Both type I interferons (IFNs) as well as lipopolysaccharide (LPS) individually compromise selected monocytic or dendritic cell (DC) functions. This study investigates the influence of these agents on the differentiation and the regulation of cell death of monocyte-derived DCs generated in the presence of granulocyte-macrophage colony-stimulating factor plus interleukin-4 (IL-4). It is reported that excessive apoptosis occurred rapidly in monocyte-derived DC cultures, if IFN-α or IFN-β was added in combination with LPS or lipoteichoic acid (LTA). The small fraction of cells surviving in such cultures displayed a mature DC phenotype with expression of CD83, CD80, and CD86. IL-10 was found in the supernatants of monocyte-derived DC cultures, if supplemented with LPS or IFN-α plus LPS but not in control cultures. When monocyte-derived DCs were generated in the presence of IFN-α without LPS, these cells displayed an immature DC phenotype with a reduction of cell recovery but no overt apoptosis. However, the addition of LPS, LTA, LPS plus IFN-γ, or tumor necrosis factor α (TNF-α) plus prostaglandin E2 to such cells again resulted in the rapid induction of apoptosis in the majority of cells, together with a reduced production of IL-12 p70 and TNF-α. Together, these data indicate an exquisite sensitivity of monocyte-derived DCs to activation-induced cell death if generated in the presence of IFN-α, indicating the existence of an important mechanism of immunosuppression caused by IFN-α–inducing agents, such as viral or bacterial stimuli.

Evidence accumulating during many years has shown that viral and bacterial infections can impair the function of many cells of the immune system. Dendritic cells (DCs) are of central relevance within the immune system because only these cells can activate naive T cells and thereby initiate an adaptive immune response. Thus, understanding their function during infections might give insight into important immunopathologic mechanisms. For in vitro studies, DCs are commonly derived either from CD34-expressing precursors or from monocytes (reviewed by Banchereau and Steinman1). With regard to monocyte-derived DCs several effects of bacterial and viral stimuli in vitro and in vivo have already been described.

For instance, interferon α (IFN-α), which is transiently released during the first wave of innate immunity in response to bacteria and viruses,2-10 has been reported to suppress interleukin-12 (IL-12) production of monocytes and DCs.11-14 The superfamily of type I IFNs comprises at least 12 IFN-α species and a single IFN-β that differentially bind to a common receptor.15 Alpha IFNs are produced in modest amounts by monocytes, macrophages, and B lymphocytes9 and in much larger quantities by the precursors of lymphoid DCs (pDC2),9,16,17 whereas IFN-β is mainly produced by fibroblasts.18 Despite their established protective role in innate immunity because of their antibacterial and antiviral activity,2,10,19-21 there is increasing evidence that type I IFNs might also act negatively on immunity not only by abrogating IL-12 production but also by impairing DC differentiation22 and reducing the phagocytic and oxidative activity of monocytes.23 

Other studies showed that lipopolysaccharide (LPS) has suppressive effects on monocytes and monocyte-derived DCs,4,10possibly in part mediated by LPS-triggered type I IFN production. A transient presence of endotoxin during the differentiation of monocyte-derived DCs abrogates IL-12 production of the resulting immature DCs on a second stimulation with LPS.24 If permanently present during DC differentiation, it completely desensitizes the cells to all further LPS-mediated maturation signals.25 Monocytes from septic patients were found to be hyporesponsive to LPS stimulation ex vivo, and this hyporesponsiveness was reversed during IFN-γ treatment that also lead to clearance of sepsis,26 suggesting that monocyte deactivation might be a major mechanism of immunosuppression.

Despite these data it is unclear how DCs respond to a combination of type I IFNs and bacterial products. With the use of the culture of monocyte-derived DCs, our study revealed a strong synergistic effect between type I IFNs and products from gram-negative as well as gram-positive bacteria for the induction of apoptosis. This effect occurs if both stimuli are present together during the whole culture but also when the bacterial stimulus is added later to immature DCs generated in the presence of type I IFNs.

Monocyte isolation and DC culture

Human mononuclear cells were obtained from blood containing citrate as anticoagulant from healthy donors by density gradient centrifugation using endotoxin-free Ficoll-Paque-PLUS (Pharmacia, Uppsala, Sweden). Monocytes were isolated from these mononuclear cells by a second density gradient centrifugation (M.L. and W.H., manuscript submitted, March 2001) using Ficoll-Paque-PLUS adjusted to 1.068 g/mL by dilution with phosphate-buffered saline (BioWhittaker, Walkersville, MD). The remaining cells, containing 80% to 90% monocytes as assessed by counting on a Sysmex F820 instrument, were frozen in RPMI 1640 (Life Technologies, Grand Island, NY) supplemented with 10% dimethyl sulfoxide (Sigma, St Louis, MO) and 20% fetal calf serum containing less than 100 pg/mL endotoxin (PAA Laboratories, Linz, Austria). For DC differentiation monocytes were cultured in 24-well culture plates (Iwaki, Japan) at 0.5 × 106/mL in RPMI 1640 supplemented with 10% fetal calf serum, 2 mM L-glutamine (Life Technologies), 1000 U/mL granulocyte-macrophage colony-stimulating factor (GM-CSF; Leucomax; Roche, Basel, Switzerland), and 1000 U/mL recombinant human IL-4 (Strathmann Biotec, Hamburg, Germany). Half of the medium (including all supplements) was exchanged every 2 days. IFN-α2c (Berofor; Boehringer Ingelheim, Vienna, Austria), recombinant human IFN-β (Sigma), LPS fromEscherichia coli 055:B5 (Sigma) and lipoteichoic acid (LTA) from Bacillus subtilis (Sigma) were added in different concentrations and combinations at the indicated time points. Further, some cultures were additionally supplemented with 1000 U/mL recombinant human tumor necrosing factor α (TNF-α; Bender MedSystems, Vienna, Austria), 1 μg/mL prostaglandin E2 (PGE2; Sigma), 1000 U/mL IFN-γ (Bender MedSystems), or 40 000/24-well irradiated (6000 rad) SNJB7 neuroblastoma cells transfected with CD40 ligand (CD40L).46 

Flow cytometric analysis

For immunophenotypic analysis the following monoclonal antibodies were used: anti-CD80–phycoerythrin (PE; clone MAB104; Immunotech, Marseille, France), anti-CD83–PE (clone HB15A; Immunotech), anti-CD86–fluorescein isothiocyanate (FITC; clone 2331; Pharmingen, San Diego, CA), anti-CD1a–FITC (clone HI149; Pharmingen), and anti-CD14–allophycocyanin (clone MoP9; Becton Dickinson, Erembodegem, Belgium). Apoptosis was detected by staining with Annexin V–FITC (Alexis, San Diego, CA) according to the manufacturer's protocol. Cells were analyzed on a FACScan flow cytometer (Becton Dickinson, Mountain View, CA), and data analysis was performed with CellQuest software (Becton Dickinson).

Cytokine measurements

On the basis of standard sandwich enzyme-linked immunosorbent assay methodology, commercially available pairs of monoclonal antibodies (Pharmingen) were used to quantify human IL-12 p70, IL-10, and TNF-α.

Combination of type I IFNs with LPS or LTA induces extensive apoptosis in cultures of monocyte-derived DCs

IFN-α and LPS each are described to affect the differentiation of monocyte-derived DCs. To investigate potential synergistic effects between type I IFNs and bacterial stimuli on monocyte differentiation toward DCs, we first analyzed the effect of IFN-α and LPS present during the whole culture period. IFN-α (1000 U/mL) as well as 1 ng/mL LPS were added on day 0 alone or in combination to monocytes in medium containing IL-4 and GM-CSF. On day 6, light microscopy indicated extensive cell death in the cultures containing both LPS and IFN-α (not shown).

Apoptotic cell death is characterized by the binding of Annexin V and by cell shrinkage (in contrast to necrosis). Figure1 shows the flow-activated cell sorter (FACS) analysis of a DC culture treated with IFN-α plus LPS compared with an untreated culture after staining with Annexin V–FITC and propidium iodide. Figure 1A illustrates that in the control cultures the majority of cells are contained within a relatively homogenous DC population with regard to cell size (estimated by forward scatter [FSC]) and granularity (estimated by the side scatter [SSC]), both parameters are decreased if IFN-α plus LPS had been added to the culture (Figure 1B). Most of the IFN-α–plus–LPS–treated cells acquired Annexin V positivity, indicating the presence of extensive apoptosis, with a subpopulation being also positive for propidium iodide (Figure 1D). Double-positive cells represent cells that have already lost integrity of their membranes during the late apoptotic process. In this representation, however, a clear demarcation of the apoptotic cell population was not possible. For the quantification of apoptosis we, therefore, used the observed reduction in the forward scatter together with increased binding of Annexin V to achieve a sufficient discrimination of apoptotic cells (Figure 1E,F). The same mode of representation was used for the quantification of apoptosis in all further experiments. We could also detect the typical condensation of apoptotic nuclei in cytospins of cells stained with Annexin V and 4,6-diamidino-2-phenylindole (not shown).

Fig. 1.

Quantification of apoptosis by FACS analysis after staining with Annexin V–FITC and propidium iodide.

Density gradient-purified monocytes were cultured for 6 days with GM-CSF plus IL-4 in the absence (A,C,E) or presence (B,D,F) of 1000 U/mL IFN-α plus 1 ng/mL LPS. Shown are the results of a single experiment by 3 alternative modes of representations displaying the forward/side scatter profile (A-B), Annexin V–FITC versus propidium iodide (PI) (C-D), and forward scatter versus Annexin V–FITC (E-F). Gates G1 (apoptotic cells) and G2 (viable cells) were drawn arbitrarily in the representation forward scatter versus Annexin (E-F), because in this representation both populations could best be discriminated. In this experiment cells in gate G1 (apoptotic cells) amounted to 12.2% of total cells in panel E (control culture) and 64.8% in panel F (culture with IFN-α plus LPS), whereas the cells in gate G2 were considered as viable cells (75% of total cells in panel E and 22% in panel F). The small population of lymphocytes (best visible in panel A) amounted to 8.3% of total events in this experiment.

Fig. 1.

Quantification of apoptosis by FACS analysis after staining with Annexin V–FITC and propidium iodide.

Density gradient-purified monocytes were cultured for 6 days with GM-CSF plus IL-4 in the absence (A,C,E) or presence (B,D,F) of 1000 U/mL IFN-α plus 1 ng/mL LPS. Shown are the results of a single experiment by 3 alternative modes of representations displaying the forward/side scatter profile (A-B), Annexin V–FITC versus propidium iodide (PI) (C-D), and forward scatter versus Annexin V–FITC (E-F). Gates G1 (apoptotic cells) and G2 (viable cells) were drawn arbitrarily in the representation forward scatter versus Annexin (E-F), because in this representation both populations could best be discriminated. In this experiment cells in gate G1 (apoptotic cells) amounted to 12.2% of total cells in panel E (control culture) and 64.8% in panel F (culture with IFN-α plus LPS), whereas the cells in gate G2 were considered as viable cells (75% of total cells in panel E and 22% in panel F). The small population of lymphocytes (best visible in panel A) amounted to 8.3% of total events in this experiment.

Close modal

In the following experiments we investigated if the observed synergy in induction of apoptosis also exists between IFN-β as an alternative to type I IFN and LTA, a cell wall component of gram-positive bacteria. Figure 2 compares the fraction of apoptotic cells on day 6 in cultures containing IFN-α or IFN-β and LPS or LTA alone or in combination. In contrast to the effect of the single factors, all combinations of a type I IFN with a bacterial stimulus induced strong apoptosis.

Fig. 2.

Type I IFNs in combination with LPS or LTA induce apoptosis.

Different combinations of 1000 U/mL IFN-α, 1000 U/mL IFN-β, 1 ng/mL LPS, and 1 μg/mL LTA were added on day 0 to cultures supplemented with IL-4 and GM-CSF. The percentage of Annexin-positive cells was determined by FACS analysis on day 6 as detailed in Figure 1. The data were obtained from 4 different donors, each represented by an individual symbol. Two donors (■, ▵) were tested twice in independent experiments.

Fig. 2.

Type I IFNs in combination with LPS or LTA induce apoptosis.

Different combinations of 1000 U/mL IFN-α, 1000 U/mL IFN-β, 1 ng/mL LPS, and 1 μg/mL LTA were added on day 0 to cultures supplemented with IL-4 and GM-CSF. The percentage of Annexin-positive cells was determined by FACS analysis on day 6 as detailed in Figure 1. The data were obtained from 4 different donors, each represented by an individual symbol. Two donors (■, ▵) were tested twice in independent experiments.

Close modal

Titration experiments with IFN-α plus LPS (Figure3A) and IFN-α plus LTA (Figure 3B) revealed strong synergistic effects of 100 to 1000 U/mL IFN-α with both bacterial components at low concentrations, and in kinetic studies it was evident that apoptosis occurred rapidly following the addition of IFN-α plus LPS (Figure 4).

Fig. 3.

Dose dependency of apoptosis in response to IFN-α plus LPS or LTA.

IFN-α and LPS (A) or LTA (B) was added at the indicated concentrations alone (▴) or together with 10 U/mL (●), 100 U/mL (♦), or 1000 U/mL IFN-α (▪) on day 0 to cultures of monocytes supplemented with IL-4 and GM-CSF. The induction of apoptosis was quantified by FACS analysis on day 6 as detailed in Figure1.

Fig. 3.

Dose dependency of apoptosis in response to IFN-α plus LPS or LTA.

IFN-α and LPS (A) or LTA (B) was added at the indicated concentrations alone (▴) or together with 10 U/mL (●), 100 U/mL (♦), or 1000 U/mL IFN-α (▪) on day 0 to cultures of monocytes supplemented with IL-4 and GM-CSF. The induction of apoptosis was quantified by FACS analysis on day 6 as detailed in Figure1.

Close modal
Fig. 4.

Rapid induction of apoptosis in cultures containing IFN-α and LPS.

On day 0, 1000 U/mL IFN-α ± 1 ng/mL LPS was added to monocyte cultures supplemented with GM-CSF plus IL-4. Annexin V positivity was quantified on days 2, 4, and 6. ■, control; ░, IFN-α; ▪, IFN-α plus LPS. Shown are the data obtained with monocytes from 3 different donors (mean ± SD).

Fig. 4.

Rapid induction of apoptosis in cultures containing IFN-α and LPS.

On day 0, 1000 U/mL IFN-α ± 1 ng/mL LPS was added to monocyte cultures supplemented with GM-CSF plus IL-4. Annexin V positivity was quantified on days 2, 4, and 6. ■, control; ░, IFN-α; ▪, IFN-α plus LPS. Shown are the data obtained with monocytes from 3 different donors (mean ± SD).

Close modal

Surviving cells in cultures containing IFN-α plus LPS are phenotypically mature DCs

The extensive apoptosis in the presence of IFN-α and LPS strongly reduced the yield of viable cells. The phenotype of the remaining cells was compared with that of control cultures of immature DCs differentiated in the absence of IFN-α plus LPS and with cells derived from such control cultures but matured by the addition of LPS for the last 3 days of the culture. FACS analysis for selected DC markers for all cultures was done on day 8 (Figure5A). The permanent presence of IFN-α plus LPS resulted in an increased expression of CD83, CD80, and CD86 in the surviving cells, indicating the presence of mature DCs. These increases were not seen in the presence of IFN-α or LPS alone (not shown). When these cells were additionally exposed to the strong maturation stimulus IFN-γ plus CD40L from day 5 on, there was almost no further response (Figure 5B).

Fig. 5.

Surviving DCs generated in the presence of IFN-α plus LPS have a mature phenotype.

Monocyte-derived DCs were generated with GM-CSF plus IL-4 in the absence (control cultures, dotted lines, A) or presence of IFN-α plus LPS (bold lines, A-B). During the last 3 days, a part of both cultures was subjected to a maturation step employing 100 ng/mL LPS (control cultures, thin line, A) or CD40L plus IFN-γ (cultures containing IFN-α plus LPS, thin line, B). Cells were stained on day 8 of total culture for the presence of CD83, CD80, and CD86; only large viable cells were analyzed.

Fig. 5.

Surviving DCs generated in the presence of IFN-α plus LPS have a mature phenotype.

Monocyte-derived DCs were generated with GM-CSF plus IL-4 in the absence (control cultures, dotted lines, A) or presence of IFN-α plus LPS (bold lines, A-B). During the last 3 days, a part of both cultures was subjected to a maturation step employing 100 ng/mL LPS (control cultures, thin line, A) or CD40L plus IFN-γ (cultures containing IFN-α plus LPS, thin line, B). Cells were stained on day 8 of total culture for the presence of CD83, CD80, and CD86; only large viable cells were analyzed.

Close modal

Although the remaining viable cells of the cultures containing IFN-α and LPS thus resembled mature DCs, there were peculiarities. Despite a homogeneous expression of CD83, CD80, and CD86 (Figure 5A), further analysis revealed CD14 expression in a subpopulation of cells (Figure6A). To some extent, varying between individuals, some CD14 expression was also seen with IFN-α alone. Because IL-10 was described to induce differentiation of immature monocyte-derived DCs into CD14+ macrophagelike cells,27 we tested if the observed CD14 expression was correlated with elevated IL-10 production. In the cultures containing both IFN-α and LPS approximately 2 ng/mL IL-10 was found on day 4 (Figure 6B). Although there was no CD14 expression at all with LPS alone, it also induced some IL-10 (300 pg/mL) as previously reported,28 whereas IFN-α alone induced some CD14 but not any IL-10.

Fig. 6.

CD14 expression and IL-10 production of cells differentiated in presence of IFN-α plus LPS.

The dot plots in panel A were obtained by FACS analysis on day 6 from cultures containing 1000 U/mL IFN-α and/or 1 ng/mL LPS as indicated. Despite homogeneous expression of the DC maturation markers CD83, CD80, and CD86 (not displayed), the expression of CD14 is retained in a subpopulation of cells cultured in the presence of IFN-α plus LPS. Shown are the data from one representative experiment; similar results were obtained in 3 further experiments. Panel B displays the amount of IL-10 found in the supernatants on day 4 (mean ± SD of 3 independent experiments).

Fig. 6.

CD14 expression and IL-10 production of cells differentiated in presence of IFN-α plus LPS.

The dot plots in panel A were obtained by FACS analysis on day 6 from cultures containing 1000 U/mL IFN-α and/or 1 ng/mL LPS as indicated. Despite homogeneous expression of the DC maturation markers CD83, CD80, and CD86 (not displayed), the expression of CD14 is retained in a subpopulation of cells cultured in the presence of IFN-α plus LPS. Shown are the data from one representative experiment; similar results were obtained in 3 further experiments. Panel B displays the amount of IL-10 found in the supernatants on day 4 (mean ± SD of 3 independent experiments).

Close modal

Addition of IFN-α plus LPS to immature DCs induces maturation without apoptosis

In the experiments described above, type I IFNs and bacterial stimuli were added to monocytes on day 0. To investigate the response of immature DCs to a combination of these stimuli, immature DCs were challenged with LPS or IFN-α alone or with a combination of both. LPS plus IFN-α as well as LPS alone triggered maturation with induction of CD83 (not shown) and a very high expression of CD80 and CD86 3 days after stimulation (Figure 7A). IFN-α alone did not induce maturation. Importantly, no increased apoptosis was observed in the cultures containing IFN-α plus LPS (Figure 7B).

Fig. 7.

The addition of IFN-α plus LPS to immature DCs on day 7 triggers maturation without apoptosis.

Panel A shows the phenotype of monocyte-derived DCs on day 10 (ie, 3 days after addition of the indicated stimuli). The percentage of apoptotic cells on day 10 is shown in panel B (mean ± SD of 3 independent experiments).

Fig. 7.

The addition of IFN-α plus LPS to immature DCs on day 7 triggers maturation without apoptosis.

Panel A shows the phenotype of monocyte-derived DCs on day 10 (ie, 3 days after addition of the indicated stimuli). The percentage of apoptotic cells on day 10 is shown in panel B (mean ± SD of 3 independent experiments).

Close modal

Immature DCs generated in the presence of IFN-α undergo apoptosis in response to a variety of activation stimuli

The above experiments had investigated the effects of type I IFNs and bacterial stimuli when added together at the start of the culture to monocytes or to monocyte-derived immature DCs. Next we asked whether bacterial products or other activating signals would induce apoptosis if added later to immature DCs, which had been differentiated in the presence of IFN-α. Therefore, immature DCs generated in the presence or absence of 1000 U/mL IFN-α were stimulated with LTA, LPS, LPS plus IFN-γ, and TNF-α plus PGE2 on day 5. Three days later, extensive apoptosis was present in all cultures that had been differentiated in the presence of IFN-α (Figure 8A). Importantly, apoptosis was induced to the same extent by activation of the IFN-α precultured DCs with TNF-α plus PGE2 as with LPS or LTA, suggesting that IFN-α had sensitized the immature DCs to activation-induced cell death in general. To some extent 1000 U/mL IFN-α alone also increased apoptosis on day 8, but addition of the maturation stimuli dramatically raised the percentage of Annexin V–binding cells. The expression of CD80, CD86, and CD83 of the surviving cells in these cultures was completely identical to that of the respective activated DCs differentiated without IFN-α (data not shown). Although quantitatively reduced, IFN-α primed immature DCs were also competent for production of IL-12 p70 and TNF-α in response to LPS plus IFN-γ, whereas IL-10 was not produced (Figure9).

Fig. 8.

Immature DCs generated by GM-CSF plus IL-4 in the presence of IFN-α undergo apoptosis in response to a variety of activation stimuli.

The response of immature DCs generated in the permanent presence of 1000 U/mL IFN-α (▪) to the indicated stimuli was compared with that of immature DCs generated in the absence of IFN-α (░). Apoptosis was quantified on day 8 (ie, 3 days after activation with 1 μg/mL LTA, 100 ng/mL LPS, 100 ng/mL LPS plus 1000 U/mL IFN-γ, and 1000 U/mL TNF-α plus 1 μg/mL PGE2 as indicated [mean ± SD of 3 independent experiments]).

Fig. 8.

Immature DCs generated by GM-CSF plus IL-4 in the presence of IFN-α undergo apoptosis in response to a variety of activation stimuli.

The response of immature DCs generated in the permanent presence of 1000 U/mL IFN-α (▪) to the indicated stimuli was compared with that of immature DCs generated in the absence of IFN-α (░). Apoptosis was quantified on day 8 (ie, 3 days after activation with 1 μg/mL LTA, 100 ng/mL LPS, 100 ng/mL LPS plus 1000 U/mL IFN-γ, and 1000 U/mL TNF-α plus 1 μg/mL PGE2 as indicated [mean ± SD of 3 independent experiments]).

Close modal
Fig. 9.

Cytokine production of DCs generated in the presence of IFN-α in response to LPS plus IFN-γ.

The production of IL-12 p70, TNF-α, and IL-10 was determined on day 8 (ie, 3 days after activation with 100 ng/mL LPS plus 1000 U/mL IFN-γ). The response of immature DCs generated in the permanent presence of 1000 U/mL IFN-α (▪) was compared with that of immature DCs generated in the absence of IFN-α (░) (mean ± SD of 3 independent experiments).

Fig. 9.

Cytokine production of DCs generated in the presence of IFN-α in response to LPS plus IFN-γ.

The production of IL-12 p70, TNF-α, and IL-10 was determined on day 8 (ie, 3 days after activation with 100 ng/mL LPS plus 1000 U/mL IFN-γ). The response of immature DCs generated in the permanent presence of 1000 U/mL IFN-α (▪) was compared with that of immature DCs generated in the absence of IFN-α (░) (mean ± SD of 3 independent experiments).

Close modal

In this study we describe the effects of the combined action of type I IFNs and microbial products on the differentiation and survival of monocyte-derived DCs. We found that IL-4/GM-CSF–stimulated cultures of monocytes differentiating toward DCs in the presence of IFN-α or IFN-β underwent strong apoptosis if microbial products such as LPS or LTA were additionally added. Provided that type I IFNs were present from the initiation of the cultures, bacterial products also induced apoptosis when added later to immature DCs. Immature DCs differentiated in the presence of IFN-α died also in response to TNF-α plus PGE2. No apoptosis was seen, however, when type I IFNs alone or together with a bacterial stimulus were added to already differentiated immature DCs. Cells surviving IFN-α plus LPS treatment developed into cells displaying a mature DC phenotype with an expression of CD83, CD80, and CD86 identical to that displayed by normal mature DCs, grown in the absence of IFN-α and further matured by an appropriate stimulus.

Our results fit well with a recent report,22 in which a reduced yield of monocyte-derived DCs grown in the presence of type I IFNs was described. We show that this reduction is due to apoptosis and is dramatically enhanced by the addition of a bacterial stimulus even at a low dose (30-100 pg/mL LPS). The kinetics of the apoptosis induction through the combined action of IFN-α and LPS was very rapid. Only 2 days after culture initiation a predominant fraction of cells underwent apoptosis. A similar synergy has been reported for the bovine system, in which macrophages rapidly undergo LPS-induced apoptosis after priming with IFN-α.29 Interestingly, however, murine macrophages go in apoptosis in response to LPS plus IFN-γ but are protected by pretreatment with IFN-α or β.30 

Somehow contrasting the detrimental effects of type I IFNs on DC differentiation seen in our study, 2 recent reports suggested IFN-α as an adjuvant supporting DC development with GM-CSF in the absence of IL-4.31,32 Interestingly, Santini et al31described also an increase of Annexin binding cells in their cultures containing IFN-α plus GM-CSF, although in their study a beneficial effect of IFN-α on the generation and maturation of DCs prevailed. In our system it was mainly the addition of a second stimulus, such as LPS, LTA, or TNF-α plus PGE2, that resulted in the apoptosis of the majority of newly generated DCs. Our data thus imply that such IFN-α–pretreated immature DCs in general are sensitized to activation-induced cell death.

The phenotype of the surviving cells differed depending on the time point of the addition of the bacterial stimulus. In the presence of IFN-α, GM-CSF, and IL-4, the cells developed into viable CD14 and CD1a+ immature DCs (Figure 6A). Addition of bacterial stimuli to the immature DCs precultured with IFN-α induced strong apoptosis, but the surviving cells expressed CD83, CD80, and CD86 completely identically to mature control DCs (Figure 8 and not shown). In the cultures in which both stimuli were present from the initiation of the culture, the few viable cells also displayed a mature DC phenotype without further activation (Figure 5A) and hardly responded to further stimulation with IFN-γ plus CD40L (Figure 5B). These cells also stimulated allogeneic T cells in a mixed lymphocyte reaction in accordance with their mature DC phenotype (not shown).

The mature population resulting from the early presence of IFN-α plus LPS further contained a varying but substantial fraction of CD14+ cells (Figure 6A). Because IL-10 was described to shift differentiation of DC cultures toward macrophagelike cells by raising the expression of the receptor for the endogenously produced macrophage colony-stimulating factor,27 we analyzed the culture supernatants for the presence of IL-10. In accordance with earlier reports,28 33 we found little amounts of IL-10 in cultures containing a low dose of LPS and markedly raised amounts in cultures supplemented with LPS plus IFN-α (Figure 6B). Because the addition of IL-10 to cultures treated with IFN-α alone strongly increased CD14 expression and a neutralizing anti–IL-10 monoclonal antibody added to IFN-α plus LPS-stimulated cultures decreased CD14 expression (data not shown), endogenously produced IL-10 indeed appeared to influence the phenotype of the cells in our system.

With regard to the underlying mechanism of apoptosis induction, endogenously produced nitric oxide (NO) could be a possible candidate mediating cell death. In murine macrophages NO is induced by LPS in synergy with a type I IFN or IFN-γ,34 and in murine dendritic cells NO production in response to LPS plus IFN-γ was reported to cause apoptosis.35 In bovine macrophages IFN-α could prime for LPS-induced apoptosis, but this effect did not correlate with enhanced NO production, instead LPS-induced NO production was strongly reduced by IFN-α.29 In the human system controversy exists as to whether and under which conditions monocytic cells can be activated for increased NO production.36-39 Sharara et al38 found a modestly increased NO production of human monocytes treated with IFN-α, and Weinberg et al39 reported that a basal NO production by human monocytes could not be increased by treatment with LPS plus IFN-γ. Together these published data suggest that significant NO production by human DCs is unlikely to occur. In accordance with such an interpretation of the published literature, we found no inhibition of IFN-α plus LPS induced apoptosis by the NO synthase inhibitor L-NMMA (NG-monomethyl-L-arginine, monoacetate) (data not shown).

Apart from being induced directly in the DCs, alternatively, apoptosis might be mediated indirectly, for instance, through the action of T cells expressing Fas ligand. Both sensitivity as well as resistance of human monocyte-derived DCs to Fas-mediated apoptosis has been described.40-42 In preliminary experiments, cross-linked Fas ligand neither induced nor a blocking CD95 Fas antibody reduced apoptosis in our system (not shown). Most of our DC cell preparations based on density gradient centrifugation purified monocytes contained between 5% and 15% T cells. However, further purification and depletion of these T cells by FACS sorting did not have any influence on the apoptosis induction in response to IFN-α plus LPS (not shown), making an involvement of contaminating T cells unlikely.

Regardless of the intracellular mechanism responsible for the observed apoptosis, what is the biological relevance of our finding? It is conceivable that bacterial products and sufficiently high concentrations of type I IFNs could coincide during bacterial infections, because bacteria and their products have been shown to induce type I IFNs in monocyte and macrophage cells at least under certain circumstances.2-4,10 Although LPS directly induces IFN-α in GM-CSF–primed human monocytes4 and human DCs,10 we have not seen marked apoptosis in response to LPS alone nor has such a phenomenon been described to our knowledge. This phenomenon could be due to a requirement of IFN priming before LPS can subsequently induce apoptosis, which would not occur if the type I IFN accumulation is delayed during culture. Indeed, such a critical timing of both stimuli was described with bovine macrophages.29 However, in vivo LPS-induced IFN-α could make other monocytic cells or DCs not yet in contact with the LPS susceptible to subsequent activation-induced cell death. Monocytes from septic patients might be primed by type I IFNs by such a mechanism and indeed have been reported to display a predilection to undergo apoptosis in response to LPS.43 

Type I IFNs are produced in large amounts by the rare type 2 DCs9,16,17 and at much lower levels by other more frequent cells such as monocytes or B cells.9 Usually, this synthesis of type I IFN by activated cells appears to be only transient,44 resulting in elevated type I IFN serum levels in acute but not in chronic viral infections.5-7 Thus, cell death by coincidence of IFN-α priming and bacterial activation seems less likely in chronic viral infections but still could occur, for instance in bacterially colonized tissues (ie, the upper respiratory and the gastrointestinal tract during an acute viral illness).

Apart from infectious situations, a prolonged and more systemic presence of type I IFNs might also be achieved during treatment with type I IFNs, as currently employed in hepatitis C, multiple sclerosis, or selected malignancies. This treatment could result in the permanent priming of monocytes and immature DCs for activation-induced apoptosis in response to any infection or other activating stimuli. Interestingly, a transient decrease in monocytes has been reported after IFN-α application to healthy humans.45 

IFN-α and endotoxin have already been individually known to influence the function of monocytes and derived DCs. Our study reveals that both agents in combination are potent inducers of apoptosis in these cells, pointing to a new and potentially relevant mechanism of immunosuppression in infectious disease.

We thank Dr G. Mehes (CCRI, Vienna, Austria) for technical assistance in fluorescence microscopy and Dr H. Kovar (CCRI) for expert advice in detection of apoptosis.

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
Diefenbach
A
Schindler
H
Donhauser
N
et al
Type 1 interferon (IFNalpha/beta) and type 2 nitric oxide synthase regulate the innate immune response to a protozoan parasite.
Immunity.
8
1998
77
87
3
Helfgott
DC
May
LT
Sthoeger
Z
Tamm
I
Sehgal
PB
Bacterial lipopolysaccharide (endotoxin) enhances expression and secretion of beta 2 interferon by human fibroblasts.
J Exp Med.
166
1987
1300
1309
4
Hayes
MP
Enterline
JC
Gerrard
TL
Zoon
KC
Regulation of interferon production by human monocytes: requirements for priming for lipopolysaccharide-induced production.
J Leukoc Biol.
50
1991
176
181
5
Zachoval
R
Abb
J
Zachoval
V
Deinhardt
F
Circulating interferon in patients with acute hepatitis A.
J Infect Dis.
153
1986
1174
1175
6
Zachoval
R
Abb
J
Zachoval
V
et al
Interferon alpha in hepatitis type B and non-A, non-B. Defective production by peripheral blood mononuclear cells in chronic infection and development of serum interferon in acute disease.
J Hepatol.
6
1988
364
368
7
von Sydow
M
Sonnerborg
A
Gaines
H
Strannegard
O
Interferon-alpha and tumor necrosis factor-alpha in serum of patients in various stages of HIV-1 infection.
AIDS Res Hum Retroviruses.
7
1991
375
380
8
Feldman
SB
Ferraro
M
Zheng
HM
et al
Viral induction of low frequency interferon-alpha producing cells.
Virology.
204
1994
1
7
9
Ferbas
JJ
Toso
JF
Logar
AJ
Navratil
JS
Rinaldo
CRJ
CD4+ blood dendritic cells are potent producers of IFN-alpha in response to in vitro HIV-1 infection.
J Immunol.
152
1994
4649
4662
10
Cella
M
Salio
M
Sakakibara
Y
et al
Maturation, activation, and protection of dendritic cells induced by double-stranded RNA.
J Exp Med.
189
1999
821
829
11
Karp
CL
Biron
CA
Irani
DN
Interferon beta in multiple sclerosis: is IL-12 suppression the key?
Immunol Today.
21
2000
24
28
12
McRae
BL
Semnani
RT
Hayes
MP
van Seventer
GA
Type I IFNs inhibit human dendritic cell IL-12 production and Th1 cell development.
J Immunol.
160
1998
4298
4304
13
Cousens
LP
Orange
JS
Su
HC
Biron
CA
Interferon-alpha/beta inhibition of interleukin 12 and interferon-gamma production in vitro and endogenously during viral infection.
Proc Natl Acad Sci U S A.
94
1997
634
639
14
Chehimi
J
Starr
SE
Frank
I
et al
Impaired interleukin 12 production in human immunodeficiency virus-infected patients.
J Exp Med.
179
1994
1361
1366
15
Lewerenz
M
Mogensen
KE
Uze
G
Shared receptor components but distinct complexes for alpha and beta interferons.
J Mol Biol.
282
1998
585
599
16
Siegal
FP
Kadowaki
N
Shodell
M
et al
The nature of the principal type 1 interferon-producing cells in human blood.
Science.
284
1999
1835
1837
17
Cella
M
Jarrossay
D
Facchetti
F
et al
Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon.
Nat Med.
5
1999
919
923
18
Sehgal
PB
Sagar
AD
Heterogeneity of poly(I) x poly(C)-induced human fibroblast interferon mRNA species.
Nature.
288
1980
95
97
19
Gribaudo
G
Ravaglia
S
Caliendo
A
et al
Interferons inhibit onset of murine cytomegalovirus immediate-early gene transcription.
Virology.
197
1993
303
311
20
Yeow
WS
Lawson
CM
Beilharz
MW
Antiviral activities of individual murine IFN-alpha subtypes in vivo: intramuscular injection of IFN expression constructs reduces cytomegalovirus replication.
J Immunol.
160
1998
2932
2939
21
Maitra
RK
Silverman
RH
Regulation of human immunodeficiency virus replication by 2′,5′-oligoadenylate-dependent RNase L.
J Virol.
72
1998
1146
1152
22
McRae
BL
Nagai
T
Semnani
RT
van Seventer
JM
van Seventer
GA
Interferon-alpha and -beta inhibit the in vitro differentiation of immunocompetent human dendritic cells from CD14(+) precursors.
Blood.
96
2000
210
217
23
Noraz
N
Lathey
JL
Spector
SA
Human cytomegalovirus-associated immunosuppression is mediated through interferon-alpha.
Blood.
89
1997
2443
2452
24
Karp
CL
Wysocka
M
Ma
X
et al
Potent suppression of IL-12 production from monocytes and dendritic cells during endotoxin tolerance.
Eur J Immunol.
28
1998
3128
3136
25
Rieser
C
Papesh
C
Herold
M
et al
Differential deactivation of human dendritic cells by endotoxin desensitization: role of tumor necrosis factor-alpha and prostaglandin E2.
Blood.
91
1998
3112
3117
26
Docke
WD
Randow
F
Syrbe
U
et al
Monocyte deactivation in septic patients: restoration by IFN-gamma treatment.
Nat Med.
3
1997
678
681
27
Rieser
C
Ramoner
R
Bock
G
et al
Human monocyte-derived dendritic cells produce macrophage colony-stimulating factor: enhancement of c-fms expression by interleukin-10.
Eur J Immunol.
28
1998
2283
2288
28
de Waal
M
Abrams
J
Bennett
B
Figdor
CG
de Vries
JE
Interleukin 10 (IL-10) inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL-10 produced by monocytes.
J Exp Med.
174
1991
1209
1220
29
Adler
B
Adler
H
Jungi
TW
Peterhans
E
Interferon-alpha primes macrophages for lipopolysaccharide-induced apoptosis.
Biochem Biophys Res Commun.
215
1995
921
927
30
Lopez-Collazo
E
Hortelano
S
Bosca
L
Interferon-alpha/beta inhibits the apoptosis induced by lipopolysaccharide and interferon-gamma in murine peritoneal macrophages.
J Interferon Cytokine Res.
18
1998
461
467
31
Santini
SM
Lapenta
C
Logozzi
M
et al
Type I interferon as a powerful adjuvant for monocyte-derived dendritic cell development and activity in vitro and in Hu-PBL-SCID mice.
J Exp Med.
191
2000
1777
1788
32
Paquette
RL
Hsu
NC
Kiertscher
SM
et al
Interferon-alpha and granulocyte-macrophage colony-stimulating factor differentiate peripheral blood monocytes into potent antigen-presenting cells.
J Leukoc Biol.
64
1998
358
367
33
Aman
MJ
Tretter
T
Eisenbeis
I
et al
Interferon-alpha stimulates production of interleukin-10 in activated CD4+ T cells and monocytes.
Blood.
87
1996
4731
4736
34
Ding
AH
Nathan
CF
Stuehr
DJ
Release of reactive nitrogen intermediates and reactive oxygen intermediates from mouse peritoneal macrophages. Comparison of activating cytokines and evidence for independent production.
J Immunol.
141
1988
2407
2412
35
Lu
L
Bonham
CA
Chambers
FG
et al
Induction of nitric oxide synthase in mouse dendritic cells by IFN-gamma, endotoxin, and interaction with allogeneic T cells: nitric oxide production is associated with dendritic cell apoptosis.
J Immunol.
157
1996
3577
3586
36
Denis
M
Human monocytes/macrophages: NO or no NO?
J Leukoc Biol.
55
1994
682
684
37
Zhang
X
Laubach
VE
Alley
EW
et al
Transcriptional basis for hyporesponsiveness of the human inducible nitric oxide synthase gene to lipopolysaccharide/interferon-gamma.
J Leukoc Biol.
59
1996
575
585
38
Sharara
AI
Perkins
DJ
Misukonis
MA
et al
Interferon (IFN)-alpha activation of human blood mononuclear cells in vitro and in vivo for nitric oxide synthase (NOS) type 2 mRNA and protein expression: possible relationship of induced NOS2 to the anti-hepatitis C effects of IFN-alpha in vivo.
J Exp Med.
186
1997
1495
1502
39
Weinberg
JB
Misukonis
MA
Shami
PJ
et al
Human mononuclear phagocyte inducible nitric oxide synthase (iNOS): analysis of iNOS mRNA, iNOS protein, biopterin, and nitric oxide production by blood monocytes and peritoneal macrophages.
Blood.
86
1995
1184
1195
40
Willems
F
Amraoui
Z
Vanderheyde
N
et al
Expression of c-FLIP(L) and resistance to CD95-mediated apoptosis of monocyte-derived dendritic cells: inhibition by bisindolylmaleimide.
Blood.
95
2000
3478
3482
41
Servet-Delprat
C
Vidalain
PO
Azocar
O
et al
Consequences of Fas-mediated human dendritic cell apoptosis induced by measles virus.
J Virol.
74
2000
4387
4393
42
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
43
Williams
MA
Withington
S
Newland
AC
Kelsey
SM
Monocyte anergy in septic shock is associated with a predilection to apoptosis and is reversed by granulocyte-macrophage colony-stimulating factor ex vivo.
J Infect Dis.
178
1998
1421
1433
44
Hayes
MP
Zoon
KC
Priming of human monocytes for enhanced lipopolysaccharide responses: expression of alpha interferon, interferon regulatory factors, and tumor necrosis factor.
Infect Immun.
61
1993
3222
3227
45
Corssmit
EP
Heijligenberg
R
Hack
CE
et al
Effects of interferon-alpha (IFN-alpha) administration on leucocytes in healthy humans.
Clin Exp Immunol.
107
1997
359
363
46
Felzmann T, Buchberger M, Lehner M, et al. Functional maturation of dendritic cells by exposure to CD40L transgenic tumor cells, fibroblasts or keratinocytes. Cancer Lett. In press.

Author notes

Wolfgang Holter, Children's Cancer Research Institute, Kinderspitalgasse 6, A-1090, Vienna, Austria; e-mail:holter@ccri.univie.ac.at.

Sign in via your Institution