Although dendritic cells (DCs) are the most potent antigen-presenting cells involved in numerous physiologic and pathologic processes, little is known about the signaling pathways that regulate DC activation and antigen-presenting function. Recently, we demonstrated that nuclear factor (NF)-κB activation is central to that process, as overexpression of IκBα blocks the allogeneic mixed lymphocyte reaction (MLR), an in vitro model of T-cell activation. In this study, we investigated the role of 2 putative NF-κB–inducing components, NF-κB–inducing kinase (NIK), and IκB kinase 2 (IKK2). Using an adenoviral gene transfer method to efficiently express dominant-negative (dn) forms of these molecules in monocyte-derived DCs, we found that IKK2dn but not NIKdn inhibited the allogeneic MLR. When DCs were fixed, this inhibitory effect of IKK2dn was lost, suggesting that IKK2 is involved in T-cell–derived signals that enhance DC antigen presentation during the allogeneic MLR period and does not have an effect on viability or differentiation state of DCs prior to coculture with T cells. One such signal is likely to be CD40 ligand (CD40L), as IKK2dn blocked CD40L but not lipopolysaccharide (LPS)–induced NF-κB activation, cytokine production, and up-regulation of costimulatory molecules and HLA-DR in DCs. In summary, our results demonstrate that IKK2 is essential for DC activation induced by CD40L or contact with allogeneic T cells, but not by LPS, whereas NIK is not required for any of these signals. In addition, our results support IKK2 as a potential therapeutic target for the down-regulation of unwanted immune responses that may occur during transplantation or autoimmunity.

Dendritic cells (DCs) are bone marrow–derived cells that constantly circulate from the tissues to the secondary lymphoid organs, where they activate antigen-specific T cells and initiate adaptive immunity to the antigens recognized.1Their unique T-cell stimulatory capacity is due to the expression of high levels of costimulatory molecules that include CD40, CD80, and CD86, and major histocompatibility complex (MHC) class I and II antigens that are essential for the presentation of endogenous and exogenous protein antigens and the production of T-cell stimulatory cytokines such as interleukin-12 (IL-12), IL-15, and IL-18.1-4 

DCs are believed to play a major role in transplant (allograft) rejection by presenting alloantigens to T cells,5 whose recognition of foreign MHC molecules is the primary event initiating allograft rejection. Both donor-derived and infiltrating recipient-derived DCs that take up alloantigen have been shown to activate T cells by migrating to lymphoid tissues.5,6 T cells, in turn, initiate an inflammatory process by the secretion of proinflammatory cytokines and the induction of cytotoxic lymphocytes, ultimately resulting in the rejection of the graft. The observation that neutralizing antibodies or fusion proteins that inhibit efficient DC costimulation by blocking CD80/86-CD28 and CD40-CD40L interactions can prevent allograft rejection in numerous models has supported the importance of DCs in that process.7 In addition, the demonstration that DCs lacking sufficient costimulatory molecule expression induce antigen-specific T-cell anergy in vitro and prolong cardiac and islet allografts in vivo8,9 has suggested that DCs also are involved in the induction of graft tolerance.

Immunosuppressive drugs commonly used in transplantation, such as corticosteroids and cyclosporin A (CsA) or tacromilus (FK506), that block T-cell receptor signaling also have been shown to affect DC differentiation and antigen presentation.10 However, the use and efficacy of these drugs is limited by their broad toxicity, especially in the long-term.11 One approach to improve the success of human allogeneic organ transplantation is to design more specific immunosuppressive drugs, targeted directly to DCs, by understanding what regulates DC antigen-presenting function. Recently, the nuclear factor (NF)–κB transcription factors have been shown to be required for efficient DC antigen-presenting function in both mouse and human DCs,12-14 but the ubiquitous involvement of NF-κB in physiologic and homeostatic processes suggests that widespread and effective inhibition of NF-κB may have hazards. Much more attractive would be the inhibition of the pathways that lead to NF-κB activation in DCs during pathologic processes but do not affect all NF-κB functions.

The activation of NF-κB is a complex process that requires the action of multiple kinases that ultimately result in the phosphorylation and degradation of IκB proteins and the nuclear translocation of NF-κB. The IκB kinases 1 and 2 (IKK1 and IKK2) that directly phosphorylate IκBα and IκBβ have been demonstrated to be important in that process in response to various inflammatory signals in vitro.15-18 IKK2, in particular, was suggested to be more important in the signaling of inflammatory mediators such as lipopolysaccharide (LPS), tumor necrosis factor α (TNFα), and IL-1,16,17,19-27 whereas IKK1 recently was suggested to be more important in development, probably through an NF-κB–independent role.28,29 Another important kinase proposed to act upstream of the IKKs is NF-κB–inducing kinase (NIK).30 NIK has been reported to play a major role in NF-κB activation in response to TNFα, IL-1, and LPS, and to be a key element in the activation of the IKK complex.31However, some recent studies in NIK-deficient mice and human primary cells have questioned its physiologic role in NF-κB activation in response to a variety of stimuli and have suggested that its function may be restricted to signaling through the lymphotoxin β receptor.32-34 Currently, it is not known whether IKK2 or NIK are involved in the activation of NF-κB in DCs that is essential for effective antigen-presenting function.

In this study, we adapted an adenoviral gene transfer technique to efficiently infect more than 95% of immature monocyte-derived DCs.35-37 Using this tool, we investigated the role of IKK2 and NIK in the allogeneic mixed lymphocyte reaction (MLR), an in vitro model of T-cell activation that takes place during allograft rejection. We report here that IKK2 but not NIK is essential for the allogeneic MLR, and we suggest that this may involve T-cell–derived signals such as CD40L that enhance DC antigen-presenting function.

Reagents

Human recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) was a kind gift of Dr G. Larsen (Genetics Institute, Boston, MA). Human recombinant IL-4 and soluble noncovalent trimeric CD40L were purchased from Peprotech (London, United Kingdom). Escherichia coli LPS and lipid A were obtained from Sigma (St Louis, MO), and the mock control and CD40L-transfected mouse fibroblast cell lines were obtained from Prof D. Gray (United Kingdom) and Prof A. Schimpl (Germany).

Isolation of peripheral blood monocytes

Peripheral blood T cells and monocytes were isolated from single donor–platelet pheresis residues of healthy volunteers and purchased from the North London Blood Transfusion Service (Colindale, United Kingdom). Mononuclear cells were isolated by Ficoll/Hypaque centrifugation (specific density, 1.077 g/mL; Nycomed Pharma A.S., Cheshire, United Kingdom) prior to cell separation in a Beckman JE6 elutriator (Torrence, CA). Elutriation was performed in RPMI 1640 containing 1% heat-inactivated fetal calf serum (FCS) (BioWhittaker, Berkshire, United Kingdom). Monocyte purity was always assessed by flow cytometry. Typical T-cell fractions contained more than 80% CD3-expressing cells, less than 5% CD19-expressing cells, and less than 1% CD14-expressing cells, whereas monocyte fractions contained 85%-90% CD14-expressing cells, less than 0.5% CD19-expressing cells, and less than 3% CD3-expressing cells.

Cell culture

Transfected mouse fibroblasts were maintained in RPMI 1640 containing 1% heat-inactivated FCS and 1% penicillin/streptomycin (BioWhittaker). Dendritic cells were generated from peripheral blood monocytes after 5 days of culture in RPMI 1640 containing 5% heat-inactivated FCS, 1% penicillin/streptomycin, 50 ng/mL GM-CSF, and 10 ng/mL IL-4. At day 3 of culture, GM-CSF and IL-4 were replenished. This method is well established to produce dendritic cells of the immature phenotype and presents several advantages when compared to differentiation of dendritic cells directly from blood or bone marrow precursors. Besides being relatively easy and giving high numbers of cells, it generates a homogenous population of cells with a stable “immature DC” phenotype.13,35-37 

Adenoviral vectors and their propagation

Recombinant, replication-deficient adenoviral vectors encoding the E coli β-galactosidase protein (Adβ-gal) or having no insert were provided by Drs A. Byrnes and M. Wood (Oxford, United Kingdom). An adenovirus encoding the jellyfish green fluorescence protein (GFP) (AdGFP) was provided by Quantum Biotech (Carlsbad, AB Canada), and adenoviruses encoding IκBα, (AdIκBα), or a kinase-defective dominant negative form of IKK2 (AdIKK2dn) were a kind gift of Dr R. de Martin (University of Vienna, Austria). Finally, adenoviruses encoding wild-type forms of IKK2 (AdIKK2wt) and NIK (AdNIKwt) or a kinase-defective dominant negative version of NIK (AdNIKdn) were constructed by us. All viruses are E1/E3-deleted, belong to the Ad5 serotype, and have been used previously in other studies.33,38,39 Briefly, viruses were propagated in the 293 human embryonic kidney cell line (ATCC, Middlesex, United Kingdom) and were purified by ultracentrifugation through 2 cesium chloride gradients. Titers of viral stocks were determined by plaque assay in 293 cells as previously described.40 

Gene transfer into monocyte-derived DCs

After 5 days of culture in GM-CSF and IL-4, monocyte-derived DCs were collected, counted, and replated at 1 × 106cells/mL in serum-free RPMI 1640 in 48- or 96-well plates (Falcon, Oxford, United Kingdom), depending on the purpose of the experiment. Cells were then infected with replication-deficient adenoviruses at various multiplicities of infection (MOI) for 2 hours. Subsequently, the adenovirus-containing medium was removed, RPMI 1640 containing 5% heat-inactivated FCS, 1% penicillin/streptomycin, 50 ng/mL GM-CSF, and 10 ng/mL IL-4 was added back, and cells were left to overexpress the transgene of interest for another 1-2 days before further use.

Analysis of infectibility

DCs left uninfected or infected with Ad0 or Adβ-gal at various MOI were collected, washed in 750 μL staining solution (phosphate-buffered saline containing 4% FCS and 10 mM HEPES [N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid]) and centrifuged at 1000 rpm for 5 minutes. They were then incubated at 37°C for 5 minutes before the addition of 50 μL of a 2 mM solution of fluorescein di-(β-D)–galactopyranoside (FDG; Sigma), a β-galactosidase substrate that fluoresces after the reaction takes place. After 1 minute, the reaction was stopped by addition of excess ice-cold staining solution (500 μL). Cell fluorescence was analyzed by flow cytometry on a Becton Dickinson FACScan (Oxford, United Kingdom). When DCs were infected with AdGFP, fluorescence was visualized by fluorescence microscopy (Nikon, Japan).

Analysis of cell death

Uninfected or adenovirus-infected DCs were cultured for 2 days in complete medium prior to being transferred to a 50 μg/mL solution of propidium iodide (PI) before analysis by fluorescence-activated cell-sorter scanner (FACS).41 Late apoptotic or dying cells are PI+. The methyl-thiazol tetrazolium (MTT) assay was also used for the same purpose as previously described (Sigma).42 Analysis of early apoptosis was done by annexin V staining and examination by FACS.43 

Mixed lymphocyte reaction

To assay the immunostimulatory capacity of monocyte-derived DCs, uninfected or adenovirus-infected DCs were cultured in graded doses with 1 × 105 allogeneic elutriated T cells in quadruplicate in a 96-well flat-bottom microtiter plate (Falcon). Proliferation was measured on day 5 by thymidine incorporation after a 16-hour pulse with [3H] thymidine (0.5 μCi/well [0.0185 MBq]; Amersham Life Science, Bucks, United Kingdom). Both irradiated (3000 rad from a 137Cs source) and nonirradiated DCs were used, with no significant differences observed. In some experiments with uninfected DCs, a neutralizing anti-CD40L antibody (Alexis, Nottingham, United Kingdom) or a murine anti–human IgG1 isotype control was added at a concentration of 10 μg/mL to the allogeneic MLR cultures.

Western blotting and electrophoretic mobility shift assay

Two days after infection, 5 × 106 DCs were stimulated with either LPS for 30 minutes or CD40 ligand for 45 minutes. Cytosolic and nuclear extracts were prepared as described.44 Cytosolic proteins were subsequently separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis on a 10% (wt/vol) polyacrylamide gel and transferred onto a nitrocellulose membrane for Western blotting. The antibody for β-galactosidase was from Roche Molecular Diagnostics (East Sussex, United Kingdom), and the antibodies for IKK2 and IκBα were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). In some cases, immunoprecipitations directed toward flag-tagged NIK or IKK2 were performed according to the manufacturer's instructions using 10 μL anti–flag M2 affinity gel (Sigma). Nuclear extracts (10 μg) were examined for NF-κB DNA binding activity by electrophoretic mobility shift assay (EMSA) as previously described.45 

Analysis of cytokines

To examine their cytokine production, monocyte-derived DCs were plated at 1 × 105 cells/well in 96-well flat-bottomed tissue culture plates (Falcon) and were either left uninfected or infected with Adβ-gal, AdNIKdn, AdIKK2dn, or AdIκBα. Cells were stimulated for another 24 hours 2 days after infection with 30 μg/mL soluble CD40L, 100 ng/mL LPS, or 1 μg/mL lipid A. In experiments not shown, these concentrations have been shown to induce maximal cytokine production in DCs. In some cases, 1 × 105 DCs were cocultured with 1 × 105transfected mouse fibroblasts for 24 hours. Supernatants were then collected and analyzed for TNFα, IL-6, and IL-8 by enzyme-linked immunosorbent assay (ELISA) (Pharmingen, Oxford, United Kingdom), and remaining cells were examined for viability by the MTT assay as previously described (Sigma).42 ELISA plates were analyzed at an absorbance of 450 nm on a spectrophotometric ELISA plate reader (Labsystems Multiscan Biochromic, Cambridge, United Kingdom) using a DeltaSoft II.4 software program (Cambridge, United Kingdom). Results are expressed as the mean concentration of triplicate cultures ± SD.

Analysis of cell surface molecules by FACS

Cell surface molecule expression of monocyte-derived DCs was analyzed by FACS staining as previously described.14Antibodies for coxsackie virus and adenovirus receptor (CAR) were donated by Dr R. Finberg (Dana-Farber Cancer Institute, Boston, MA), and antibodies for αVβ5 were provided by Drs W. Smith and J. Gamble (Hanson Centre, Adelaide, Australia). Directly conjugated antibodies for αVβ3, CD80, CD86, HLA-DR, and appropriate isotype-controls were purchased from Pharmingen.

Statistical methods

Mean, standard deviation (SD), standard error of the mean (SEM), normal distribution of data, and statistical tests were calculated using GraphPad version 3 (GraphPad Software, San Diego, CA). For statistical analysis, a 2-sided Student t test of paired comparisons was used.

Adenoviral infection of monocyte-derived DCs

A major problem that has hampered the study of the biochemical signaling pathways involved in DC function is the difficulty of transfecting DNA into DCs.46,47 To circumvent that problem, we developed an alternative approach using replication-deficient adenoviruses. At a relatively low MOI of 300, we previously showed that mature monocyte-derived DCs can be efficiently infected to express the gene of interest14 without the need to be combined with cationic lipids.48 In this study, we modified this technique to infect immature monocyte-derived DCs, the predominant form of DCs found in tissues specialized in taking up and processing antigen.

We first examined whether immature monocyte-derived DCs express receptors involved in adenoviral infection such as the CAR, MHC class I, or the integrins αVβ3 and αVβ5.49-52 We found that although monocyte-derived DCs are not a natural target of adenoviral infection, they do express CAR and MHC class I as well as high levels of αVβ5 but negligible αVβ3 (Figure1A). This suggested susceptibility to infection, and at an MOI of 100, more than 95% of the cells expressed the bacterial protein β-galactosidase after infection with an adenoviral vector encoding the β-galactosidase gene under the control of the cytomegalovirus promoter (Adβ-gal) (Figure 1B) after 1 to 2 days of culture. The presence of β-galactosidase protein was detected by addition of its substrate FDG and detection of fluorescence by FACS. Infectivity was also confirmed by using an adenoviral vector encoding green fluorescent protein (Figure 1C). Another group has also reported that replication-deficient adenoviruses infect immature monocyte-derived DCs and demonstrated that this does not perturb DC function.46 

Fig. 1.

Immature DCs express CAR, MHC class I, and αVβ5, and can be efficiently infected by replication-deficient adenoviruses.

(A) Immature DCs were examined for the expression of CAR, MHC I, αVβ3, and αVβ5 by FACS staining. Cells treated with isotype control antibody (filled histogram) were compared with cells treated with αVβ5, αVβ3, CAR, or MHC class I (HLA-A, -B, -C) monoclonal antibodies (open histogram). (B) Immature DCs were left uninfected or infected with Adβ-gal at various MOI, and examined after for the presence of β-galactosidase by FACS as described in “Materials and methods.” (C) Immature DCs were infected with AdGFP at an MOI of 100 and then examined for the presence of GFP by fluorescence microscopy (original magnification for both panels, × 20). Representative phase-contrast and fluorescent pictures are shown. (D) Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, or AdIKK2dn at an MOI of 100. After 2 days, cells were lysed and extracts examined for the presence of NIKdn or IKK2dn by immunoprecipitation and Western blotting. An antibody directed against the flag epitope attached to NIKdn and IKK2dn but not β-gal was used as previously described.33,39 In all panels, the results of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Fig. 1.

Immature DCs express CAR, MHC class I, and αVβ5, and can be efficiently infected by replication-deficient adenoviruses.

(A) Immature DCs were examined for the expression of CAR, MHC I, αVβ3, and αVβ5 by FACS staining. Cells treated with isotype control antibody (filled histogram) were compared with cells treated with αVβ5, αVβ3, CAR, or MHC class I (HLA-A, -B, -C) monoclonal antibodies (open histogram). (B) Immature DCs were left uninfected or infected with Adβ-gal at various MOI, and examined after for the presence of β-galactosidase by FACS as described in “Materials and methods.” (C) Immature DCs were infected with AdGFP at an MOI of 100 and then examined for the presence of GFP by fluorescence microscopy (original magnification for both panels, × 20). Representative phase-contrast and fluorescent pictures are shown. (D) Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, or AdIKK2dn at an MOI of 100. After 2 days, cells were lysed and extracts examined for the presence of NIKdn or IKK2dn by immunoprecipitation and Western blotting. An antibody directed against the flag epitope attached to NIKdn and IKK2dn but not β-gal was used as previously described.33,39 In all panels, the results of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Close modal

Adenoviral infection of monocyte-derived DCs resulted in the expression of high levels of kinase-deficient dominant negative forms of IKK2 (IKK2dn) and NIK (NIKdn) when AdIKK2dn and AdNIKdn were used at a MOI of 100 (Figure 1D). As a consequence of this overexpression, IKK2dn appears as a doublet, with the upper band representing the whole protein and the lower band a degradation product of it. As both antibodies used for immunoprecipitation and Western blotting are of the same species, the Ig heavy chain of the antibody used to immunoprecipitate IKK2dn and NIKdn is also shown.

Expression of IKK2dn or NIKdn over a 2-day period prior to culture with T cells did not affect the DC viability or differentiation state. DC viability was assessed by PI staining (Figure2A) and the MTT assay as previously described (Sigma)42 (data not shown). Annexin V expression, indicative of early apoptotic events,43 revealed no difference between virus-infected cells and control uninfected cells (data not shown). DC differentiation state was assessed by examining the levels of expression of the costimulatory molecules CD80 and CD86 and the antigen-presenting molecules HLA-A, -B, -C, and -DR. These molecules are expressed in higher levels in cells of the DC phenotype compared to cells of the monocyte/macrophage lineage.35-37Figure 2B (and Figure 6) shows that AdIKK2dn- or AdNIKdn-infected DCs still express similar levels of CD80, CD86, HLA-A, -B, -C, and -DR as uninfected or Adβ-gal–infected cells. This suggests that IKK2dn or NIKdn expression has no effect on the differentiation state of immature DCs.

Fig. 2.

The expression of IKK2dn or NIKdn in immature DCs does not affect their viability or differentiation state.

Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, AdIKK2dn, or AdIκBα at a MOI of 100. (A) After 2 days, cells were examined for viability by PI staining and FACS. Late apoptotic and dying cells stain positive in this assay. (B) After 2 days, cells were stained for cell surface expression of CD80, CD86, HLA-A, -B, -C, and DR and examined by FACS. DCs stained with isotype control (filled histograms) or specific antibody (open histograms) are compared. The results of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Fig. 2.

The expression of IKK2dn or NIKdn in immature DCs does not affect their viability or differentiation state.

Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, AdIKK2dn, or AdIκBα at a MOI of 100. (A) After 2 days, cells were examined for viability by PI staining and FACS. Late apoptotic and dying cells stain positive in this assay. (B) After 2 days, cells were stained for cell surface expression of CD80, CD86, HLA-A, -B, -C, and DR and examined by FACS. DCs stained with isotype control (filled histograms) or specific antibody (open histograms) are compared. The results of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Close modal

Expression of IKK2dn but not NIKdn inhibits the allogeneic MLR

To examine the role of IKK2 and NIK activity in the allogeneic MLR, monocyte-derived DCs were infected at an MOI of 100 with replication-deficient adenoviruses encoding the protein of interest, and after further culture for 2 days they were mixed with allogeneic T cells. We found that inhibition of IKK2 activity via overexpression of IKK2dn but not of a control protein β-galactosidase abrogated the DC-induced T-cell proliferation at all DC doses used (P < .05) (Figure 3A). Overexpression, on the other hand, of NIKdn had no effect on T-cell proliferation in the allogeneic MLR, implying that NIK is not essential in this process. As expected, overexpression of IκBα also abrogated the allogeneic MLR.

Fig. 3.

The allogeneic MLR is inhibited by overexpression of IκBα or IKK2dn but not NIKdn, but this effect is lost after fixation of DCs.

Immature DCs were left uninfected (white) or were infected with Adβ-gal (gray), AdNIKdn (cross-hatched), AdIKK2dn (checkered), or AdIκBα (black). A MOI of 100 was used. (A) DCs were then plated in graded doses with 105 purified, allogeneic T cells in quadruplicate in a 96-well microtiter plate. (B) DCs were first fixed with 1% paraformaldehyde and then plated in graded doses with 105 purified, allogeneic T cells in quadruplicate in a 96-well microtiter plate. Proliferation was determined on day 6 using the [3H]thymidine uptake assay. Each point represents the mean ± SEM from 5 (A) or 4 (B) independent experiments performed on samples from unrelated donors. Statistical significance was assessed by using a one-tailed Student t test using uninfected cells as a control (*P < .05, **P < .01).

Fig. 3.

The allogeneic MLR is inhibited by overexpression of IκBα or IKK2dn but not NIKdn, but this effect is lost after fixation of DCs.

Immature DCs were left uninfected (white) or were infected with Adβ-gal (gray), AdNIKdn (cross-hatched), AdIKK2dn (checkered), or AdIκBα (black). A MOI of 100 was used. (A) DCs were then plated in graded doses with 105 purified, allogeneic T cells in quadruplicate in a 96-well microtiter plate. (B) DCs were first fixed with 1% paraformaldehyde and then plated in graded doses with 105 purified, allogeneic T cells in quadruplicate in a 96-well microtiter plate. Proliferation was determined on day 6 using the [3H]thymidine uptake assay. Each point represents the mean ± SEM from 5 (A) or 4 (B) independent experiments performed on samples from unrelated donors. Statistical significance was assessed by using a one-tailed Student t test using uninfected cells as a control (*P < .05, **P < .01).

Close modal

The inhibition of immature DC-induced T-cell proliferation by IKK2dn and IκBα could be due to an effect on the differentiation or antigen presentation ability of DCs that occurs either before or during the coculture period with T cells. To distinguish between the 2 possibilities, we used fixed DCs that can also induce allogeneic T-cell proliferation, although much less pronounced than that observed with nonfixed DCs.53 We found that fixation of DCs prior to culture with T cells abolished the inhibitory effect of IKK2dn or IκBα overexpression in the allogeneic MLR (Figure 3B). As expected, β-galactosidase or NIKdn expression did not affect this process either. These data are in agreement with the observation that the differentiation state of immature DCs as assessed by expression of costimulatory (CD80, CD86) and antigen-presenting (HLA-DR, HLA-A, -B, -C) molecules is not affected by IKK2dn (Figures 2B and 6) or IκBα (Figure 6). In addition, these data suggest that IKK2dn and IκBα inhibit the allogeneic MLR by blocking a T-cell–derived signal during the coculture period that enhances DC antigen-presenting function.

IKK2dn inhibits CD40L but not LPS-induced NF-κB activation and cytokine production, whereas NIKdn has no effect

We decided then to examine whether IKK2 or NIK is involved in DC activation induced by CD40L and LPS. These stimuli are known to enhance DC antigen-presenting function and T-cell stimulatory capacity by inducing cytokines and by up-regulating antigen-presenting and costimulatory molecules.54,55 In addition, CD40L has been shown previously to be essential for the allogeneic MLR in vitro.56 We have also confirmed this in our system by using an anti-CD40L antibody that we previously showed blocks CD40L-induced activation of the endothelium57 (Figure4A).

Fig. 4.

IKK2dn DC blocks CD40L-induced NF-κB activation and cytokine production in immature DCs.

Immature DCs were left uninfected (white) or were infected with Adβ-gal (gray), AdNIKdn (cross-hatched), AdIKK2dn (checkered), or AdIκBα (black), and cultured for 2 additional days. (A) Graded doses of uninfected cells were cultured with 105 allogeneic T cells in the presence of a neutralizing CD40L antibody or its isotype-matched control. Proliferation was determined on day 6 using the [3H]thymidine uptake assay. (B-D) Cells were stimulated for 24 hours with irradiated 1 × 105 mock control– or CD40L-transfected mouse fibroblasts, and supernatants were collected and assayed for the presence of TNFα, IL-6, and IL-8 by ELISA. Mean values ± SDs of 1 experiment representative of 4 independent experiments are shown. (E) Cells were stimulated with 30 μg/mL of sCD40L for 45 minutes, cytosolic and nuclear extracts collected, and IκBα levels as well as NF-κB DNA-binding activity were determined by Western blotting and EMSA, respectively. (F) Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, AdNIKwt, or combinations of them. An MOI of 100 was used for each virus except in the 2 coinfection conditions, where an MOI of 500 was used to provide a 5-fold excess of Adβ-gal or AdNIKdn virus. After further stimulation for 24 hours, supernatants were collected and assayed for the presence of TNFα by ELISA. Mean values ± SDs of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Fig. 4.

IKK2dn DC blocks CD40L-induced NF-κB activation and cytokine production in immature DCs.

Immature DCs were left uninfected (white) or were infected with Adβ-gal (gray), AdNIKdn (cross-hatched), AdIKK2dn (checkered), or AdIκBα (black), and cultured for 2 additional days. (A) Graded doses of uninfected cells were cultured with 105 allogeneic T cells in the presence of a neutralizing CD40L antibody or its isotype-matched control. Proliferation was determined on day 6 using the [3H]thymidine uptake assay. (B-D) Cells were stimulated for 24 hours with irradiated 1 × 105 mock control– or CD40L-transfected mouse fibroblasts, and supernatants were collected and assayed for the presence of TNFα, IL-6, and IL-8 by ELISA. Mean values ± SDs of 1 experiment representative of 4 independent experiments are shown. (E) Cells were stimulated with 30 μg/mL of sCD40L for 45 minutes, cytosolic and nuclear extracts collected, and IκBα levels as well as NF-κB DNA-binding activity were determined by Western blotting and EMSA, respectively. (F) Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, AdNIKwt, or combinations of them. An MOI of 100 was used for each virus except in the 2 coinfection conditions, where an MOI of 500 was used to provide a 5-fold excess of Adβ-gal or AdNIKdn virus. After further stimulation for 24 hours, supernatants were collected and assayed for the presence of TNFα by ELISA. Mean values ± SDs of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Close modal

For CD40L-induced DC activation, we used a commercially available trimeric soluble recombinant CD40L (sCD40L) or mouse fibroblasts transfected with either a plasmid-expressing CD40L or an empty plasmid that served as a control.58 DCs again were either left uninfected or infected with the replication-deficient adenovirus of interest, and after 2 days the stimulus was added in the cultures for another 24 hours. When used at a 1:1 ratio, irradiated CD40L-transfected cells but not control fibroblasts induced high levels of expression of TNFα, IL-6, and IL-8 (Figure 4B-D). This could be inhibited by AdIKK2dn and AdIκBα but not NIKdn, suggesting that IKK2 is essential for CD40L-induced NF-κB activation in immature DCs. Similar data were obtained with sCD40L used at a previously determined optimal concentration of 30 μg/mL (data not shown). AdIKK2dn but not AdNIKdn could also inhibit sCD40L-induced IκBα degradation and NF-κB activation as measured by the electrophoretic mobility shift assay (Figure 4E). In all cases, viability of the cells was checked after collecting the supernatants by doing an MTT assay42 and was not affected (data not shown). To exclude the possibility that NIKdn could not act as a functional dominant negative inhibitor in immature DCs, coinfection experiments were performed using wild-type NIK (NIKwt) as the inducing stimulus as previously described.33 Expression of NIKwt-induced TNFα production was inhibited when NIKdn was coinfected at an equal or 5-fold excess titer (Figure 4F).

In contrast, activation of DCs with LPS or its lipid A portion induced TNFα, IL-6, and IL-8 production that was not inhibited by the expression of either IKK2dn or NIKdn (Figure5A-C). This was independent of the LPS concentrations used, as at lower LPS doses TNFα (Figure 5D), IL-6, and IL-8 (data not shown) were not blocked by IKK2dn or NIKdn. The fact that LPS- or lipid A–induced DC cytokine production is not blocked further demonstrates that DC viability is not affected by the expression of IKK2dn or NIKdn. At the same time, neither AdIKK2dn (Figure 5E) nor AdNIKdn (data not shown) inhibited IκBα degradation and NF-κB activation in response to LPS or lipid A, whereas AdIκBα did (Figure 5E). These observations suggest that the involvement of IKK2 in NF-κB activation and NF-κB–dependent cytokine production in DCs depends on the stimulus, as it is required for CD40 but not for LPS receptor signaling. However, in both instances activation is NF-κB–dependent since it is blocked by IκBα overexpression. In contrast, NIK seems to be redundant with respect to NF-κB activation when LPS, CD40L, or contact with activated T cells are used as a stimulus.

Fig. 5.

Expression of IKK2dn or NIKdn into DCs does not block LPS- or lipid A–induced NF-κB activation and cytokine production.

Immature DCs were left uninfected (white) or were infected with Adβ-gal (gray), AdNIKdn (cross-hatched), AdIKK2dn (checkered), and AdIκBα (black), and cultured for 2 additional days. (A-C) Cells were stimulated for 24 hours with 100 ng/mL LPS or 1 μg/mL of lipid A, and supernatants collected and assayed for the presence of TNFα, IL-6, and IL-8 by ELISA. Mean values ± SDs of 1 experiment representative of 4 independent experiments performed on samples from from unrelated donors are shown. (D) Cells were stimulated with various concentrations of LPS, and supernatants collected and assayed for the presence of TNFα by ELISA. Mean values ± SDs of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown. (E) After stimulation with 100 ng/mL LPS for 45 minutes, cytosolic and nuclear extracts were collected. The presence of cytosolic IKK2 and IκBα was examined by Western blotting and the nuclear NF-κB DNA binding activity determined by EMSA.

Fig. 5.

Expression of IKK2dn or NIKdn into DCs does not block LPS- or lipid A–induced NF-κB activation and cytokine production.

Immature DCs were left uninfected (white) or were infected with Adβ-gal (gray), AdNIKdn (cross-hatched), AdIKK2dn (checkered), and AdIκBα (black), and cultured for 2 additional days. (A-C) Cells were stimulated for 24 hours with 100 ng/mL LPS or 1 μg/mL of lipid A, and supernatants collected and assayed for the presence of TNFα, IL-6, and IL-8 by ELISA. Mean values ± SDs of 1 experiment representative of 4 independent experiments performed on samples from from unrelated donors are shown. (D) Cells were stimulated with various concentrations of LPS, and supernatants collected and assayed for the presence of TNFα by ELISA. Mean values ± SDs of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown. (E) After stimulation with 100 ng/mL LPS for 45 minutes, cytosolic and nuclear extracts were collected. The presence of cytosolic IKK2 and IκBα was examined by Western blotting and the nuclear NF-κB DNA binding activity determined by EMSA.

Close modal

IKK2dn blocks CD40L but not LPS-induced up-regulation of CD80, CD86, and HLA-DR

The unique ability of DCs to stimulate T-cell responses is due in part to the high levels of costimulatory molecules that they display. For that reason we examined whether the expression of IKK2dn, NIKdn, or IκBα prevents the up-regulation of these molecules after CD40L and LPS-induced DC activation. Again, monocyte-derived DCs were infected with replication-deficient adenoviruses. After 1 day, one group of cells was left unstimulated, whereas the other was stimulated with 30 μg/mL of soluble CD40L or 100 ng/mL LPS for another 2 days, and cells were then stained for the presence of surface CD80, CD86, and HLA-DR. These markers increase upon DC activation and play a major role in enhancing the T-cell stimulatory ability of DCs.54 We found that although IKK2dn had no effect on the constitutive expression of these costimulatory molecules, it abrogated the up-regulation of CD80 (P < .05), CD86 (P < .05), and HLA-DR (P < .05) after CD40 triggering (Figure6A, Table1). In contrast, IKK2dn could not abrogate LPS-induced up-regulation of CD80, CD86, and HLA-DR (Figure6B), whereas NIKdn had no effect in the up-regulation of these molecules regardless of which stimulus was used (Figure 6A-B). At the same time, IκBα inhibited both CD40L and LPS-induced up-regulation of CD80 (P < .05 and P < .01, respectively), CD86 (P < .01 in both cases), and HLA-DR (P < .05 and P < .01, respectively) (Figure 6A-B, Table 1). Statistical significance was analyzed by using mean fluorescence intensity units from experiments from 4 independent donors and the 2-sided Student t test for parametric data. Because CD40L is a T-cell surface molecule believed to be essential for the allogeneic MLR,56 these data suggest that IKK2dn may inhibit DC antigen-presenting function during the allogeneic MLR by blocking CD4OL-induced DC activation after contact with T cells.

Fig. 6.

Expression of IKK2dn blocks CD40L-induced up-regulation of MHC antigen-presenting and costimulatory molecules.

Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, AdIKK2dn, and AdIκBα. After culture for 2 additional days, DCs were stimulated for 48 hours with 30 μg/mL of sCD40L (A) or 100 ng/mL LPS (B) and then examined for the expression of CD80, CD86, and HLA-DR by FACS staining. Unstimulated DCs (filled histogram) were compared with sCD40L or LPS-stimulated DCs (open histogram) treated with isotype control (IC), CD80, CD86, and HLA-DR directly conjugated monoclonal antibodies. The results of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Fig. 6.

Expression of IKK2dn blocks CD40L-induced up-regulation of MHC antigen-presenting and costimulatory molecules.

Immature DCs were left uninfected or infected with Adβ-gal, AdNIKdn, AdIKK2dn, and AdIκBα. After culture for 2 additional days, DCs were stimulated for 48 hours with 30 μg/mL of sCD40L (A) or 100 ng/mL LPS (B) and then examined for the expression of CD80, CD86, and HLA-DR by FACS staining. Unstimulated DCs (filled histogram) were compared with sCD40L or LPS-stimulated DCs (open histogram) treated with isotype control (IC), CD80, CD86, and HLA-DR directly conjugated monoclonal antibodies. The results of 1 experiment representative of 3 independent experiments performed on samples from unrelated donors are shown.

Close modal
Table 1.

Mean fluorescence intensity units ± SEMs of CD80, CD86, and HLA-DR expression of DCs before and after stimulation with LPS or CD40L

No stimulusLPS (P)CD40L (P)
Isotype control    
 Uninfected 7 ± 1 3 ± 1 7 ± 1 
 Adβ-gal 8 ± 2 3 ± 1 7 ± 1 
 AdNIKdn 8 ± 1 3 ± 1 7 ± 1 
 AdIKK2dn 8 ± 1 3 ± 1 7 ± 2 
 AdIκBα 7 ± 2 4 ± 0 7 ± 2 
CD80    
 Uninfected 22 ± 6 58 ± 2 43 ± 6 
 Adβ-gal 22 ± 5 56 ± 9 43 ± 6 
 AdNIKdn 20 ± 5 61 ± 2 41 ± 6 
 AdIKK2dn 17 ± 2 54 ± 9 18 ± 5 (< .05) 
 AdIκBα 17 ± 4 22 ± 2 (< .01) 16 ± 3 (< .05) 
CD86    
 Uninfected 20 ± 4 51 ± 7 46 ± 5 
 Adβ-gal 21 ± 5 55 ± 3 46 ± 4 
 AdNIKdn 24 ± 5 58 ± 2 48 ± 4 
 AdIKK2dn 22 ± 4 54 ± 2 27 ± 5 (< .05) 
 AdIκBα 22 ± 5 19 ± 5 (< .01) 23 ± 3 (< .01) 
HLA-DR    
 Uninfected 82 ± 7 113 ± 10 137 ± 36 
 Adβ-gal 83 ± 3 117 ± 1 149 ± 33 
 AdNIKdn 78 ± 9 127 ± 1 132 ± 25 
 AdIKK2dn 90 ± 3 129 ± 6 79 ± 14 (< .05) 
 AdIκBα 80 ± 8 69 ± 9 (< .01) 66 ± 8 (< .05) 
No stimulusLPS (P)CD40L (P)
Isotype control    
 Uninfected 7 ± 1 3 ± 1 7 ± 1 
 Adβ-gal 8 ± 2 3 ± 1 7 ± 1 
 AdNIKdn 8 ± 1 3 ± 1 7 ± 1 
 AdIKK2dn 8 ± 1 3 ± 1 7 ± 2 
 AdIκBα 7 ± 2 4 ± 0 7 ± 2 
CD80    
 Uninfected 22 ± 6 58 ± 2 43 ± 6 
 Adβ-gal 22 ± 5 56 ± 9 43 ± 6 
 AdNIKdn 20 ± 5 61 ± 2 41 ± 6 
 AdIKK2dn 17 ± 2 54 ± 9 18 ± 5 (< .05) 
 AdIκBα 17 ± 4 22 ± 2 (< .01) 16 ± 3 (< .05) 
CD86    
 Uninfected 20 ± 4 51 ± 7 46 ± 5 
 Adβ-gal 21 ± 5 55 ± 3 46 ± 4 
 AdNIKdn 24 ± 5 58 ± 2 48 ± 4 
 AdIKK2dn 22 ± 4 54 ± 2 27 ± 5 (< .05) 
 AdIκBα 22 ± 5 19 ± 5 (< .01) 23 ± 3 (< .01) 
HLA-DR    
 Uninfected 82 ± 7 113 ± 10 137 ± 36 
 Adβ-gal 83 ± 3 117 ± 1 149 ± 33 
 AdNIKdn 78 ± 9 127 ± 1 132 ± 25 
 AdIKK2dn 90 ± 3 129 ± 6 79 ± 14 (< .05) 
 AdIκBα 80 ± 8 69 ± 9 (< .01) 66 ± 8 (< .05) 

A Student 2-sided t test for parametric data was used for statistical analysis. The null hypothesis was that expression of the respective surface molecule in adenovirus-infected cells was not different from the control uninfected cells.

The pathophysiology of transplant rejection is characterized by the activation of alloantigen-specific CD4+ T cells, the secretion of proinflammatory cytokines and chemokines, the formation of an infiltrate in the graft consisting of helper and cytotoxic T cells as well as macrophages, and the production of highly specific alloreactive antibodies by B cells59 that ultimately result in graft destruction. The initiation of this process requires the recognition of alloantigens by T cells. Alloantigen presentation can be mediated either “directly” by intact allo-MHC on the surface of donor DCs or “indirectly” by recipient DCs that process peptides derived from donor MHC.5,6 The aim of our study was to characterize some of the signaling pathways involved in DC alloantigen presentation in the hope that a better understanding of that process may lead to improved and less-toxic ways of preventing allograft rejection.

First, we developed an adenoviral gene transfer method to efficiently infect immature DCs. We found that immature monocyte-derived DCs express high levels of CAR, MHC class I, and the integrin αVβ5 that have been proposed to act as receptors/coreceptors for the adenoviral entry into cells. Adenoviral infection took place at relatively low MOI, with an MOI of 100 resulting in more than 95% of DCs expressing the transgene. This efficiency allows the inhibition of DC function in complex systems such as the allogeneic MLR and the examination of endogenous genes without the need to use reporter constructs.

Next, we concentrated our study on the role of intracellular signaling pathways involved in the allogeneic MLR, an in vitro model of T-cell activation that occurs during allograft rejection. Previous work from us13,14 and others12,60 has demonstrated that NF-κB activation is required for efficient DC antigen-presenting function during the allogeneic MLR, because prevention of IκBα degradation completely abrogates the allogeneic T-cell proliferation. In addition, other studies in mice have suggested that the NF-κB subunit relB is essential for effective antigen presentation of DCs,61,62 whereas c-rel is required for cross-priming of DCs to induce cytotoxic T-lymphocyte responses.63To understand what regulates NF-κB in DCs, we decided to examine the involvement of IKK2 and NIK, 2 upstream kinases that have been shown to be essential for NF-κB activation in several settings.18We found that IKK2 but not NIK activity is required for effective alloantigen presentation during the MLR in vitro. Interestingly, IKK2dn did not affect the differentiation of DCs prior to coculture with T cells, which was assessed by examining the expression of antigen-presenting and costimulatory molecules such as HLA-DR, HLA-A, -B, -C, CD80, and CD86, or the ability of fixed DCs to induce T-cell proliferation. This suggested that IKK2 activity is required for a T-cell–derived signal that enhances DC antigen-presenting function during the MLR rather than an effect of IKK2dn on DC differentiation prior to the coculture period. T-cell–induced DC activation in response to allo-MHC determinants during the allogeneic MLR has been described.64,65 

It is very difficult to determine what activates NF-κB and enhances DC antigen-presenting function during the complex process and the complex cell interactions of the allogeneic MLR. It is considered that one of the major signals involved is CD40L expressed by T cells.66 CD40L enhances DC T-cell stimulatory ability by inducing their activation with the consequent production of cytokines and the up-regulation of antigen-presenting and costimulatory molecules.54,67 Blockade of CD40-CD40L interactions prevents TH1 cytokine production and virtually abolishes cytotoxic T-cell effector generation during the primary MLR in vitro.56 Moreover, disruption of CD40-CD40L interactions abrogates the induction of cytotoxic T-cell responses in vivo.68,69 Our observation that IKK2 but not NIK is essential for CD40L-induced NF-κB activation, cytokine production, and up-regulation of the costimulatory molecules CD80 and CD86, and HLA-DR in DCs, suggests that its major role in the allogeneic MLR could be due mainly to its ability to prevent effective CD40-CD40L interactions from taking place.

Although IKK2 is essential for allogeneic T-cell– and CD40L-induced DC activation, it is not always involved in that process, as we have found that LPS or its lipid A portion can induce NF-κB activation and cytokine production in an IKK2-independent manner. This contradicts the current belief that IKK2 is essential for LPS-induced NF-κB activation18 and suggests that at least in DCs this is not the case. In DCs, IKK2 use is receptor-restricted with CD40L and contact with allogeneic T cells inducing IKK2-dependent and LPS inducing IKK2-independent NF-κB activation. This may have potential advantages in the design of therapeutic strategies, because agents that inhibit IKK2 will not compromise the ability of the immune system to recognize and respond to the bacterial product LPS upon bacterial infections, and probably other signals too. NIK, on the other hand, is not involved in NF-κB activation in response to CD40L, contact with allogeneic T cells, or LPS stimulation in DCs. This, however, does not exclude the involvement of NIK in other yet-unidentified pathways that also lead to NF-κB activation. We33 and others32,34 have recently found evidence that NIK is essential for lymphotoxin β receptor (LTβR)–induced NF-κB activation and may thus have a more restricted role in the regulation of NF-κB.

Some other interesting points arise from this study. First, NF-κB activity is not involved in the constitutive expression of antigen-presenting and costimulatory molecules in immature DCs but only plays a role in their up-regulation during DC maturation. This is supported by studies that we previously performed in mature DCs, where the overexpression of IκBα down-regulates the expression of antigen-presenting and costimulatory molecules only to levels found in immature DCs.13 This is in contrast to data obtained with pharmacological NF-κB inhibitors such as N-acetyl-L-cysteine (NAC) and aspirin that also decrease the cell surface expression of CD80, CD86, and HLA-DR in immature DCs,70,71 but whether this is due to a direct consequence of NF-κB activation or nonspecific effects of the inhibition of other pathways too is not clear. Certainly, in our study the specific inhibition of NF-κB through overexpression of IκBα does not have such effects (Figure 2A). Second, NF-κB is not required for survival of DCs. This confirms recent data of others that phosphatidolinositol-3-kinase and p42/44 mitogen-activated protein kinase rather than NF-κB are required for that process.12,72 

The implications of our findings are relevant clinically. The allogeneic MLR is not only a system that measures DC antigen-presenting function but also an in vitro model of allograft rejection. Although it is simplistic to believe that it reflects exactly how T-cell activation takes place early in vivo in the process of transplant rejection, its value as a tool for the study of the molecular mechanisms involved cannot be contested. Neutralizing antibodies or proteins that prevent IL-2 signaling or CD80/86-CD28 interactions that were originally shown to inhibit the allogeneic MLR73-75 also have been shown to be efficient in vivo7,76 and are beginning to enter the clinical arena.11 A humanized murine monoclonal antibody, in particular, targeted against the 55-kDa α chain of the IL-2 receptor has received approval even for its use in humans in combination with standard cyclosporin-based immunosuppressive therapy.77 Our observations thus suggest that IKK2 may be a good therapeutic target for the prevention of transplant rejection as well as other inflammatory and autoimmune diseases where DCs also are thought to play a major role by presenting antigen and initiating inappropriate inflammatory responses.78,79 In addition, DCs are known to be important in tolerance induction.80,81We recently have shown that prevention of NF-κB activation in DCs induces T-cell tolerance in vitro.13 Our observation that this process requires IKK2 suggests that inhibition of its activity also should promote tolerance that could again be beneficial in transplantation and autoimmunity.

We would like to thank Prof B. Vogelstein, Prof D. Wallach, Prof R. Hay, Dr A. Byrnes, Dr M. Wood, and Dr R. de Martin for the reagents used in this study. We would also like to thank Dr M. Osman for reading the manuscript.

Prepublished online as Blood First Edition Paper, September 19, 2002; DOI 10.1182/blood-2002-06-1835.

Supported by the Arthritis Research Campaign (United Kingdom) and the Wellcome Trust.

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
Banchereau
J
Steinman
RM
Dendritic cells and the control of immunity.
Nature.
392
1998
245
252
2
Caux
C
Massacrier
C
Dezutter-Dambuyant
C
et al
Human dendritic Langerhans cells generated in vitro from CD34+ progenitors can prime naive CD4+ T cells and process soluble antigen.
J Immunol.
155
1995
5427
5435
3
Hart
DN
Dendritic cells: unique leukocyte populations which control the primary immune response.
Blood.
90
1997
3245
3287
4
de Saint-Vis
B
Fugier-Vivier
I
Massacrier
C
et al
The cytokine profile expressed by human dendritic cells is dependent on cell subtype and mode of activation.
J Immunol.
160
1998
1666
1676
5
Larsen
CP
Steinman
RM
Witmer-Pack
M
Hankins
DF
Morris
PJ
Austyn
JM
Migration and maturation of Langerhans cells in skin transplants and explants.
J Exp Med.
172
1990
1483
1493
6
Gould
DS
Auchincloss
H
Jr
Direct and indirect recognition: the role of MHC antigens in graft rejection.
Immunol Today.
20
1999
77
82
7
Dai
Z
Lakkis
FG
The role of cytokines, CTLA-4 and costimulation in transplant tolerance and rejection.
Curr Opin Immunol.
11
1999
504
508
8
Fu
F
Li
Y
Qian
S
et al
Costimulatory molecule-deficient dendritic cell progenitors (MHC class II+, CD80dim, CD86-) prolong cardiac allograft survival in nonimmunosuppressed recipients.
Transplantation.
62
1996
659
665
9
Rastellini
C
Lu
L
Ricordi
C
Starzl
TE
Rao
AS
Thomson
AW
Granulocyte/macrophage colony-stimulating factor–stimulated hepatic dendritic cell progenitors prolong pancreatic islet allograft survival.
Transplantation.
60
1995
1366
1370
10
Woltman
AM
de Fijter
JW
Kamerling
SW
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
11
Vella
JP
Sayegh
MH
Current and future immunosuppressive therapies: impact on chronic allograft dysfunction.
J Nephrol.
10
1997
229
231
12
Rescigno
M
Martino
M
Sutherland
CL
Gold
MR
Ricciardi-Castagnoli
P
Dendritic cell survival and maturation are regulated by different signaling pathways.
J Exp Med.
188
1998
2175
2180
13
Yoshimura
S
Bondeson
J
Brennan
FM
Foxwell
BM
Feldmann
M
Role of NFkappaB in antigen presentation and development of regulatory T cells elucidated by treatment of dendritic cells with the proteasome inhibitor PSI.
Eur J Immunol.
31
2001
1883
1893
14
Yoshimura
S
Bondeson
J
Foxwell
BM
Brennan
FM
Feldmann
M
Effective antigen presentation by dendritic cells is NF-kappaB dependent: coordinate regulation of MHC, co-stimulatory molecules and cytokines.
Int Immunol.
13
2001
675
683
15
DiDonato
JA
Hayakawa
M
Rothwarf
DM
Zandi
E
Karin
M
A cytokine-responsive IkappaB kinase that activates the transcription factor NF-kappaB.
Nature.
388
1997
548
554
16
Mercurio
F
Zhu
H
Murray
BW
et al
IKK-1 and IKK-2: cytokine-activated IkappaB kinases essential for NF-kappaB activation.
Science.
278
1997
860
866
17
Regnier
CH
Song
HY
Gao
X
Goeddel
DV
Cao
Z
Rothe
M
Identification and characterization of an IkappaB kinase.
Cell.
90
1997
373
383
18
Karin
M
Delhase
M
The I kappa B kinase (IKK) and NF-kappa B: key elements of proinflammatory signalling.
Semin Immunol.
12
2000
85
98
19
Woronicz
JD
Gao
X
Cao
Z
Rothe
M
Goeddel
DV
IkappaB kinase-beta: NF-kappaB activation and complex formation with IkappaB kinase-alpha and NIK.
Science.
278
1997
866
869
20
Zandi
E
Rothwarf
DM
Delhase
M
Hayakawa
M
Karin
M
The IkappaB kinase complex (IKK) contains two kinase subunits, IKKalpha and IKKbeta, necessary for IkappaB phosphorylation and NF-kappaB activation.
Cell.
91
1997
243
252
21
O'Connell
MA
Bennett
BL
Mercurio
F
Manning
AM
Mackman
N
Role of IKK1 and IKK2 in lipopolysaccharide signaling in human monocytic cells.
J Biol Chem.
273
1998
30410
30414
22
Fischer
C
Page
S
Weber
M
Eisele
T
Neumeier
D
Brand
K
Differential effects of lipopolysaccharide and tumor necrosis factor on monocytic IkappaB kinase signal some activation and IkappaB proteolysis.
J Biol Chem.
274
1999
24625
24632
23
Algarte
M
Nguyen
H
Heylbroeck
C
Lin
R
Hiscott
J
IkappaB-mediated inhibition of virus-induced beta interferon transcription.
J Virol.
73
1999
2694
2702
24
Delhase
M
Hayakawa
M
Chen
Y
Karin
M
Positive and negative regulation of IkappaB kinase activity through IKKbeta subunit phosphorylation.
Science.
284
1999
309
313
25
Li
ZW
Chu
W
Hu
Y
et al
The IKKbeta subunit of IkappaB kinase (IKK) is essential for nuclear factor kappaB activation and prevention of apoptosis.
J Exp Med.
189
1999
1839
1845
26
Li
Q
Van Antwerp
D
Mercurio
F
Lee
KF
Verma
IM
Severe liver degeneration in mice lacking the IkappaB kinase 2 gene.
Science.
284
1999
321
325
27
Tanaka
M
Fuentes
ME
Yamaguchi
K
et al
Embryonic lethality, liver degeneration, and impaired NF-kappa B activation in IKK-beta–deficient mice.
Immunity.
10
1999
421
429
28
Hu
Y
Baud
V
Delhase
M
et al
Abnormal morphogenesis but intact IKK activation in mice lacking the IKKalpha subunit of IkappaB kinase.
Science.
284
1999
316
320
29
Takeda
K
Takeuchi
O
Tsujimura
T
et al
Limb and skin abnormalities in mice lacking IKKalpha.
Science.
284
1999
313
316
30
Malinin
NL
Boldin
MP
Kovalenko
AV
Wallach
D
MAP3K-related kinase involved in NF-kappaB induction by TNF, CD95 and IL-1.
Nature.
385
1997
540
544
31
Karin
M
Ben-Neriah
Y
Phosphorylation meets ubiquitination: the control of NF-κB activity.
Annu Rev Immunol.
18
2000
621
663
32
Matsushima
A
Kaisho
T
Rennert
PD
et al
Essential role of nuclear factor (NF)-kappaB-inducing kinase and inhibitor of kappaB (IkappaB) kinase alpha in NF-kappaB activation through lymphotoxin beta receptor, but not through tumor necrosis factor receptor I.
J Exp Med.
193
2001
631
636
33
Smith
C
Andreakos
E
Crawley
JB
Brennan
FM
Feldmann
M
Foxwell
BM
NF-kappaB–inducing kinase is dispensable for activation of NF-kappaB in inflammatory settings but essential for lymphotoxin beta receptor activation of NF-kappaB in primary human fibroblasts.
J Immunol.
167
2001
5895
5903
34
Yin
L
Wu
L
Wesche
H
et al
Defective lymphotoxin-beta receptor–induced NF-kappaB transcriptional activity in NIK-deficient mice.
Science.
291
2001
2162
2165
35
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
36
Romani
N
Reider
D
Heuer
M
et al
Generation of mature dendritic cells from human blood: an improved method with special regard to clinical applicability.
J Immunol Methods.
196
1996
137
151
37
Bender
A
Sapp
M
Schuler
G
Steinman
RM
Bhardwaj
N
Improved methods for the generation of dendritic cells from nonproliferating progenitors in human blood.
J Immunol Methods.
196
1996
121
135
38
Wrighton
CJ
Hofer-Warbinek
R
Moll
T
Eytner
R
Bach
FH
de Martin
R
Inhibition of endothelial cell activation by adenovirus-mediated expression of I kappa B alpha, an inhibitor of the transcription factor NF-kappa B.
J Exp Med.
183
1996
1013
1022
39
Oitzinger
W
Hofer-Warbinek
R
Schmid
JA
Koshelnick
Y
Binder
BR
de Martin
R
Adenovirus-mediated expression of a mutant IkappaB kinase 2 inhibits the response of endothelial cells to inflammatory stimuli.
Blood.
97
2001
1611
1617
40
Graham
FL
Prevec
L
Methods for construction of adenovirus vectors.
Mol Biotechnol.
3
1995
207
220
41
Nicoletti
I
Migliorati
G
Pagliacci
MC
Grignani
F
Riccardi
C
A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry.
J Immunol Methods.
139
1991
271
279
42
Mosmann
T
Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays.
J Immunol Methods.
65
1983
55
63
43
Koopman
G
Reutelingsperger
CP
Kuijten
GA
Keehnen
RM
Pals
ST
van Oers
MH
Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis.
Blood.
84
1994
1415
1420
44
Whiteside
ST
Visvanathan
KV
Goodbourn
S
Identification of novel factors that bind to the PRD I region of the human beta-interferon promoter.
Nucleic Acids Res.
20
1992
1531
1538
45
Clarke
CJ
Taylor-Fishwick
DA
Hales
A
et al
Interleukin-4 inhibits kappa light chain expression and NF kappa B activation but not I kappa B alpha degradation in 70Z/3 murine pre-B cells.
Eur J Immunol.
25
1995
2961
2966
46
Zhong
L
Granelli-Piperno
A
Choi
Y
Steinman
RM
Recombinant adenovirus is an efficient and non-perturbing genetic vector for human dendritic cells.
Eur J Immunol.
29
1999
964
972
47
Arthur
JF
Butterfield
LH
Roth
MD
et al
A comparison of gene transfer methods in human dendritic cells.
Cancer Gene Ther.
4
1997
17
25
48
Dietz
AB
Vuk-Pavlovic
S
High efficiency adenovirus-mediated gene transfer to human dendritic cells.
Blood.
91
1998
392
398
49
Wickham
TJ
Carrion
ME
Kovesdi
I
Targeting of adenovirus penton base to new receptors through replacement of its RGD motif with other receptor-specific peptide motifs.
Gene Ther.
2
1995
750
756
50
Bergelson
JM
Cunningham
JA
Droguett
G
et al
Isolation of a common receptor for Coxsackie B viruses and adenoviruses 2 and 5.
Science.
275
1997
1320
1323
51
Tomko
RP
Xu
R
Philipson
L
HCAR and MCAR: the human and mouse cellular receptors for subgroup C adenoviruses and group B coxsackieviruses.
Proc Natl Acad Sci U S A.
94
1997
3352
3356
52
Hong
SS
Karayan
L
Tournier
J
Curiel
DT
Boulanger
PA
Adenovirus type 5 fiber knob binds to MHC class I alpha2 domain at the surface of human epithelial and B lymphoblastoid cells.
Embo J.
16
1997
2294
2306
53
Morel
AS
Quaratino
S
Douek
DC
Londei
M
Split activity of interleukin-10 on antigen capture and antigen presentation by human dendritic cells: definition of a maturative step.
Eur J Immunol.
27
1997
26
34
54
Cella
M
Scheidegger
D
Palmer-Lehmann
K
Lane
P
Lanzavecchia
A
Alber
G
Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation.
J Exp Med.
184
1996
747
752
55
Sallusto
F
Cella
M
Danieli
C
Lanzavecchia
A
Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: downregulation by cytokines and bacterial products.
J Exp Med.
182
1995
389
400
56
Blazar
BR
Taylor
PA
Panoskaltsis-Mortari
A
et al
Blockade of CD40 ligand-CD40 interaction impairs CD4+ T cell–mediated alloreactivity by inhibiting mature donor T cell expansion and function after bone marrow transplantation.
J Immunol.
158
1997
29
39
57
Monaco
C
Andreakos
E
Young
S
Feldmann
M
Paleolog
E
T cell–mediated signaling to vascular endothelium: induction of cytokines, chemokines, and tissue factor.
J Leukoc Biol.
71
2002
659
668
58
Wohlleben
G
Gray
D
Schimpl
A
In vitro immunization of naive mouse B cells: establishment of IgM secreting hybridomas specific for soluble protein or hapten from B cells cultured on CD40 ligand transfected mouse fibroblasts.
Int Immunol.
8
1996
343
349
59
Hernandez-Fuentes
MP
Baker
RJ
Lechler
RI
The alloresponse.
Rev Immunogenet.
1
1999
282
296
60
Oyama
T
Ran
S
Ishida
T
et al
Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factor-kappa B activation in hemopoietic progenitor cells.
J Immunol.
160
1998
1224
1232
61
Burkly
L
Hession
C
Ogata
L
et al
Expression of relB is required for the development of thymic medulla and dendritic cells.
Nature.
373
1995
531
536
62
Carrasco
D
Ryseck
RP
Bravo
R
Expression of relB transcripts during lymphoid organ development: specific expression in dendritic antigen-presenting cells.
Development.
118
1993
1221
1231
63
Mintern
JD
Belz
G
Gerondakis
S
Carbone
FR
Heath
WR
The cross-priming APC requires a Rel-dependent signal to induce CTL.
J Immunol.
168
2002
3283
3287
64
McLellan
AD
Heiser
A
Hart
DN
Induction of dendritic cell costimulator molecule expression is suppressed by T cells in the absence of antigen-specific signalling: role of cluster formation, CD40 and HLA-class II for dendritic cell activation.
Immunology.
98
1999
171
180
65
Muraille
E
De Trez
C
Pajak
B
Brait
M
Urbain
J
Leo
O
T cell–dependent maturation of dendritic cells in response to bacterial superantigens.
J Immunol.
168
2002
4352
4360
66
Hollenbaugh
D
Grosmaire
LS
Kullas
CD
et al
The human T cell antigen gp39, a member of the TNF gene family, is a ligand for the CD40 receptor: expression of a soluble form of gp39 with B cell co-stimulatory activity.
Embo J.
11
1992
4313
4321
67
Koch
F
Stanzl
U
Jennewein
P
et al
High level IL-12 production by murine dendritic cells: upregulation via MHC class II and CD40 molecules and downregulation by IL-4 and IL-10.
J Exp Med.
184
1996
741
746
68
Bennett
SR
Carbone
FR
Karamalis
F
Flavell
RA
Miller
JF
Heath
WR
Help for cytotoxic T-cell responses is mediated by CD40 signalling.
Nature.
393
1998
478
480
69
Schoenberger
SP
Toes
RE
van der Voort
EI
Offringa
R
Melief
CJ
T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions.
Nature.
393
1998
480
483
70
Verhasselt
V
Vanden Berghe
W
Vanderheyde
N
Willems
F
Haegeman
G
Goldman
M
N-acetyl-L-cysteine inhibits primary human T cell responses at the dendritic cell level: association with NF-kappaB inhibition.
J Immunol.
162
1999
2569
2574
71
Hackstein
H
Morelli
AE
Larregina
AT
et al
Aspirin inhibits in vitro maturation and in vivo immunostimulatory function of murine myeloid dendritic cells.
J Immunol.
166
2001
7053
7062
72
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
73
Depper
JM
Leonard
WJ
Robb
RJ
Waldmann
TA
Greene
WC
Blockade of the interleukin-2 receptor by anti-Tac antibody: inhibition of human lymphocyte activation.
J Immunol.
131
1983
690
696
74
Azuma
M
Cayabyab
M
Buck
D
Phillips
JH
Lanier
LL
CD28 interaction with B7 costimulates primary allogeneic proliferative responses and cytotoxicity mediated by small, resting T lymphocytes.
J Exp Med.
175
1992
353
360
75
Tan
P
Anasetti
C
Hansen
JA
et al
Induction of alloantigen-specific hyporesponsiveness in human T lymphocytes by blocking interaction of CD28 with its natural ligand B7/BB1.
J Exp Med.
177
1993
165
173
76
Waldmann
TA
O'Shea
J
The use of antibodies against the IL-2 receptor in transplantation.
Curr Opin Immunol.
10
1998
507
512
77
Vincenti
F
Kirkman
R
Light
S
et al
Interleukin-2 receptor blockade with daclizumab to prevent acute rejection in renal transplantation: Daclizumab Triple Therapy Study Group.
N Engl J Med.
338
1998
161
165
78
Ludewig
B
Odermatt
B
Ochsenbein
AF
Zinkernagel
RM
Hengartner
H
Role of dendritic cells in the induction and maintenance of autoimmune diseases.
Immunol Rev.
169
1999
45
54
79
Drakesmith
H
Chain
B
Beverley
P
How can dendritic cells cause autoimmune disease?
Immunol Today.
21
2000
214
217
80
Forster
I
Lieberam
I
Peripheral tolerance of CD4 T cells following local activation in adolescent mice.
Eur J Immunol.
26
1996
3194
3202
81
Kurts
C
Kosaka
H
Carbone
FR
Miller
JF
Heath
WR
Class I–restricted cross-presentation of exogenous self-antigens leads to deletion of autoreactive CD8(+) T cells.
J Exp Med.
186
1997
239
245

Author notes

Marc Feldmann, Kennedy Institute of Rheumatology Division, Faculty of Medicine, Imperial College of Science, Technology and Medicine, 1 Aspenlea Rd, Hammersmith, London W6 8LH, United Kingdom; e-mail: m.feldmann@ic.ac.uk.

Sign in via your Institution