The longevity of dendritic cells (DCs) is a critical regulatory factor influencing the outcome of immune responses. Recently, we demonstrated that the immunosuppressive drug rapamycin (Rapa) specifically induces apoptosis in DCs but not in other myeloid cell types. The present study unraveled the mechanism used by Rapa to induce apoptosis in human monocyte-derived DCs. Our data demonstrate that granulocyte-macrophage colony-stimulating factor (GM-CSF) preserves DC survival specifically via the phosphatidylinositol-3 lipid kinase/mammalian target of rapamycin (PI3K/mTOR) signaling pathway, which is abrogated by Rapa at the level of mTOR. Disruption of this GM-CSF signaling pathway induced loss of mitochondrial membrane potential, phosphatidyl-serine exposure, and nuclear changes. Apoptosis of these nonproliferating DCs was preceded by an up-regulation of the cell cycle inhibitor p27KIP1. Overexpression of p27KIP1 in DCs using adenoviral gene transduction revealed that apoptosis is directly regulated by p27KIP1. Furthermore, both overexpression of p27KIP1 and disruption of the GM-CSF/PI3K/mTOR signaling pathway decreased the expression of the antiapoptotic protein mcl-1. This mTOR/p27KIP1/mcl-1 survival seems unique for DCs and may provide novel opportunities to influence immune responses by specific interference with the life span of these cells.

Apoptosis, or programmed cell death, is a physiologic process that is required for the normal development and maintenance of tissue homeostasis.1 It is an active process that is regulated by gene products, which either block or accelerate programmed cell death. In most cells, the apoptotic program is always ready to be executed unless continuously inhibited by extracellular survival factors.2 Apoptosis regulates many aspects of immunologic homeostasis, including initiation, magnitude, and termination of immune responses. Dendritic cells (DCs) play a critical role in the diverse facets of immune regulation, ranging from tolerance induction and the prevention of autoimmunity to the induction of antitumor immunity and the protection against infectious agents.3,4 

DCs are the most potent antigen-presenting cells. They play a major role in the uptake, transport, and presentation of antigens and have the unique capacity to stimulate naive T lymphocytes.5 In addition to their polarizing capacity on contact with naive T cells,4 they can interact with B cells6 and natural killer (NK) cells7 and thus direct the character of the immune response. Although of possible biologic importance to down-regulate immune responses, apoptosis in DCs has been scarcely investigated. Several different death receptors have been identified on DCs, including Fas (CD95), tumor necrosis factor (TNF) receptor, and TNF receptor–related apoptosis-inducing ligand receptor (TRAIL-R), suggesting a role for death ligand–induced DC apoptosis.8-10 DC apoptosis can also be triggered by UVB radiation,11 glucocorticoids, reactive haptens, infectious pathogens, tumor cells, and NK cells.12 Although recently, specific nuclear factor κB (NF-κB) subunits were described to be important for DC survival in mice,13 the molecular mechanisms underlying DC apoptosis have not yet been unraveled. DCs can also actively protect themselves against cell death. Moreover, DCs protect themselves from cytotoxic T lymphocyte (CTL) attack,14 suggesting that the survival of DCs is an important regulatory mechanism in immune responses.

Previously, we demonstrated that rapamycin (Rapa) specifically induces apoptosis in both monocyte-derived DCs and DCs generated from CD34+ precursors but not in monocytes or macrophages.15 Rapa, which is an immunosuppressive drug introduced to prevent allograft rejection,16 is extensively studied for its effect on T lymphocytes and is known mainly for its antiproliferative effect.17 The drug is structurally related to FK506 that also inhibits T-cell proliferation. Although FK506 and Rapa bind to the same intracellular protein, FKBP-12, the resulting complexes interfere with distinct signaling pathways.18,19 FK506 inhibits production of interleukin 2 (IL-2), via inhibition of calcineurin, whereas Rapa inhibits growth factor signaling rather than growth factor synthesis.

The Rapa/FKBP12 complex acts to inhibit the activity of mammalian target of rapamycin (mTOR).17,20 mTOR is a member of the lipid kinase family with homology to phosphatidylinositol-3 lipid kinase (PI3K). In T cells, Rapa abrogates the IL-2–induced activity of mTOR, thereby blocking G1-S transition and proliferation. A potential mechanism by which PI3K and mTOR exert their proliferative effects is by down-regulation of the cyclin-dependent kinase (CDK) inhibitor p27KIP1.21 However, up-regulation of p27KIP1 is not only linked to cell cycle arrest in G0/1,22 but it seems also associated with apoptosis induced by growth factor withdrawal or PI3K inhibition.23,24 

In the present study we examined the mechanism used by Rapa to induce apoptosis in primary human DCs. Our data demonstrate that the survival of monocyte-derived DCs, which are nonproliferating cells, requires granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling via the PI3K/mTOR signaling pathway. Inhibition of mTOR via Rapa leads to increased expression of the cell cycle inhibitor p27KIP1 in nonproliferating DCs. Overexpression of p27KIP1 by using adenoviral gene transduction was shown to be directly responsible for the down-regulation of the antiapoptotic bcl-2 family protein mcl-1 and the induction of apoptosis.

Reagents

LY294002 (Alexis, San Diego, CA) and PD98059, SB203580, and Rapamycin (all from Calbiochem, Cambridge, MA) were dissolved in dimethyl sulfoxide (DMSO) and used at the concentrations indicated. FK506 (Prograft; Fujisawa Benelux, Houten, The Netherlands) was added to the cultures at 5 × 10−6 M.

Cell culture

Monocyte-derived DCs were generated as described previously.25 In brief, human monocytes were isolated from a buffy coat obtained from healthy donors using Ficoll-Hypaque (Sigma, St Louis, MO) and Percoll (Pharmacia, Uppsala, Sweden) density gradient centrifugation, followed by plastic adherence (2 hours) in T75 culture flasks (15 × 106 cells/flask; Nunc/Life Technologies, Breda, The Netherlands). Adherent monocytes were cultured in RPMI 1640 containing 10% heat-inactivated fetal calf serum (FCS) and penicillin/streptomycin (P/S) supplemented with 5 ng/mL GM-CSF (Sandoz, Uden, The Netherlands) and 10 ng/mL IL-4 (Sanvertech, Heerhugowaard, The Netherlands) for 6 days. Differentiation into CD1a+CD14CD83 immature DCs was confirmed by fluorescence-activated cell sorter (FACS) analysis.

Induction and detection of apoptosis

Apoptosis induction experiments were performed in 12- or 24-well culture plates (Costar, Cambridge, MA) containing 5 × 105 DC/mL.

Determination of nuclear condensation and fragmentation.

Cells were harvested at the indicated time points and fixed with 1% paraformaldehyde on ice. Cytospin preparations were made and stained with Hoechst 33258 (Molecular Probes, Leiden, The Netherlands) for 3 minutes at room temperature. The percentage of apoptotic cells was determined by examining 200 cells and counting the cells that were characterized by condensed or fragmented nuclei.

Determination of phosphatidyl-serine exposure.

Cells were harvested at the indicated time points, washed, labeled with annexin V–fluorescein isothiocyanate (FITC; Apoptest FITC kit; Nexins Research BV, Kattendijke, The Netherlands) for 30 minutes on ice, and subsequently taken up in 1 μg/mL propidium iodide (PI; Molecular Probes). Annexin V/PI staining was conducted on a FACScan and analyzed using WinMDI software (Becton Dickinson).

Determination of DNA fragmentation (sub-G0 analysis).

Cells were harvested, washed in 1 mM EDTA/PBS (ethylenediaminetetraacetic acid/phosphate-buffered saline), and fixed in 90% ethanol for at least 30 minutes at −20°C. Then the cells were washed, and cellular DNA was stained by treating the cells with 50 μg/mL RNase A (Sigma) and 10 μg/mL PI (Molecular Probes) for 45 minutes at room temperature. Fluorescence intensity was quantified on a per cell basis by flow cytometry (FACScan).

Determination of mitochondrial transmembrane potential.

Mitochondrial dysfunction was assessed by using rhodamine 123 (Rh123; Molecular Probes). Cells were incubated at 37°C for 30 minutes in the presence of 0.1 μg/mL Rh123. Then cells were washed and resuspended in PBS, either with or without 1 μg/mL PI, and Rh123 retention was conducted on a FACScan.

Preparation of whole cell lysates and Western blot analysis

Cultured DCs were harvested, washed, and lysed in Triton X-100 buffer, containing 20 mM Tris (tris(hydroxymethyl)aminomethane)–HCl (pH 7.4), 137 mM NaCl, 10% glycerol, 1% Triton X-100, 2 mM EDTA, 1 mM phenylmethyl sulfonyl fluoride (PMSF), 2 μg/mL leupeptin, 2 μg/mL antipain, 2 μg/mL chymostatin, and 5 U/mL trasylol. The protein concentration was determined by using the bicinchonic acid (BCA) protein assay (Pierce Chemical, Rockford, IL). Each protein sample was separated under reducing conditions on a 12% polyacrylamide sodium dodecyl sulfate (SDS) gel and semi-dry electroblotted on polyvinylidene fluoride (PVDF) membranes (Immobilon-p; Millipore, Bedford, MA). Membranes were incubated with 2% casein in PBS-0.05% Tween 20 for blocking, and then the primary antibody, either mouse-antihuman p27KIP1 (F-8; Santa Cruz Biotechnology, Santa Cruz, CA) or rabbit-antihuman mcl-1 (S-19; Santa Cruz Biotechnology), was added. After incubation with the appropriate secondary antibody (horseradish peroxidase [HRP]–conjugated swine-antirabbit immunoglobulin or goat-antimouse immunoglobulin both from Dako, Glostrup, Denmark) detection was performed with Supersignal (Pierce), and the blots were exposed to Hyperfilm films (Amersham Pharmacia Biotech, United Kingdom). Membranes were stripped by using Restore Stripping Buffer (Pierce) to investigate the expression of several specific proteins within one experiment. Equal protein loading was verified by Coomassie blue staining of the blot, which allows comparison over the whole molecular weight range.

Recombinant adenoviral vectors

Replication-deficient adenoviral vectors containing either human p27KIP1 cDNA (rAd-p27) or the LacZ gene (rAd-LacZ)24 were obtained from Drs I. Naruse and H. Hoshino (Gunma University School of Medicine, Maebashi, Japan). Adenoviral stocks were generated and purified by double CsCl density gradient centrifugation as described previously.26 To remove the CsCl, the virus bands were dialyzed against a Tris buffer (25 mM Tris-Cl, 137 mM NaCl, 5 mM KCl, 0.73 mM Na2HPO4, 0.9 mM CaCl, 0.5 mM MgCl2, pH 7.45). The final dialysis was performed with this Tris buffer containing 5% sucrose, and then virus stocks were stored at −80°C until further use.

Infection of DCs with rAd

Day 6 immature DCs (0.4 × 106) were resuspended in 100 μL PBS and incubated with rAd-LacZ (2.0 × 1010pfu/mL) or rAd-p27 (1.2 × 1010 pfu/mL) (multiplicity of infection [MOI], 1000).27 After 2 hours at 37°C, DCs were washed twice with PBS to remove free adenoviruses. Then cells were resuspended in RPMI 1640 containing 10% ΔFCS and P/S supplemented with 5 ng/mL GM-CSF and 10 ng/mL IL-4 and cultured at 37°C in a 5% CO2 incubator.

After 24 hours of culture, DCs were fixed with 0.2% glutaraldehyde/2% formaldehyde in PBS for 5 minutes at 4°C. Fixed cells were incubated in a stain solution containing 5 mM potassium ferrocyanide2+, 5 mM potassium ferrocyanide3+, 2 mM MgCl2, and 1 mg/mL X-gal (Sigma) for 4 hours at 37°C. The percentage of LacZ+ cells (blue cells) was calculated by light microscopy.

Monocytes and DCs differ in their survival mechanisms

As demonstrated previously, addition of GM-CSF to monocyte cultures increased the yield of functional immature DCs on culture with IL-4.28 To examine whether GM-CSF increases cell recovery by affecting cell survival at the start of culture, ie, an effect on freshly isolated monocytes, or by promoting DC survival, both monocytes and DCs were cultured with or without GM-CSF for 48 hours. GM-CSF withdrawal, but not IL-4 withdrawal, from DC cultures strongly induced apoptosis (Figure 1). GM-CSF withdrawal induced the typical characteristics of apoptosis, including nuclear condensation and fragmentation visualized by Hoechst staining (Figure1A) and DNA fragmentation as demonstrated by an increased sub-G0 fraction after DNA staining with propidium iodide (Figure 1B). The loss of mitochondrial transmembrane potential (ΔΨm) on GM-CSF deprivation, as determined by Rh123 retention, suggested an active role of mitochondria in the induction of apoptosis (Figure 1C).

Fig. 1.

GM-CSF withdrawal induces apoptosis in DCs.

DCs were cultured as described in “Materials and methods.” At day 6, DCs were harvested, extensively washed, and further cultured in RPMI-10% FCS supplemented with IL-4 (10 ng/mL) plus GM-CSF (5 ng/mL; designated as “GM-CSF” in panels B and C), IL-4 alone (10 ng/mL; designated as “No GM-CSF”), or GM-CSF alone (5 ng/mL; designated as “No IL-4”). After 48 hours of incubation, cells were analyzed for the amount of apoptosis using Hoechst to analyze nuclear morphology (A), PI to detect DNA fragmentation (B), or Rh123 to investigate ΔΨm (C). Photographs (original magnification, × 400) taken from cells stained with Hoechst were inverted and demonstrate condensed (“C”) and fragmented nuclei (“F”) indicated by arrows (A). Data presented are representative of 5 independent experiments with different donors.

Fig. 1.

GM-CSF withdrawal induces apoptosis in DCs.

DCs were cultured as described in “Materials and methods.” At day 6, DCs were harvested, extensively washed, and further cultured in RPMI-10% FCS supplemented with IL-4 (10 ng/mL) plus GM-CSF (5 ng/mL; designated as “GM-CSF” in panels B and C), IL-4 alone (10 ng/mL; designated as “No GM-CSF”), or GM-CSF alone (5 ng/mL; designated as “No IL-4”). After 48 hours of incubation, cells were analyzed for the amount of apoptosis using Hoechst to analyze nuclear morphology (A), PI to detect DNA fragmentation (B), or Rh123 to investigate ΔΨm (C). Photographs (original magnification, × 400) taken from cells stained with Hoechst were inverted and demonstrate condensed (“C”) and fragmented nuclei (“F”) indicated by arrows (A). Data presented are representative of 5 independent experiments with different donors.

Close modal

Both mitogen-activated protein kinase (MAPK) and PI3K/mTOR signaling pathways have been suggested to play a role in GM-CSF–induced survival of various hematopoietic cells. To investigate the involvement of the distinct signaling pathways in GM-CSF–mediated survival of DCs, the activation of the MAPK family members extracellular signal–related kinase (ERK) and p38, PI3K, and mTOR was blocked by culturing DCs in the presence of PD98059, SB203580, LY294002, and Rapa, respectively. PD98059 slightly induced apoptosis, whereas inhibition of p38 by SB203580 did not (Figure 2A). Both inhibition of PI3K using LY294002 and inhibition of mTOR using Rapa induced apoptosis comparable to GM-CSF deprivation, demonstrating a critical role for PI3K/mTOR signaling in the regulation of DC survival. Inhibition of PI3K, ERK, or p38 in monocytes slightly induced apoptosis (Figure 2A). As shown previously,15 monocytes were completely insensitive for the proapoptotic activity of Rapa, demonstrating that the longevity of DCs and their precursors, ie, the monocytes, are regulated by different survival mechanisms.

Fig. 2.

Monocytes and DCs differ in their survival mechanisms.

(A) DCs were cultured in RPMI-10% FCS with the addition of IL-4 and GM-CSF, whereas freshly isolated peripheral blood monocytes were cultured in RPMI-10% FCS alone. After 48 hours of culture, the effects of GM-CSF deprivation (No GM-CSF) or treatment with PD98059 (50 μM), SB203580 (10 μM), LY294002 (20 μM), Rapa (1 μM), or DMSO, were analyzed using Hoechst. Results demonstrate mean ± SD percentage of apoptosis of 3 independent experiments with different donors. * indicates significant apoptotic effect of “No GM-CSF” compared with “GM-CSF” or an inhibitor compared with its solvent “DMSO” (P < .05; Student t test for paired samples). (B) DCs were cultured with IL-4 and GM-CSF in the presence of various concentrations of the PI3K-inhibitor LY294002, or DMSO. Cells were analyzed after 24 hours or 48 hours using Hoechst (n = 2). (C) Day-6 DCs were harvested, extensively washed, and further cultured in RPMI-10% FCS supplemented with IL-4 plus GM-CSF (control), IL-4 alone (No GM-CSF), or IL-4 plus GM-CSF with either Rapa (1 μM), LY294002 (20 μM), PD98059 (50 μM), or SB203580 (10 μM) added. After 48 hours, ΔΨm was investigated using Rh123. The percentage of cells with a decreased ΔΨm are plotted in a histogram. Data are shown as the mean ± SD percentage of apoptosis of duplicate cultures, representative for 3 to 6 independent experiments with different donors. * indicates significant apoptotic effect compared with “GM-CSF” or “DMSO” (P < .05; Student t test for paired samples).

Fig. 2.

Monocytes and DCs differ in their survival mechanisms.

(A) DCs were cultured in RPMI-10% FCS with the addition of IL-4 and GM-CSF, whereas freshly isolated peripheral blood monocytes were cultured in RPMI-10% FCS alone. After 48 hours of culture, the effects of GM-CSF deprivation (No GM-CSF) or treatment with PD98059 (50 μM), SB203580 (10 μM), LY294002 (20 μM), Rapa (1 μM), or DMSO, were analyzed using Hoechst. Results demonstrate mean ± SD percentage of apoptosis of 3 independent experiments with different donors. * indicates significant apoptotic effect of “No GM-CSF” compared with “GM-CSF” or an inhibitor compared with its solvent “DMSO” (P < .05; Student t test for paired samples). (B) DCs were cultured with IL-4 and GM-CSF in the presence of various concentrations of the PI3K-inhibitor LY294002, or DMSO. Cells were analyzed after 24 hours or 48 hours using Hoechst (n = 2). (C) Day-6 DCs were harvested, extensively washed, and further cultured in RPMI-10% FCS supplemented with IL-4 plus GM-CSF (control), IL-4 alone (No GM-CSF), or IL-4 plus GM-CSF with either Rapa (1 μM), LY294002 (20 μM), PD98059 (50 μM), or SB203580 (10 μM) added. After 48 hours, ΔΨm was investigated using Rh123. The percentage of cells with a decreased ΔΨm are plotted in a histogram. Data are shown as the mean ± SD percentage of apoptosis of duplicate cultures, representative for 3 to 6 independent experiments with different donors. * indicates significant apoptotic effect compared with “GM-CSF” or “DMSO” (P < .05; Student t test for paired samples).

Close modal

DC survival requires GM-CSF signaling via PI3K and mTOR

Like treatment of DCs with Rapa to inhibit mTOR,15blockade of PI3K in DCs by addition of LY294002 time- and dose-dependently induced apoptosis that was characterized by nuclear condensation and fragmentation (Figure 2B) and DNA fragmentation (data not shown). In addition to the data obtained from GM-CSF deprivation experiments, both LY294002 and Rapa demonstrated loss of ΔΨm associated with the percentage of apoptosis induced (Figure 2C). In line with the induction of apoptosis, inhibition of ERK by PD98059 only slightly induced loss of ΔΨm, and inhibition of p38 did not significantly change ΔΨm(Figure 2C).

Rapa increases the expression of p27KIP1

Because mTOR is the most downstream target in GM-CSF–driven survival of DCs as determined so far, and because Rapa is clinically used as an immunosuppressive drug, we further explored the mechanism used by Rapa to specifically induce apoptosis in DCs. In T lymphocytes, Rapa exerts its effects via the up-regulation of the cell cycle inhibitor p27KIP1. An increased expression of p27KIP1 in nonproliferating eosinophils has been shown to correlate with the induction of apoptosis.23 Therefore, we investigated whether Rapa or GM-CSF signaling could influence p27KIP1 protein expression in monocyte-derived DCs, which are also nonproliferating cells. A 48-hour incubation in the presence of Rapa strongly increased the expression of p27KIP1(Figure 3A). When DCs were simultaneously treated with FK506, which antagonizes the apoptotic effect of Rapa (Figure 3B), the Rapa-induced p27KIP1 expression also could be inhibited (Figure 3A). Although Rapa required more than 24 hours to induce strong apoptosis,15 the effect on the expression of p27KIP1 was observed relatively early. Treatment with Rapa for 5 hours already increased the expression of p27KIP1that was not further increased by an extended incubation period in the presence of the drug (Figure 3C-D).

Fig. 3.

Rapa increases expression of p27KIP1 protein.

(A-B) DCs were cultured with IL-4 and GM-CSF in the absence or presence of Rapa (10−7 M) and either with or without addition of FK506 (5 × 10−6 M) for 48 hours. Part of the cells was used to prepare whole cell lysates. Equal amounts of protein (20 μg/lane) were loaded, and the levels of p27KIP1 were determined (A). Apoptosis was detected in remaining cells by flow cytometry, using annexin V–FITC and PI staining (B). Data shown are representative for 3 independent experiments performed with different donors. (C-D) DCs were cultured in IL-4 and GM-CSF with or without addition of 1 μM Rapa. Cells were harvested after 5 hours, 24 hours, or 48 hours of incubation. Whole cell lysates (20 μg /lane) were loaded, and the levels of p27KIP1 were determined by Western blot analysis (C). The relative expression of p27KIP1 was determined by using Stratagene-EagleSight (La Jolla, CA) and represents the ratio of each band from Rapa-treated DCs to that of untreated DCs. Results are expressed as mean ratio (± SD) of 4 independent experiments (D).

Fig. 3.

Rapa increases expression of p27KIP1 protein.

(A-B) DCs were cultured with IL-4 and GM-CSF in the absence or presence of Rapa (10−7 M) and either with or without addition of FK506 (5 × 10−6 M) for 48 hours. Part of the cells was used to prepare whole cell lysates. Equal amounts of protein (20 μg/lane) were loaded, and the levels of p27KIP1 were determined (A). Apoptosis was detected in remaining cells by flow cytometry, using annexin V–FITC and PI staining (B). Data shown are representative for 3 independent experiments performed with different donors. (C-D) DCs were cultured in IL-4 and GM-CSF with or without addition of 1 μM Rapa. Cells were harvested after 5 hours, 24 hours, or 48 hours of incubation. Whole cell lysates (20 μg /lane) were loaded, and the levels of p27KIP1 were determined by Western blot analysis (C). The relative expression of p27KIP1 was determined by using Stratagene-EagleSight (La Jolla, CA) and represents the ratio of each band from Rapa-treated DCs to that of untreated DCs. Results are expressed as mean ratio (± SD) of 4 independent experiments (D).

Close modal

p27KIP1 expression is increased after GM-CSF withdrawal or PI3K inhibition

To support the hypotheses that Rapa-induced apoptosis is mediated by an increased p27KIP1 expression, we analyzed the effect of cytokine withdrawal and PI3K inhibition on the expression of p27KIP1. All culture conditions that lead to apoptosis of DCs, including cultures without GM-CSF or with the PI3K inhibitor LY294002, were accompanied by an increased p27KIP1expression, whereas no changes were observed in conditions without apoptosis, such as IL-4 withdrawal or DMSO (Figure4A).

Fig. 4.

p27KIP1 protein levels correlate with the induction of apoptosis.

(A) Day-6 DCs were harvested, extensively washed, and further cultured in RPMI-10% FCS in the presence or absence of IL-4 and GM-CSF (left panel) or in the presence of both IL-4 and GM-CSF with or without LY294002 (20 μM) or DMSO (right panel). Data demonstrated in left and right panels are derived from different experiments with different donors. (B) Freshly isolated peripheral blood monocytes were cultured in RPMI-10% FCS with or without addition of Rapa (1 μM).

Fig. 4.

p27KIP1 protein levels correlate with the induction of apoptosis.

(A) Day-6 DCs were harvested, extensively washed, and further cultured in RPMI-10% FCS in the presence or absence of IL-4 and GM-CSF (left panel) or in the presence of both IL-4 and GM-CSF with or without LY294002 (20 μM) or DMSO (right panel). Data demonstrated in left and right panels are derived from different experiments with different donors. (B) Freshly isolated peripheral blood monocytes were cultured in RPMI-10% FCS with or without addition of Rapa (1 μM).

Close modal

Monocytes were completely resistant to Rapa-induced apoptosis (Figure2A). Rapa did not increase p27KIP1 in cultured peripheral blood monocytes (Figure 4B), confirming the hypothesis that Rapa-induced apoptosis is preceded and accompanied by an increased expression of p27KIP1.

p27KIP1 gene transduction induces apoptosis in DCs

To determine whether up-regulation of p27KIP1 has a causative role in Rapa-induced apoptosis of monocyte-derived DCs, DCs were infected with recombinant adenoviruses (rAd) containing human p27KIP1 cDNA. Infection of DCs with rAd-LacZ served as a control and showed that transduction efficiencies of above 90% were achieved at an MOI of 1000 (Figure 5A). Expression of p27KIP1 protein after infection with the adenoviral vectors was measured by Western blot analysis. A strong overexpression of p27KIP1 was detected at 48 hours after rAd-p27 infection (Figure 5B), which was not further increased after 72 hours. Expression of p27KIP1 protein in DCs infected with rAd-LacZ was not different from control levels.

Fig. 5.

Overexpression of p27KIP1 induces apoptosis in DCs.

(A) Evaluation transduction efficiency of rAd-LacZ or rAd-p27–infected immature DCs using X-gal. (B) DCs were infected with rAd-LacZ or rAd-p27. After 48 hours of culture in the presence of IL-4 and GM-CSF, infected DCs and control DCs were harvested for the preparation of whole cell lysates. Then equal amounts of protein (20 μg/lane) were loaded, and the levels of p27KIP1 were determined. Data shown are representative for 3 independent experiments with different donors. (C) DCs were infected with rAd-LacZ or rAd-p27 and subsequently cultured in IL-4 and GM-CSF. After 48 hours of culture, annexin V–FITC/PI staining was performed to determine the percentage of apoptotic cells. Data shown are representative for 2 independent experiments with different donors. (D) DCs were left untreated or infected with rAd-LacZ or rAd-p27 and subsequently cultured in IL-4 and GM-CSF. Cells were harvested after 24, 48, and 72 hours of culture. Annexin V–FITC/PI stainings were performed to determine the percentage of annexin V+/PI (early apoptotic) cells (n = 2).

Fig. 5.

Overexpression of p27KIP1 induces apoptosis in DCs.

(A) Evaluation transduction efficiency of rAd-LacZ or rAd-p27–infected immature DCs using X-gal. (B) DCs were infected with rAd-LacZ or rAd-p27. After 48 hours of culture in the presence of IL-4 and GM-CSF, infected DCs and control DCs were harvested for the preparation of whole cell lysates. Then equal amounts of protein (20 μg/lane) were loaded, and the levels of p27KIP1 were determined. Data shown are representative for 3 independent experiments with different donors. (C) DCs were infected with rAd-LacZ or rAd-p27 and subsequently cultured in IL-4 and GM-CSF. After 48 hours of culture, annexin V–FITC/PI staining was performed to determine the percentage of apoptotic cells. Data shown are representative for 2 independent experiments with different donors. (D) DCs were left untreated or infected with rAd-LacZ or rAd-p27 and subsequently cultured in IL-4 and GM-CSF. Cells were harvested after 24, 48, and 72 hours of culture. Annexin V–FITC/PI stainings were performed to determine the percentage of annexin V+/PI (early apoptotic) cells (n = 2).

Close modal

Cells were harvested after 48 hours of incubation. Whole cell lysates were prepared, equal amounts of protein (20 μg/lane) were loaded, and the levels of p27KIP1 were determined. Data shown are representative for 3 independent experiments with different donors.

In the presence of GM-CSF, DCs infected with rAd-p27 went into apoptosis as demonstrated by an increased annexin V binding within the PI population (Figure 5C) and nuclear condensation and fragmentation (data not shown). Kinetic experiments showed that p27KIP1-induced apoptosis of DCs was already present 24 hours after infection, which further increased in time (Figure 5D). No differences were found in annexin V binding and nuclear morphology between rAd-LacZ–infected DCs and control DC cultures.

Increased expression of p27KIP1 is responsible for down-regulation of mcl-1

As demonstrated earlier, apoptosis of DCs induced as a consequence of a disruptive GM-CSF/PI3K/mTOR signaling pathway is associated with loss of mitochondrial integrity. The bcl-2 family of proteins, containing both antiapoptotic and proapoptotic members, controls mitochondrial permeability and thus plays a critical role in the regulation of apoptosis. mcl-1 is an antiapoptotic protein, which is thought to be an important bcl-2 family member in GM-CSF–mediated survival of hematopoietic cells.29 

Western blot analysis of lysates prepared after rAd-p27 infection demonstrated that increased expression of p27KIP1 directly causes down-regulation of mcl-1 protein expression in DCs (Figure6A). The correlation between the down-regulation of mcl-1 and the interference in the GM-CSF signaling pathway at the level of PI3K and mTOR finally leading to apoptosis was further examined. Addition of either LY294002 or Rapa to DC cultures, as well as deprivation of GM-CSF, induced a down-regulation of mcl-1 that correlated with the percentage of apoptosis induced, thereby demonstrating that expression of mcl-1 is tightly regulated via PI3K and mTOR in response to GM-CSF (Figure 6B).

Fig. 6.

Overexpression of p27KIP1 as well as disruptive GM-CSF/PI3K/mTOR signaling down-regulate mcl-1 protein expression.

(A) DCs were left untreated or infected with rAd-LacZ or rAd-p27. After 48 hours of culture in the presence of IL-4 and GM-CSF, DCs were harvested for the preparation of whole cell lysates. Then equal amounts of protein (25 μg/lane) were loaded, and the levels of mcl-1 were determined. Data shown are representative for 2 independent experiments with different donors. (B) DCs were cultured either in IL-4 (No GM-CSF) alone or IL-4 and GM-CSF. Cultures containing IL-4 and GM-CSF were supplemented with or without 10 μM LY294002, 1 μM Rapa, or DMSO. Equal amounts of protein (20 μg whole cell lysate/lane) were loaded, and the levels of mcl-1 were determined. Data shown are representative for 3 independent experiments performed with different donors. (C) Quantification of mcl-1 expression in control and Rapa-treated DCs after either 24 hours or 48 hours of culture as described in “Materials and methods.” Data shown are representative for 4 independent experiments performed with different donors.

Fig. 6.

Overexpression of p27KIP1 as well as disruptive GM-CSF/PI3K/mTOR signaling down-regulate mcl-1 protein expression.

(A) DCs were left untreated or infected with rAd-LacZ or rAd-p27. After 48 hours of culture in the presence of IL-4 and GM-CSF, DCs were harvested for the preparation of whole cell lysates. Then equal amounts of protein (25 μg/lane) were loaded, and the levels of mcl-1 were determined. Data shown are representative for 2 independent experiments with different donors. (B) DCs were cultured either in IL-4 (No GM-CSF) alone or IL-4 and GM-CSF. Cultures containing IL-4 and GM-CSF were supplemented with or without 10 μM LY294002, 1 μM Rapa, or DMSO. Equal amounts of protein (20 μg whole cell lysate/lane) were loaded, and the levels of mcl-1 were determined. Data shown are representative for 3 independent experiments performed with different donors. (C) Quantification of mcl-1 expression in control and Rapa-treated DCs after either 24 hours or 48 hours of culture as described in “Materials and methods.” Data shown are representative for 4 independent experiments performed with different donors.

Close modal

A more detailed analysis of the effect of Rapa on the expression of mcl-1 in DCs was performed. The Rapa-induced reduction of mcl-1 protein levels was already observed within 24 hours of incubation, as demonstrated by Western blot analysis, but became more pronounced after 48 hours of treatment, finally resulting in a 2-fold reduction in mcl-1 protein levels (Figure 6C).

In the present study we show that defective GM-CSF signaling in human nonproliferating DCs, either because of selective inhibition of PI3K or mTOR, or GM-CSF deprivation, caused an increased expression of the cyclin-dependent kinase (CDK) inhibitor p27KIP1 prior to a decreased expression of the antiapoptotic protein mcl-1 and apoptosis. Overexpression of p27KIP1 in DCs by using adenoviral gene transduction revealed that both mcl-1 expression and cell survival are directly regulated by p27KIP1.

By examining the mechanism used by Rapa for the induction of apoptosis in DCs, we found that the PI3K/mTOR pathway is a critical signaling route for GM-CSF–driven survival of monocyte-derived DCs. GM-CSF is a potent growth factor for a variety of hematopoietic cells. On binding to its heterodimeric receptor, GM-CSF activates several signaling pathways, including the Jak-Stat (Janus kinase and signal transducer and activator of transcription) and Ras pathways,30,31 resulting in both mitogenic and antiapoptotic signals. Although downstream from Ras, both Raf/MAPK and PI3K/mTOR pathways have been suggested to play a role in cytokine-driven survival of hematopoietic cells,31,32 we found that for monocyte-derived DCs only the PI3K/mTOR pathway is absolutely necessary. In line with a previous study on lipopolysaccharide (LPS)–stimulated monocyte-derived DCs,33 which also demonstrate a cell death–inducing effect of LY294002 but not of PD98059 or SB203580, we observed only a limited role for ERK activation in DC survival. Although our data suggest that Rapa specifically counteracts the antiapoptotic activity of GM-CSF that results in an increased expression of p27KIP1, the fact that Rapa further increases apoptosis on GM-CSF withdrawal (data not shown) might suggest additional modes of action of Rapa.

p27KIP1 is an inhibitor of cell cycle progression, exerting its effect through interaction with cyclin-CDK complexes and arresting cells in G0/1.22 p27KIP1 is present at relatively high levels in quiescent cells and is down-regulated by mitogenic stimulation21,34,35; the latter process being blocked by Rapa.21 It is important to note that a cell cycle inhibitor can also play a decisive role in a nonproliferating cell type such as DCs or, as shown, nonproliferating eosinophils.23 However, although both in eosinophils and in the murine IL-3–dependent cell line Ba/F3 apoptosis was linked to increased p27KIP1 and interference with the PI3K pathway, apoptosis could not be induced by Rapa.23 This finding further underlines the specificity of Rapa toward DC apoptosis. It has been shown that p27KIP1-deficient mice demonstrate an increased survival of bone marrow–derived stem cells when cultured ex vivo compared with wild-type mice,23 but the immune status has not been specifically investigated.36 The mechanism by which p27KIP1 can regulate cell survival is not known. In search of a potential mechanism by which p27KIP1 could modulate GM-CSF–driven DC survival, we focused on the regulation of mcl-1.

mcl-1, initially cloned as a gene differentially expressed in human ML-1 myeloid leukemia cells,37 shows extensive homology to bcl-2. Like bcl-2 and bcl-xL, mcl-1 heterodimerizes with bax38 and thus plays an important role in the prevention of apoptosis. Although mcl-1 and bcl-2 show strong homology, their distribution, expression levels, and regulation of apoptosis are independently regulated.39-41 mcl-1 is thought to be an important protein in IL-3 and GM-CSF–mediated survival of hematopoietic cells,29,42 a finding supported by the fact that mcl-1 transgenic mice possess an enhanced viability in a wide range of hematopoietic cell types.43 

We demonstrate that the protein level of mcl-1 in DCs is tightly regulated by GM-CSF signaling via PI3K and mTOR. Despite the large decrease in the level of mcl-1 protein following treatment with Rapa or LY294002, no significant decrease in the level of mcl-1 mRNA was observed (data not shown). This finding is consistent with previous work demonstrating that GM-CSF–mediated mcl-1 expression is regulated at the translational level by a PI3K-controlled pathway.44LY294002-treated human monocyte-derived macrophages45showed also a marked decrease of mcl-1 protein expression, but they appear to have different PI3K/mcl-1 survival mechanisms than monocyte-derived DCs, because monocytes and macrophages do not undergo Rapa-induced apoptosis (Figure 2).15 In addition, we showed that monocytes do not increase their p27KIP1expression on Rapa treatment, whereas DCs clearly do. Therefore, alternative survival mechanisms might be present in monocytes/macrophages that regulate the expression of mcl-1 independently of mTOR and p27KIP1.

In conclusion, given their central role in the immune system, DCs are important targets for both immunosuppressive or immunostimulatory therapies.46-50 Understanding the survival program of DCs will provide the opportunity to either increase immune responses by prolonged DC survival or to terminate these responses by specific depletion of the cells. Rapa, which has been introduced recently as an effective drug to prevent allograft rejection, might partially exert its immunosuppressive effect by virtue of its proapoptotic effect on DCs. Our finding that the survival of monocyte-derived DCs is specifically regulated by the GM-CSF signaling pathway via PI3K/mTOR and involves the regulation of p27KIP1 and mcl-1 might provide additional tools to control immune responses.

We thank Drs I. Naruse and H. Hoshino (Gunma University School of Medicine, Maebashi, Japan) for providing the adenoviral vectors containing either p27KIP1 or LacZ, and M. J. W. E. Rabelink (Department of Molecular Cell Biology), Dr L. T. C. Peltenburg (Department of Clinical Oncology), and N. Schlagwein (Department of Nephrology; all from LUMC, Leiden, The Netherlands) for technical assistance.

C. van K. is a fellow of the Royal Netherlands Academy of Arts and Sciences (KNAW).

Prepublished online as Blood First Edition Paper, September 26, 2002; DOI 10.1182/blood-2002-06-1688.

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 U.S.C. section 1734.

1
Vaux
DL
Korsmeyer
SJ
Cell death in development.
Cell.
96
1999
245
254
2
Raff
MC
Social controls on cell survival and cell death.
Nature.
356
1992
397
400
3
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature.
392
1998
245
252
4
Liu
YJ
Kanzler
H
Soumelis
V
Gilliet
M
Dendritic cell lineage, plasticity and cross-regulation.
Nat Immunol.
2
2001
585
589
5
Banchereau
J
Briere
F
Caux
C
et al
Immunobiology of dendritic cells.
Annu Rev Immunol.
18
2000
767
811
6
Dubois
B
Bridon
JM
Fayette
J
et al
Dendritic cells directly modulate B cell growth and differentiation.
J Leukoc Biol.
66
1999
224
230
7
Gerosa
F
Baldani-Guerra
B
Nisii
C
Marchesini
V
Carra
G
Trinchieri
G
Reciprocal activating interaction between natural killer cells and dendritic cells.
J Exp Med.
195
2002
327
333
8
Matsue
H
Edelbaum
D
Hartmann
AC
et al
Dendritic cells undergo rapid apoptosis in vitro during antigen-specific interaction with CD4+ T cells.
J Immunol.
162
1999
5287
5298
9
Koppi
TA
Tough-Bement
T
Lewinsohn
DM
Lynch
DH
Alderson
MR
CD40 ligand inhibits Fas/CD95-mediated apoptosis of human blood-derived dendritic cells.
Eur J Immunol.
27
1997
3161
3165
10
Bjorck
P
Banchereau
J
Flores-Romo
L
CD40 ligation counteracts Fas-induced apoptosis of human dendritic cells.
Int Immunol.
9
1997
365
372
11
Nicolo
C
Tomassini
B
Rippo
MR
Testi
R
UVB-induced apoptosis of human dendritic cells: contribution by caspase-dependent and caspase-independent pathways.
Blood.
97
2001
1803
1808
12
Matsue
H
Takashima
A
Apoptosis in dendritic cell biology.
J Dermatol Sci.
20
1999
159
171
13
Ouaaz
F
Arron
J
Zheng
Y
Choi
Y
Beg
AA
Dendritic cell development and survival require distinct NF-kappaB subunits.
Immunity.
16
2002
257
270
14
Medema
JP
Schuurhuis
DH
Rea
D
et al
Expression of the serpin serine protease inhibitor 6 protects dendritic cells from cytotoxic T lymphocyte-induced apoptosis: differential modulation by T helper type 1 and type 2 cells.
J Exp Med.
194
2001
657
667
15
Woltman
AM
De Fijter
JW
Kamerling
SW
et al
Rapamycin induces apoptosis in monocyte- and CD34-derived dendritic cells, but not in monocytes and macrophages.
Blood.
98
2001
174
180
16
Kahan
BD
Efficacy of sirolimus compared with azathioprine for reduction of acute renal allograft rejection: a randomised multicentre study.
N Engl J Med.
356
2000
194
202
17
Abraham
RT
Wiederrecht
GJ
Immunopharmacology of rapamycin.
Annu Rev Immunol.
14
1996
483
510
18
Bierer
BE
Mattila
PS
Standaert
RF
et al
Two distinct signal transmission pathways in T lymphocytes are inhibited by complexes formed between an immunophilin and either FK506 or rapamycin.
Proc Natl Acad Sci U S A.
87
1990
9231
9235
19
Dumont
FJ
Melino
MR
Staruch
MJ
Koprak
SL
Fischer
PA
Sigal
NH
The immunosuppressive macrolides FK-506 and rapamycin act as reciprocal antagonists in murine T cells.
J Immunol.
144
1990
1418
1424
20
Sabers
CJ
Martin
MM
Brunn
GJ
et al
Isolation of a protein target of the FKBP12-rapamycin complex in mammalian cells.
J Biol Chem.
270
1995
815
822
21
Nourse
J
Firpo
E
Flanagan
WM
et al
Interleukin-2-mediated elimination of the p27Kip1 cyclin-dependent kinase inhibitor prevented by rapamycin.
Nature.
372
1994
570
573
22
Toyoshima
H
Hunter
T
p27, a novel inhibitor of G1 cyclin-Cdk protein kinase activity, is related to p21.
Cell.
78
1994
67
74
23
Dijkers
PF
Medema
RH
Pals
C
et al
Forkhead transcription factor FKHR-L1 modulates cytokine-dependent transcriptional regulation of p27(KIP1).
Mol Cell Biol.
20
2000
9138
9148
24
Naruse
I
Hoshino
H
Dobashi
K
Minato
K
Saito
R
Mori
M
Over-expression of p27kip1 induces growth arrest and apoptosis mediated by changes of pRb expression in lung cancer cell lines.
Int J Cancer.
88
2000
377
383
25
Woltman
AM
Fijter
JW
Kamerling
SWA
Paul
LC
Daha
MR
Van Kooten
C
The effect of calcineurin inhibitors and corticosteroids on the differentiation of human dendritic cells.
Eur J Immunol.
30
2000
1807
1812
26
Fallaux
FJ
Kranenburg
O
Cramer
SJ
et al
Characterization of 911: a new helper cell line for the titration and propagation of early region 1-deleted adenoviral vectors.
Hum Gene Ther.
7
1996
215
222
27
Rea
D
Schagen
FH
Hoeben
RC
et al
Adenoviruses activate human dendritic cells without polarization toward a T-helper type 1-inducing subset.
J Virol.
73
1999
10245
10253
28
Sallusto
F
Lanzavecchia
A
Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha.
J Exp Med.
179
1994
1109
1118
29
Chao
JR
Wang
JM
Lee
SF
et al
mcl-1 is an immediate-early gene activated by the granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling pathway and is one component of the GM-CSF viability response.
Mol Cell Biol.
18
1998
4883
4898
30
Al Shami
A
Mahanna
W
Naccache
PH
Granulocyte-macrophage colony-stimulating factor-activated signaling pathways in human neutrophils: selective activation of Jak2, Stat3, and Stat5b.
J Biol Chem.
273
1998
1058
1063
31
Sato
N
Sakamaki
K
Terada
N
Arai
K
Miyajima
A
Signal transduction by the high-affinity GM-CSF receptor: two distinct cytoplasmic regions of the common beta subunit responsible for different signaling.
EMBO J.
12
1993
4181
4189
32
Kinoshita
T
Yokota
T
Arai
K
Miyajima
A
Suppression of apoptotic death in hematopoietic cells by signalling through the IL-3/GM-CSF receptors.
EMBO J.
14
1995
266
275
33
Ardeshna
KM
Pizzey
AR
Devereux
S
Khwaja
A
The PI3 kinase, p38 SAP kinase, and NF-kappaB signal transduction pathways are involved in the survival and maturation of lipopolysaccharide-stimulated human monocyte-derived dendritic cells.
Blood.
96
2000
1039
1046
34
Reynisdottir
I
Polyak
K
Iavarone
A
Massague
J
Kip/Cip and Ink4 Cdk inhibitors cooperate to induce cell cycle arrest in response to TGF-beta.
Genes Dev.
9
1995
1831
1845
35
Kato
JY
Matsuoka
M
Polyak
K
Massague
J
Sherr
CJ
Cyclic AMP-induced G1 phase arrest mediated by an inhibitor (p27Kip1) of cyclin-dependent kinase 4 activation.
Cell.
79
1994
487
496
36
Kiyokawa
H
Kineman
RD
Manova-Todorova
KO
et al
Enhanced growth of mice lacking the cyclin-dependent kinase inhibitor function of p27(Kip1).
Cell.
85
1996
721
732
37
Kozopas
KM
Yang
T
Buchan
HL
Zhou
P
Craig
RW
MCL1, a gene expressed in programmed myeloid cell differentiation, has sequence similarity to BCL2.
Proc Natl Acad Sci U S A.
90
1993
3516
3520
38
Sedlak
TW
Oltvai
ZN
Yang
E
et al
Multiple Bcl-2 family members demonstrate selective dimerizations with Bax.
Proc Natl Acad Sci U S A.
92
1995
7834
7838
39
Ohta
K
Iwai
K
Kasahara
Y
et al
Immunoblot analysis of cellular expression of Bcl-2 family proteins, Bcl-2, Bax, Bcl-X and Mcl-1, in human peripheral blood and lymphoid tissues.
Int Immunol.
7
1995
1817
1825
40
Krajewski
S
Bodrug
S
Krajewska
M
et al
Immunohistochemical analysis of Mcl-1 protein in human tissues: differential regulation of Mcl-1 and Bcl-2 protein production suggests a unique role for Mcl-1 in control of programmed cell death in vivo.
Am J Pathol.
146
1995
1309
1319
41
Yang
T
Kozopas
KM
Craig
RW
The intracellular distribution and pattern of expression of Mcl-1 overlap with, but are not identical to, those of Bcl-2.
J Cell Biol.
128
1995
1173
1184
42
Wang
JM
Chao
JR
Chen
W
Kuo
ML
Yen
JJ
Yang-Yen
HF
The antiapoptotic gene mcl-1 is up-regulated by the phosphatidylinositol 3-kinase/Akt signaling pathway through a transcription factor complex containing CREB.
Mol Cell Biol.
19
1999
6195
6206
43
Zhou
P
Qian
L
Bieszczad
CK
et al
Mcl-1 in transgenic mice promotes survival in a spectrum of hematopoietic cell types and immortalization in the myeloid lineage.
Blood.
92
1998
3226
3239
44
Schubert
KM
Duronio
V
Distinct roles for extracellular-signal-regulated protein kinase (ERK) mitogen-activated protein kinases and phosphatidylinositol 3-kinase in the regulation of Mcl-1 synthesis.
Biochem J.
356
2001
473
480
45
Liu
H
Perlman
H
Pagliari
LJ
Pope
RM
Constitutively activated Akt-1 is vital for the survival of human monocyte-differentiated macrophages: role of Mcl-1, independent of nuclear factor (NF)-kappaB, Bad, or caspase activation.
J Exp Med.
194
2001
113
126
46
Dhodapkar
MV
Steinman
RM
Krasovsky
J
Munz
C
Bhardwaj
N
Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells.
J Exp Med.
193
2001
233
238
47
Thomson
AW
Lu
L
Dendritic cells as regulators of immune reactivity: implications for transplantation.
Transplantation.
68
1999
1
8
48
Fairchild
PJ
Waldmann
H
Dendritic cells and prospects for transplantation tolerance.
Curr Opin Immunol.
12
2000
528
535
49
Tarte
K
Klein
B
Dendritic cell-based vaccine: a promising approach for cancer immunotherapy.
Leukemia.
13
1999
653
663
50
Banchereau
J
Schuler-Thurner
B
Palucka
AK
Schuler
G
Dendritic cells as vectors for therapy.
Cell.
106
2001
271
274

Author notes

Andrea M. Woltman, Leiden University Medical Center, Department of Nephrology, C3-P, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; e-mail: a.m.woltman@lumc.nl.

Sign in via your Institution