and cell growth control.
J Immunol
2005
, vol. 
174
 (pg. 
4161
-
4171
)
12
Costello
 
PS
Gallagher
 
M
Cantrell
 
DA
Sustained and dynamic inositol lipid metabolism inside and outside the immunological synapse.
Nat Immunol
2002
, vol. 
3
 (pg. 
1082
-
1089
)
13
Harriague
 
J
Bismuth
 
G
Imaging antigen-induced PI3K activation in T cells.
Nat Immunol
2002
, vol. 
3
 (pg. 
1090
-
1096
)
14
Kane
 
LP
Mollenauer
 
MN
Weiss
 
A
A proline-rich motif in the C terminus of akt contributes to its localization in the immunological synapse.
J Immunol
2004
, vol. 
172
 (pg. 
5441
-
5449
)
15
Huppa
 
JB
Gleimer
 
M
Sumen
 
C
Davis
 
MM
Continuous T cell receptor signaling required for synapse maintenance and full effector potential.
Nat Immunol
2003
, vol. 
4
 (pg. 
749
-
755
)
16
Bruyns
 
E
Marie-Cardine
 
A
Kirchgessner
 
H
, et al. 
T cell receptor (TcR) interacting molecule (TRIM), a novel disulfide-linked dimer associated with the TcR-CD3-zeta complex, recruits intracellular signaling proteins to the plasma membrane.
J Exp Med
1998
, vol. 
188
 (pg. 
561
-
575
)
17
Moon
 
KD
Post
 
CB
Durden
 
DL
, et al. 
Molecular basis for a direct interaction between the Syk protein-tyrosine kinase and phosphoinositide 3-kinase.
J Biol Chem
2005
, vol. 
280
 (pg. 
1543
-
1551
)
18
Shim
 
EK
Moon
 
CS
Lee
 
GY
Ha
 
YJ
Chae
 
SK
Lee
 
JR
Association of the Src homology 2 domain-containing leukocyte phosphoprotein of 76 kD (SLP-76) with the p85 subunit of phosphoinositide 3-kinase.
FEBS Lett
2004
, vol. 
575
 (pg. 
35
-
40
)
19
Prasad
 
KV
Janssen
 
O
Kapeller
 
R
Raab
 
M
Cantley
 
LC
Rudd
 
CE
Src-homology 3 domain of protein kinase p59fyn mediates binding to phosphatidylinositol 3-kinase in T cells.
Proc Natl Acad Sci U S A
1993
, vol. 
90
 (pg. 
7366
-
7370
)
20
Pleiman
 
CM
Hertz
 
WM
Cambier
 
JC
Activation of phosphatidylinositol-3′ kinase by Src-family kinase SH3 binding to the p85 subunit.
Science
1994
, vol. 
263
 (pg. 
1609
-
1612
)
21
Rodriguez-Viciana
 
P
Warne
 
PH
Dhand
 
R
, et al. 
Phosphatidylinositol-3-OH kinase as a direct target of Ras.
Nature
1994
, vol. 
370
 (pg. 
527
-
532
)
22
Suire
 
S
Hawkins
 
P
Stephens
 
L
Activation of phosphoinositide 3-kinase gamma by Ras.
Curr Biol
2002
, vol. 
12
 (pg. 
1068
-
1075
)
23
Suire
 
S
Condliffe
 
AM
Ferguson
 
GJ
, et al. 
Gbetagammas and the Ras binding domain of p110gamma are both important regulators of PI(3)Kgamma signalling in neutrophils.
Nat Cell Biol
2006
, vol. 
8
 (pg. 
1303
-
1309
)
24
Rodriguez-Viciana
 
P
Sabatier
 
C
McCormick
 
F
Signaling specificity by Ras family GTPases is determined by the full spectrum of effectors they regulate.
Mol Cell Biol
2004
, vol. 
24
 (pg. 
4943
-
4954
)
25
Wabnitz
 
GH
Nebl
 
G
Klemke
 
M
Schroder
 
AJ
Samstag
 
Y
Phosphatidylinositol 3-kinase functions as a Ras effector in the signaling cascade that regulates dephosphorylation of the actin-remodeling protein cofilin after costimulation of untransformed human T lymphocytes.
J Immunol
2006
, vol. 
176
 (pg. 
1668
-
1674
)
26
Prasad
 
KV
Cai
 
YC
Raab
 
M
, et al. 
T-cell antigen CD28 interacts with the lipid kinase phosphatidylinositol 3-kinase by a cytoplasmic Tyr(P)-Met-Xaa-Met motif.
Proc Natl Acad Sci U S A
1994
, vol. 
91
 (pg. 
2834
-
2838
)
27
Pages
 
F
Ragueneau
 
M
Rottapel
 
R
, et al. 
Binding of phosphatidylinositol-3-OH kinase to CD28 is required for T-cell signalling.
Nature
1994
, vol. 
369
 (pg. 
327
-
329
)
28
Cai
 
YC
Cefai
 
D
Schneider
 
H
Raab
 
M
Nabavi
 
N
Rudd
 
CE
Selective CD28pYMNM mutations implicate phosphatidylinositol 3-kinase in CD86-CD28-mediated costimulation.
Immunity
1995
, vol. 
3
 (pg. 
417
-
426
)
29
Okkenhaug
 
K
Wu
 
L
Garza
 
KM
, et al. 
A point mutation in CD28 distinguishes proliferative signals from survival signals.
Nat Immunol
2001
, vol. 
2
 (pg. 
325
-
332
)
30
Stein
 
PH
Fraser
 
JD
Weiss
 
A
The cytoplasmic domain of CD28 is both necessary and sufficient for costimulation of interleukin-2 secretion and association with phosphatidylinositol 3′-kinase.
Mol Cell Biol
1994
, vol. 
14
 (pg. 
3392
-
3402
)
31
Ueda
 
Y
Levine
 
BL
Huang
 
ML
, et al. 
Both CD28 ligands CD80 (B7-1) and CD86 (B7-2) activate phosphatidylinositol 3-kinase, and wortmannin reveals heterogeneity in the regulation of T cell IL-2 secretion.
Int Immunol
1995
, vol. 
7
 (pg. 
957
-
966
)
32
Ward
 
SG
Ley
 
SC
MacPhee
 
C
Cantrell
 
DA
Regulation of D-3 phosphoinositides during T cell activation via the T cell antigen receptor/CD3 complex and CD2 antigens.
Eur J Immunol
1992
, vol. 
22
 (pg. 
45
-
49
)
33
Ward
 
SG
Westwick
 
J
Hall
 
ND
Sansom
 
DM
Ligation of CD28 receptor by B7 induces formation of D-3 phosphoinositides in T lymphocytes independently of T cell receptor/CD3 activation.
Eur J Immunol
1993
, vol. 
23
 (pg. 
2572
-
2577
)
34
Burr
 
JS
Savage
 
ND
Messah
 
GE
, et al. 
Cutting edge: distinct motifs within CD28 regulate T cell proliferation and induction of Bcl-XL.
J Immunol
2001
, vol. 
166
 (pg. 
5331
-
5335
)
35
Andres
 
PG
Howland
 
KC
Nirula
 
A
, et al. 
Distinct regions in the CD28 cytoplasmic domain are required for T helper type 2 differentiation.
Nat Immunol
2004
, vol. 
5
 (pg. 
435
-
442
)
36
Crooks
 
ME
Littman
 
DR
Carter
 
RH
Fearon
 
DT
Weiss
 
A
Stein
 
PH
CD28-mediated costimulation in the absence of phosphatidylinositol 3-kinase association and activation.
Mol Cell Biol
1995
, vol. 
15
 (pg. 
6820
-
6828
)
37
Okkenhaug
 
K
Bilancio
 
A
Farjot
 
G
, et al. 
Impaired B and T cell antigen receptor signaling in p110δ PI 3-kinase mutant mice.
Science
2002
, vol. 
297
 (pg. 
1031
-
1034
)
38
Deane
 
JA
Kharas
 
MG
Oak
 
JS
, et al. 
T-cell function is partially maintained in the absence of class IA phosphoinositide 3-kinase signaling.
Blood
2007
, vol. 
109
 (pg. 
2894
-
2902
)
39
Kane
 
LP
Andres
 
PG
Howland
 
KC
Abbas
 
AK
Weiss
 
A
Akt provides the CD28 costimulatory signal for up-regulation of IL-2 and IFN-γ but not TH2 cytokines.
Nat Immunol
2001
, vol. 
2
 (pg. 
37
-
44
)
40
Harada
 
Y
Tokushima
 
M
Matsumoto
 
Y
, et al. 
Critical requirement for the membrane-proximal cytosolic tyrosine residue for CD28-mediated costimulation in vivo.
J Immunol
2001
, vol. 
166
 (pg. 
3797
-
3803
)
41
Rudd
 
CE
Schneider
 
H
Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling.
Nat Rev Immunol
2003
, vol. 
3
 (pg. 
544
-
556
)
42
Okkenhaug
 
K
Patton
 
DT
Bilancio
 
A
Garcon
 
F
Rowan
 
WC
Vanhaesebroeck
 
B
The p110δ isoform of phosphoinositide 3-kinase controls clonal expansion and differentiation of Th cells.
J Immunol
2006
, vol. 
177
 (pg. 
5122
-
5128
)
43
Patton
 
DT
Garden
 
OA
Pearce
 
WP
, et al. 
Cutting edge: the phosphoinositide 3-kinase p110δ is critical for the function of CD4+CD25+Foxp3+ regulatory T cells.
J Immunol
2006
, vol. 
177
 (pg. 
6598
-
6602
)
44
Patton
 
DT
Garcon
 
F
Okkenhaug
 
K
The PI3K p110δ controls T-cell development, differentiation and regulation.
Biochem Soc Trans
2007
, vol. 
35
 (pg. 
167
-
171
)
45
Vanhaesebroeck
 
B
Higashi
 
K
Raven
 
C
, et al. 
Autophosphorylation of p110δ phosphoinositide 3-kinase: a new paradigm for the regulation of lipid kinases in vitro and in vivo.
EMBO J
1999
, vol. 
18
 (pg. 
1292
-
1302
)
46
Sasaki
 
T
Irie-Sasaki
 
J
Jones
 
RG
, et al. 
Function of PI3Kγ in thymocyte development, T cell activation, and neutrophil migration.
Science
2000
, vol. 
287
 (pg. 
1040
-
1046
)
47
Webb
 
LM
Vigorito
 
E
Wymann
 
MP
Hirsch
 
E
Turner
 
M
Cutting edge: T cell development requires the combined activities of the p110γ and p110δ catalytic isoforms of phosphatidylinositol 3-kinase.
J Immunol
2005
, vol. 
175
 (pg. 
2783
-
2787
)
48
Swat
 
W
Montgrain
 
V
Doggett
 
TA
, et al. 
Essential role of PI3Kdelta and PI3Kgamma in thymocyte survival.
Blood
2006
, vol. 
107
 (pg. 
2415
-
2422
)
49
Kumar
 
A
Humphreys
 
TD
Kremer
 
KN
, et al. 
CXCR4 physically associates with the T cell receptor to signal in T cells.
Immunity
2006
, vol. 
25
 (pg. 
213
-
224
)
50
Molon
 
B
Gri
 
G
Bettella
 
M
, et al. 
T cell costimulation by chemokine receptors.
Nat Immunol
2005
, vol. 
6
 (pg. 
465
-
471
)
51
Nishio
 
M
Watanabe
 
K
Sasaki
 
J
, et al. 
Control of cell polarity and motility by the PtdIns(3,4,5)P(3) phosphatase SHIP1.
Nat Cell Biol
2007
, vol. 
9
 (pg. 
36
-
44
)
52
Hirsch
 
E
Katanaev
 
VL
Garlanda
 
C
, et al. 
Central role for G protein-coupled phosphoinositide 3-kinase gamma in inflammation.
Science
2000
, vol. 
287
 (pg. 
1049
-
1053
)
53
Shahinian
 
A
Pfeffer
 
K
Lee
 
KP
, et al. 
Differential T cell costimulatory requirements in CD28-deficient mice.
Science
1993
, vol. 
261
 (pg. 
609
-
612
)
54
Barnden
 
MJ
Allison
 
J
Heath
 
WR
Carbone
 
FR
Defective TcR expression in transgenic mice constructed using cDNA-based α- and β-chain genes under the control of heterologous regulatory elements.
Immunol Cell Biol
1998
, vol. 
76
 (pg. 
34
-
40
)
55
Sadhu
 
C
Masinovsky
 
B
Dick
 
K
Sowell
 
CG
Staunton
 
DE
Essential role of phosphoinositide 3-kinase δ in neutrophil directional movement.
J Immunol
2003
, vol. 
170
 (pg. 
2647
-
2654
)
56
Sasaki
 
T
Sasaki
 
J
Watanabe
 
K
Suzuki
 
A
Non-invasive visualization of the lipid product of class I PI3K in transgenic mouse models.
Biochem Soc Trans
2007
, vol. 
35
 (pg. 
215
-
218
)
57
Monks
 
CR
Freiberg
 
BA
Kupfer
 
H
Sciaky
 
N
Kupfer
 
A
Three-dimensional segregation of supramolecular activation clusters in T cells.
Nature
1998
, vol. 
395
 (pg. 
82
-
86
)
58
Sanchez-Lockhart
 
M
Marin
 
E
Graf
 
B
, et al. 
Cutting edge: CD28-mediated transcriptional and posttranscriptional regulation of IL-2 expression are controlled through different signaling pathways.
J Immunol
2004
, vol. 
173
 (pg. 
7120
-
7124
)
59
Sun
 
Z
Arendt
 
CW
Ellmeier
 
W
, et al. 
PKC-theta is required for TcR-induced NF-kappaB activation in mature but not immature T lymphocytes.
Nature
2000
, vol. 
404
 (pg. 
402
-
407
)
60
Mirenda
 
V
Jarmin
 
SJ
David
 
R
, et al. 
Physiologic and aberrant regulation of memory T-cell trafficking by the costimulatory molecule CD28.
Blood
2007
, vol. 
109
 (pg. 
2968
-
2977
)
61
Kawai
 
K
Shahinian
 
A
Mak
 
TW
Ohashi
 
PS
Skin allograft rejection in CD28-deficient mice.
Transplantation
1996
, vol. 
61
 (pg. 
352
-
355
)
62
Ha
 
J
Bingaman
 
AW
Durham
 
MM
Pearson
 
TC
Larsen
 
CP
Aggressive skin allograft rejection in CD28−/− mice independent of the CD40/CD40L costimulatory pathway.
Transpl Immunol
2001
, vol. 
9
 (pg. 
13
-
17
)
63
Ji
 
H
Rintelen
 
F
Waltzinger
 
C
, et al. 
Inactivation of PI3Kγ and PI3Kδ distorts T-cell development and causes multiple organ inflammation.
Blood
2007
, vol. 
110
 (pg. 
2940
-
2947
)
64
Sasaki
 
T
Irie-Sasaki
 
J
Horie
 
Y
, et al. 
Colorectal carcinomas in mice lacking the catalytic subunit of PI(3)Kgamma.
Nature
2000
, vol. 
406
 (pg. 
897
-
902
)
65
Barbier
 
M
Attoub
 
S
Calvez
 
R
, et al. 
Tumour biology. Weakening link to colorectal cancer?
Nature
2001
, vol. 
413
 pg. 
796
 
66
Del Prete
 
A
Vermi
 
W
Dander
 
E
, et al. 
Defective dendritic cell migration and activation of adaptive immunity in PI3Kgamma-deficient mice.
EMBO J
2004
, vol. 
23
 (pg. 
3505
-
3515
)
67
Pages
 
F
Ragueneau
 
M
Klasen
 
S
, et al. 
Two distinct intracytoplasmic regions of the T-cell adhesion molecule CD28 participate in phosphatidylinositol 3-kinase association.
J Biol Chem
1996
, vol. 
271
 (pg. 
9403
-
9409
)
68
Marengere
 
LE
Okkenhaug
 
K
Clavreul
 
A
, et al. 
The SH3 domain of Itk/Emt binds to proline-rich sequences in the cytoplasmic domain of the T cell costimulatory receptor CD28.
J Immunol
1997
, vol. 
159
 (pg. 
3220
-
3229
)
69
Okkenhaug
 
K
Rottapel
 
R
Grb2 forms an inducible protein complex with CD28 through a Src homology 3 domain-proline interaction.
J Biol Chem
1998
, vol. 
273
 (pg. 
21194
-
21202
)
70
Kim
 
HH
Tharayil
 
M
Rudd
 
CE
Growth factor receptor-bound protein 2 SH2/SH3 domain binding to CD28 and its role in co-signaling.
J Biol Chem
1998
, vol. 
273
 (pg. 
296
-
301
)
71
Holdorf
 
AD
Green
 
JM
Levin
 
SD
, et al. 
Proline residues in CD28 and the Src homology (SH)3 domain of Lck are required for T cell costimulation.
J Exp Med
1999
, vol. 
190
 (pg. 
375
-
384
)
72
Ellis
 
JH
Ashman
 
C
Burden
 
MN
Kilpatrick
 
KE
Morse
 
MA
Hamblin
 
PA
GRID: a novel Grb-2-related adapter protein that interacts with the activated T cell costimulatory receptor CD28.
J Immunol
2000
, vol. 
164
 (pg. 
5805
-
5814
)
73
Holdorf
 
AD
Lee
 
KH
Burack
 
WR
Allen
 
PM
Shaw
 
AS
Regulation of Lck activity by CD4 and CD28 in the immunological synapse.
Nat Immunol
2002
, vol. 
3
 (pg. 
259
-
264
)
74
Friend
 
LD
Shah
 
DD
Deppong
 
C
, et al. 
A dose-dependent requirement for the proline motif of CD28 in cellular and humoral immunity revealed by a targeted knockin mutant.
J Exp Med
2006
, vol. 
203
 (pg. 
2121
-
2133
)
75
Andres
 
PG
Howland
 
KC
Dresnek
 
D
Edmondson
 
S
Abbas
 
AK
Krummel
 
MF
CD28 signals in the immature immunological synapse.
J Immunol
2004
, vol. 
172
 (pg. 
5880
-
5886
)
76
Acuto
 
O
Michel
 
F
CD28-mediated co-stimulation: a quantitative support for TcR signalling.
Nat Rev Immunol
2003
, vol. 
3
 (pg. 
939
-
951
)
77
Dennehy
 
KM
Elias
 
F
Na
 
SY
Fischer
 
KD
Hunig
 
T
Luhder
 
F
Mitogenic CD28 signals require the exchange factor Vav1 to enhance TcR signaling at the SLP-76-Vav-Itk signalosome.
J Immunol
2007
, vol. 
178
 (pg. 
1363
-
1371
)
78
Bluestone
 
JA
St Clair
 
EW
Turka
 
LA
CTLA4Ig: bridging the basic immunology with clinical application.
Immunity
2006
, vol. 
24
 (pg. 
233
-
238
)
79
Rommel
 
C
Camps
 
M
Ji
 
H
PI3K delta and PI3K gamma: partners in crime in inflammation in rheumatoid arthritis and beyond?
Nat Rev Immunol
2007
, vol. 
7
 (pg. 
191
-
201
)
80
Ruckle
 
T
Schwarz
 
MK
Rommel
 
C
PI3Kgamma inhibition: towards an ‘aspirin of the 21st century’?
Nat Rev Drug Discov
2006
, vol. 
5
 (pg. 
903
-
918
)
81
Elliott
 
JI
Raguz
 
S
Higgins
 
CF
Multidrug transporter activity in lymphocytes.
Br J Pharmacol
2004
, vol. 
143
 (pg. 
899
-
907
)
82
Miller
 
RA
Biomarkers of aging: prediction of longevity by using age-sensitive T-cell subset determinations in a middle-aged, genetically heterogeneous mouse population.
J Gerontol A Biol Sci Med Sci
2001
, vol. 
56
 (pg. 
B180
-
B186
)
Sign in via your Institution