On recognition of influenza virus (Flu) by TLR7, plasmacytoid dendritic cells (pDCs) produce type I IFN in significant amounts. Synthetic TLR7 ligands induce the maturation of pDCs, as evidenced by the expression of costimulatory molecules and the production of proinflammatory cytokines; however, they induce only low-level production of IFN-α. To dissect the TLR7 signaling in pDCs and how these different profiles are induced, we studied the effects of 2 TLR7 ligands (Flu and CL097) on the activation of blood-isolated pDCs and the human GEN2.2 pDC cell line. Type I IFN production by pDCs correlates with differential interferon regulatory factor 7 (IRF7) translocation into the nucleus induced by the 2 TLR7 ligands. Surprisingly, with both activators we nevertheless observed the rapid expression of the IFN-inducible genes mxa, cxcl10, and trail within 4 hours of stimulation. This expression, controlled by STAT1 phosphorylation, was independent of type I IFN. STAT1 activation was found to be strictly dependent on the PI3K-p38MAPK pathway, showing a new signaling pathway leading to rapid expression of IFN-inducible genes after TLR7 triggering. Thus, pDCs, through this unusual TLR7 signaling, have the capacity to promptly respond to viral infection during the early phases of the innate immune response.

Toll-like receptors (TLRs) contain a leucine-rich repeat ectodomain that enables the recognition of pathogen-associated molecular patterns.1  Among the 10 TLRs identified in humans, TLR7 is the least studied. This TLR binds single-stranded viral RNA, localizes to endosomal compartments, and is particularly expressed by plasmacytoid dendritic cells (pDCs).2  Ligand binding to TLR7 activates human pDCs that respond by either producing proinflammatory cytokines or substantial levels of type I IFN and activating specific T cells.3,5  In addition, we have recently shown that Flu- and TLR7 agonist–activated pDCs express TNF-related apoptosis-inducing ligand (TRAIL). This renders them capable of direct cytotoxic activity toward infected and tumor cells.6  This has been observed in vivo: Stary et al7  describe the presence of TRAIL-expressing and IFN-α–producing pDCs in tumors after topical use of the TLR7 ligand imiquimod in basal cell carcinoma treatment. Taken together, these data suggest that pDCs represent key target cells in the striking antitumor effects observed with synthetic TLR7 agonists, such as imidazoquinolines, in the treatment of cutaneous virus–induced neoplasia (eg, condyloma-HPV),8  and other skin tumors, such as basal cell carcinoma.9 

The signaling pathways triggered by TLR7 activation have been partially described. After ligand recognition, endosomal TLR7 interacts with the key adaptor molecule MyD88 (myeloid differentiation primary-response gene 88) that recruits a signal complex comprising IRAK1 (interleukin-1 receptor-associated kinase 1), IRAK4, and TRAF6 (tumor necrosis factor receptor–associated factor 6). TLR7 triggering leads to the activation of NF-κB (nuclear factor-κB) and MAPKs (mitogen-activated protein kinases), inducing the expression of proinflammatory cytokines and costimulatory molecules and the translocation of IRF7 (interferon-regulatory factor 7) into the nucleus, where it can induce the transcription of type I IFN genes.10  Agonistic engagement of TLR7 also leads to the expression of the IFN-inducible genes mxa, cxcl10, and trail.11  A type I IFN autocrine loop is thought to explain the expression of these genes, as has been described in mouse bone marrow–derived DCs.12  However, the precise pathway downstream of TLR7 leading to human pDC activation remains to be determined.

In this study we analyzed the activation profile and signaling events triggered by 2 TLR7 ligands: influenza virus (Flu; natural ligand) and CL097 (synthetic agonist). We studied the effects of these 2 ligands on both blood-isolated pDCs and the pDC model cell line GEN2.2. Results of these studies show the existence of a novel pathway downstream of TLR7 involving early STAT1 phosphorylation and expression of IFN-inducible genes in the absence of type I IFN.

Antibodies, flow cytometry

Surface or intracellular phenotype was determined by flow cytometry on a FACSCanto II (Becton Dickinson), using specific antibodies, by direct or indirect labeling. The following antibodies were from Immunotech (Beckman Coulter): PE-conjugated anti-CD40 (mAb89), PE-conjugated anti-CD80 (mAb104), and PE-conjugated goat anti–mouse IgG (H+L). PE-conjugated anti-CD123 (9F5) was purchased from PharMingen. PE-conjugated antiphospho-STAT1 (4a/ pY701-stat1) was from BD (Becton Dickinson). FITC-conjugated anti–BDCA-2 (AC144) and FITC-conjugated anti–IFN-α (LT27:295) were from Miltenyi Biotec, and purified anti-TRAIL (2E5) from was from Alexis.

Cells

Normal pDCs were isolated from peripheral blood mononuclear cells (PBMCs) of healthy volunteers with a BDCA-4 cell isolation kit (Miltenyi Biotec). Their purity, as determined with anti–BDCA-2 and anti-CD123 mAbs, was approximately 80% (Figure 1A).

Figure 1

Influenza virus and CL097 induce human pDC activation by triggering TLR7. (A, left) Flow cytometric evaluation of purity of unstimulated pDCs enriched from healthy donor blood labeled with anti-BDCA2 and anti-CD123 antibodies. Cells were untreated (medium) or stimulated with UV-formol–inactivated influenza virus (Flu) or synthetic TLR7 ligand (CL097). Expression of CD40 was measured by flow cytometry on CD123+ cells. Dot plots are shown, the percentage of CD40+ cells is indicated on each plot. Results shown are representative of 4 independent experiments. (B) GEN2.2 cells were untreated or stimulated with Flu or CL097 for 24 hours. CD40 expression was evaluated on forward scatter (FSC)/side scatter (SSC)–gated live cells by flow cytometry. Percentages indicated on dot correspond to the proportion of CD40+ cells. Results shown are representative of at least 5 independent experiments. (C) Expression levels of TLR7 and TLR9 in the GEN2.2 cell line and in lentiviral shRNA TLR7-transfected GEN2.2 cells (GENshTLR7) measured by real-time PCR. Expression levels are normalized to G6PDH. Data are shown as the mean and SD from duplicate values of 2 independent experiments. (D) GEN2.2 and GENshTLR7 cells were untreated or stimulated with Flu, CL097, or 2 different synthetic TLR9 ligands (CpG A and CpG B) for 24 hours. Expression of CD40 was measured by flow cytometry. The mean percentages and SDs from duplicate values of 3 independent experiments are shown.

Figure 1

Influenza virus and CL097 induce human pDC activation by triggering TLR7. (A, left) Flow cytometric evaluation of purity of unstimulated pDCs enriched from healthy donor blood labeled with anti-BDCA2 and anti-CD123 antibodies. Cells were untreated (medium) or stimulated with UV-formol–inactivated influenza virus (Flu) or synthetic TLR7 ligand (CL097). Expression of CD40 was measured by flow cytometry on CD123+ cells. Dot plots are shown, the percentage of CD40+ cells is indicated on each plot. Results shown are representative of 4 independent experiments. (B) GEN2.2 cells were untreated or stimulated with Flu or CL097 for 24 hours. CD40 expression was evaluated on forward scatter (FSC)/side scatter (SSC)–gated live cells by flow cytometry. Percentages indicated on dot correspond to the proportion of CD40+ cells. Results shown are representative of at least 5 independent experiments. (C) Expression levels of TLR7 and TLR9 in the GEN2.2 cell line and in lentiviral shRNA TLR7-transfected GEN2.2 cells (GENshTLR7) measured by real-time PCR. Expression levels are normalized to G6PDH. Data are shown as the mean and SD from duplicate values of 2 independent experiments. (D) GEN2.2 and GENshTLR7 cells were untreated or stimulated with Flu, CL097, or 2 different synthetic TLR9 ligands (CpG A and CpG B) for 24 hours. Expression of CD40 was measured by flow cytometry. The mean percentages and SDs from duplicate values of 3 independent experiments are shown.

Close modal

The pDC cell line GEN2.2 was grown in complete medium (RPMI 1640 Glutamax; GibcoBRL) supplemented with 1 mM sodium pyruvate, 20 μg/mL gentamicin, nonessential amino acids) to which 10% heat-inactivated fetal calf serum (Gibco) was added.

Generation of lentiviral shRNA TLR7-transfected GEN2.2 cells

GEN2.2 cells were transfected with MISSION Lentiviral transduction particles targeting TLR7 (NM_016562) Clone ID: TRCN0000056975 (Sigma-Aldrich) at MOI 2. Transfected cells were maintained in the presence of 10 μg/mL puromycin for 2 weeks to allow the selection of resistant clones that we called GENshTLR7 cells. The level of TLR7 mRNA expression in GENshTLR7 cells was assessed by real-time polymerase chain reaction (PCR). Silencing was found to diminish TLR7 mRNA expression by 80%.

Activation of GEN2.2 cells

Cells were cultured at 106 cells/mL in complete medium with 10% fetal calf serum. Cells were stimulated with either 640 UHA/mL UV-formol–inactivated influenza virus strain A/H3N2/Wisconsin/67/05 (Sanofi Pasteur) or 1 μg/mL CL097 (TLR7/8 ligand; Invivogen) or 10 μg/mL CpG-A ODN 2336 (TLR9 ligand; Coley Pharmaceuticals) or 10 μg/mL CpG-B ODN 2216 (TLR9 ligand; Invivogen) or 50 000 U/mL human recombinant IFN-α (PeproTech). For some experiments, blocking anti–IFN-α (50 000 U/mL; PBL Medical Laboratories), anti–IFN-β (25 000 U/mL; PBL Medical Laboratories), and anti–IFN-α/βR2 (5 μg/mL; PBL Medical Laboratories) or inhibitors from Calbiochem were added: 5 μM BAY11-7082, 5μM BMS-345541, 10 μM LY-294002, or 50 μM SB203580. After stimulation, phenotypic analyses were performed by flow cytometry. Culture supernatants were cryopreserved for cytokine measurements. These supernatants were tested for IFN-α content by enzyme-linked immunoabsorbent assay (PBL Medical Laboratories) and for IL-6, IL-8, TNF-α, and CXCL10 by Cytometric Bead Array multiplex (CBA; BD Biosciences).

Protein extraction and signaling factor analysis

After stimulation of GEN2.2 cells, cytosolic and nuclear fractions were extracted with the use of the protein extraction kit from Active Motif. Nuclear extracts were probed for NF-κB subunits c-Rel, p50, p52, p65, and RelB content with the use of the TransAM NF-κB family kit (Active Motif). Whole-cell extracts were used for quantification of phospho-STAT1 and phospho-p38MAPK by CBA multiplex (BD Biosciences).

Western blot analysis

After activation, GEN2.2 cells were washed in phosphate-buffered saline (PBS), lysed in 100 mL sample buffer, and heated at 100°C for 5 minutes. Whole-cell extract (20 μg) was loaded onto a 12% SDS–polyacrylamide gel. After electrophoresis, proteins were transferred to a PVDF membrane (Bio-Rad). Nonspecific binding sites were blocked with 5% nonfat milk in PBS Tween20 0.1%. Membranes were then incubated with primary antibodies: antiphospho-STAT1 (4a/pY701), antiphospho-STAT2 (7a/pY; PharMingen), and anti-actin (Sigma-Aldrich). Antibody labeling was shown with horseradish peroxidase–conjugated secondary antibodies (Dako) and was visualized with enhanced chemiluminescence (Amersham Life Science).

Immunofluorescence

Twenty-one–well Teflon-printed slides (Immuno-Cell International) were coated with 1 μg/mL poly-l-lysine (Sigma-Aldrich). GEN2.2 cells (104) were added to each well and allowed to adhere for 1 hour. Cells were then stimulated for 2 and 3 hours with 640 UHA/mL UV-formol–inactivated influenza virus strain A/H3N2/Wisconsin/67/05, 1 μg/mL CL097, or 5000 U/mL human recombinant IFN-α. After activation, medium was removed, slides were fixed in −20°C methanol for 10 minutes and dried at room temperature for 1 hour. Slides were rehydrated in PBS for 10 minutes and then incubated with primary antibody anti-IRF7 (H-246; Santa Cruz Biotechnology). Antibody labeling was revealed with the use of FITC-conjugated goat anti–rabbit secondary antibody (PharMingen). Cells were also colored with 1 mg/mL Evans blue (Sigma-Aldrich) before visualization by fluorescence microscopy. Cells were observed with a Zeiss HBO 50/AC microscope with lens 63×/1.25 oil iris, and images were acquired with a Zeiss MC 80 DX camera by the image-acquisition software Axiovision 4 Version 4.0.1.0.

Quantitative reverse transcription PCR

Total RNA was isolated from stimulated GEN2.2 cells with the use of RNeasy kit (QIAGEN). Reverse transcription to cDNA was carried out by standard methods with reverse transcriptase (Roche Diagnostics) and dNTP (Roche). These cDNAs were amplified with naked primers, LightCycler TaqMan Master mix, and Universal ProbeLibrary probe (Roche). PCRs were conducted in a LightCycler instrument (Roche). Primers were synthesized by Roche; their sequences were as follows (listed 5′-3′): G6PDH (forward, AACAGAGTGAGCCCTTCTTCA; reverse, GGAGGCTGCATCATCGTACT); human TLR7 (forward, GCTAGACTGTCTCAAAAGAACAAAAA; reverse, GCCCACACTCAATCTGCAC); human TLR9 (forward, ATAGCCGTGAGCCGGAAT; reverse, GCAGGCAGAGGTGAGGTG); IFN-α1 (forward, CCCTCTCTTTATCAACAAACTTGC; reverse, TTGTTTTCATGTTGGACCAGA); IFN-α2 (forward, TCCTGCTTGAAGGACAGACA; reverse, TTTCAGCCTTTTGGAACTGG); IFN-β1 (forward, CTTTGCTATTTTCAGACAAGATTCA; reverse, GCCAGGAGGTTCTCAACAAT); IFN-ω1 (forward, ACCAGCTATAGCCCTGTTGG; reverse, AAGTAGGCCATGGTTCTGAGG); MxA (forward, TCCAGCCACCATTCCAAG; reverse, CAACAAGTTAAATGGTATCACAGAGC). Relative threshold cycle values for each gene were normalized to the housekeeping gene G6PDH with the use of the equation 2(−dCp), where Cp is the mean cross point of duplicate runs calculated by Lightcycler software and dCp = gene Cp − G6PDH Cp.

Influenza virus and CL097 induce human pDC activation via TLR7

This study focuses on TLR7 signaling in pDCs. The recently developed human pDC cell line, GEN2.2, was used as a model of pDCs.13,14  We evaluated 2 kinds of TLR ligands: CL097, an imidazoquinoline15,16  and influenza virus (Flu).17  We first verified their activity and their specificity for TLR7. After Flu or CL097 stimulation, both enriched primary pDCs (Figure 1A), and the GEN2.2 cells (Figure 1B) displayed the same activation profile with a high increase of CD40 expression; thus, both ligands were capable of inducing pDC activation. To verify the TLR7 specificity of the effect, TLR7 shRNA lentiviral transfection of GEN2.2 cells was performed, stably transfected cells were selected leading to the establishment of the GENshTLR7 cell line. These cells showed an 80% decrease in TLR7 mRNA expression, whereas levels of TLR9 mRNA were unchanged (Figure 1C). On Flu or CL097 stimulation, GENshTLR7 cells were unable to up-regulate CD40 but maintained their capacity to respond to TLR9 ligands (CpG A and B; Figure 1D). Moreover, GENshTLR7 cells produced neither IFN-α nor inflammatory cytokines after stimulation (data not shown). Control shRNA-transfected cells displayed the same activation profile as untransfected GEN2.2 cells, confirming that the previous results were not attributable to an alteration caused by lentiviral transfection (data not shown). Thus, both Flu and CL097 act specifically on TLR7 to induce CD40 up-regulation and cytokine production. Altogether, these data show that Flu and CL097 activate human pDCs by triggering TLR7.

Influenza virus and CL097 induce differential activation of pDCs

pDCs are known to secrete proinflammatory cytokines or substantial amounts of type I IFN on activation.3,4  The nature of the cytokines produced by GEN2.2 cells after activation by the 2 TLR7 ligands was evaluated. CL097-stimulated GEN2.2 cells produced the proinflammatory cytokines IL-6, IL-8, and TNF-α, whereas Flu-activated cells mainly secreted IFN-α (Figure 2A). To further characterize this secretion, the transcription of 4 type I IFN genes was assessed. Only Flu activation induced the transcription of ifn1, ifn2, ifn1, and ifn1 genes in GEN2.2 cells, whereas CL097 activation did not result in transcription of any of the IFN genes tested (Figure 2B). These results were confirmed with human blood pDCs. A higher number of IFN-α–secreting cells were observed after Flu activation (56%) than after CL097 stimulation (7%; Figure 2C). Thus, the 2 TLR7 ligands chosen induced different activation profiles for pDCs.

Figure 2

Differential pDC maturation is triggered by different TLR7 ligands. GEN2.2 cells were untreated or stimulated with Flu or CL097 for 24 hours. (A) Production of proinflammatory cytokines was measured in culture supernatants by enzyme-linked immunoabsorbent assay and CBA. The mean and SD from duplicate values of 3 independent experiments are shown. (B) After 24 hours of culture, RNA was extracted, and RNA expression levels of type I IFNs: IFN-α1, -α2, -β1, and -ω1 were measured by quantitative PCR. Data shown are normalized to G6PDH and are representative of 2 independent experiments. (C) PBMCs from healthy donors were cultured in the absence or presence of Flu or CL097 for 3 hours. Secretion was blocked by adding brefeldin A for a further 4 hours. Intracellular IFN-α production was measured in HLA-DR+ BDCA4+ pDCs by flow cytometry. Dot plots show the percentage of IFN-α–producing cells among pDCs. Representative results from 3 different donors are shown. (D) GEN2.2 cells were cultured in the absence or presence of Flu or CL097 for 3 hours. Cells were lysed and proteins were extracted. The different NF-κB subunits were quantified in nuclear fractions with the use of the TransAM kit. The mean OD and SD from duplicate values of 3 independent experiments are shown. (E) GEN2.2 cells were cultured in the absence or presence of Flu or CL097 for 3 hours. Cells were immunostained for IRF7 and Evans blue colored. Immunofluorescence was assessed by microscopy. Representative images from 3 independent experiments are shown.

Figure 2

Differential pDC maturation is triggered by different TLR7 ligands. GEN2.2 cells were untreated or stimulated with Flu or CL097 for 24 hours. (A) Production of proinflammatory cytokines was measured in culture supernatants by enzyme-linked immunoabsorbent assay and CBA. The mean and SD from duplicate values of 3 independent experiments are shown. (B) After 24 hours of culture, RNA was extracted, and RNA expression levels of type I IFNs: IFN-α1, -α2, -β1, and -ω1 were measured by quantitative PCR. Data shown are normalized to G6PDH and are representative of 2 independent experiments. (C) PBMCs from healthy donors were cultured in the absence or presence of Flu or CL097 for 3 hours. Secretion was blocked by adding brefeldin A for a further 4 hours. Intracellular IFN-α production was measured in HLA-DR+ BDCA4+ pDCs by flow cytometry. Dot plots show the percentage of IFN-α–producing cells among pDCs. Representative results from 3 different donors are shown. (D) GEN2.2 cells were cultured in the absence or presence of Flu or CL097 for 3 hours. Cells were lysed and proteins were extracted. The different NF-κB subunits were quantified in nuclear fractions with the use of the TransAM kit. The mean OD and SD from duplicate values of 3 independent experiments are shown. (E) GEN2.2 cells were cultured in the absence or presence of Flu or CL097 for 3 hours. Cells were immunostained for IRF7 and Evans blue colored. Immunofluorescence was assessed by microscopy. Representative images from 3 independent experiments are shown.

Close modal

The transcription factors NF-κB and IRF7 play essential roles in inducing the expression of proinflammatory cytokines and type I IFN, respectively. Therefore, we examined the translocation of these 2 factors into the nucleus after TLR7 triggering. Both Flu and CL097 induced similar nuclear translocation of p50 and p65 subunits of the canonical NF-κB dimer (Figure 2D). The quantities of p52 and RelB subunits of the noncanonical NF-κB pathway were not modified during activation. Interestingly, the NF-κB c-Rel subunit was detectable only in the nucleus of the CL097-stimulated GEN2.2 cells (Figure 2D). In contrast, IRF7 nuclear translocation was observed only in Flu-activated GEN2.2 cells (Figure 2E). This result corroborates the type I IFN production profile described in the second paragraph. These data suggest that, although both ligands induce inflammatory cytokine secretion via the NF-κB pathway, Flu but not CL097 induces IFN-α secretion after IRF7 translocation.

IFN-inducible genes are expressed after TLR7 triggering in a type I IFN–independent manner

Secretion of type I IFNs by pDCs leads to the expression of a group of IFN-inducible genes, including mxa, cxcl10, and trail.18,20  We next examined whether the differential type I IFN secretion observed after TLR7 triggering could induce differential expression of these IFN-inducible genes. IFN-α secretion was observed within 4 hours after TLR7 stimulation by Flu, whereas no detectable IFN-α was measured at any time point (up to 24 hours) after CL097 activation (Figure 3A). Surprisingly, the IFN-inducible gene products were nevertheless detected within 4 hours after activation by both TLR7 ligands (Figure 3B-D). The expression profiles were identical whatever the ligand applied: expression peaked at 4 hours and decreased at 24 hours for MxA and TRAIL gene products (Figure 3C-D), whereas CXCL10 production continuously increased up to and beyond 24 hours (Figure 3B). This suggests that, in our model, the induction of MxA, CXCL10, and TRAIL could be independent of IFN-α.

Figure 3

TLR7 triggers expression of IFN-inducible genes in a type I IFN–independent manner. GEN2.2 cells were untreated or stimulated with Flu or CL097 for different times. Supernatants were collected for evaluation of (A) IFN-α and (B) CXCL10 production. (C) RNA was extracted for measurement of MxA transcription levels by real-time PCR. (D) TRAIL expression was evaluated by flow cytometry. GEN2.2 cells were untreated or stimulated with Flu or CL097 in the absence or presence of anti–IFN-α/β and anti–IFN-α/βR neutralizing antibodies for 4 hours. (E) TRAIL expression was evaluated by flow cytometry, and (F) CXCL10 production was measured in culture supernatants by CBA. Data shown are the means and SDs from duplicate values from 2 independent experiments except data in panel C, which are representative of 2 independent experiments.

Figure 3

TLR7 triggers expression of IFN-inducible genes in a type I IFN–independent manner. GEN2.2 cells were untreated or stimulated with Flu or CL097 for different times. Supernatants were collected for evaluation of (A) IFN-α and (B) CXCL10 production. (C) RNA was extracted for measurement of MxA transcription levels by real-time PCR. (D) TRAIL expression was evaluated by flow cytometry. GEN2.2 cells were untreated or stimulated with Flu or CL097 in the absence or presence of anti–IFN-α/β and anti–IFN-α/βR neutralizing antibodies for 4 hours. (E) TRAIL expression was evaluated by flow cytometry, and (F) CXCL10 production was measured in culture supernatants by CBA. Data shown are the means and SDs from duplicate values from 2 independent experiments except data in panel C, which are representative of 2 independent experiments.

Close modal

Very low levels of type I IFN could be responsible for inducing the expression of IFN-inducible genes after CL097 stimulation of GEN2.2 cells. To investigate this possibility, GEN2.2 cells were stimulated by the 2 TLR7 ligands in the presence or absence of neutralizing anti–IFN-α/β and anti–IFN-α/βR antibodies. In accordance with the absence of any detectable IFN-α production, the blocking of type I IFN signaling had no effect on TRAIL and CXCL10 expression induced by TLR7 ligands (Figure 3E-F). The preincubation of cells with the neutralizing antibodies for 30 or 60 minutes before ligand exposure to exclude the involvement of any autocrine intracellular type I IFN pathway had no effect on these expressions (supplemental Figure 1, available on the Blood website; see the Supplemental Materials link at the top of the online article). However, the neutralizing antibodies could block both TRAIL and CXCL10 expression induced by exogenous IFNα/β. Altogether, these data show that expression of IFN-inducible genes could be triggered independently of type I IFN within 4 hours after TLR7 triggering.

Expression of IFN-inducible genes is independent of the NF-κB pathway and extracellular factors

To analyze whether NF-κB was involved in the observed expression of IFN-inducible genes, we measured TRAIL expression in GEN2.2 cells 4 hours after activation by Flu and CL097 in the presence of the I-κB kinase inhibitors BAY11-708221  and BMS-345541.22  Neither of these inhibitors altered the TRAIL expression profile (Figure 4A), whereas they both abrogated NF-κB–dependent CD40 up-regulation (Figure 4B). These results indicate that the expression of IFN-inducible genes after TLR7 triggering is independent of NF-κB activation.

Figure 4

Expression of IFN-inducible genes is independent of the NF-κB pathway and of extracellular factors. GEN2.2 cells were untreated or stimulated with Flu or CL097 in the absence or presence of the NF-κB inhibitors BAY and BMS. TRAIL (A) and CD40 (B) expressions were evaluated by flow cytometry after 4 hours and 24 hours of culture, respectively. The mean percentages and SDs from duplicate values of 4 independent experiments are shown. (C) GEN2.2 cells were untreated or stimulated for 2 hours. After washing, cells were placed in 24-well plates; untreated cells were added in transwells. After 4 hours of culture, TRAIL expression was evaluated by flow cytometry on cells in wells and transwells. Data shown are the means and SDs from duplicate values of 3 independent experiments. (D) GEN2.2 cells were untreated or stimulated for 2 hours in the absence or presence of anti–IFN-α/β and anti–IFN-α/βR neutralizing antibodies. After washing, cells were placed in 24-well plates; untreated cells were added in transwells. After 4 hours of culture, TRAIL expression was evaluated by flow cytometry on cells in wells and transwells. Data from 1 representative experiment are shown.

Figure 4

Expression of IFN-inducible genes is independent of the NF-κB pathway and of extracellular factors. GEN2.2 cells were untreated or stimulated with Flu or CL097 in the absence or presence of the NF-κB inhibitors BAY and BMS. TRAIL (A) and CD40 (B) expressions were evaluated by flow cytometry after 4 hours and 24 hours of culture, respectively. The mean percentages and SDs from duplicate values of 4 independent experiments are shown. (C) GEN2.2 cells were untreated or stimulated for 2 hours. After washing, cells were placed in 24-well plates; untreated cells were added in transwells. After 4 hours of culture, TRAIL expression was evaluated by flow cytometry on cells in wells and transwells. Data shown are the means and SDs from duplicate values of 3 independent experiments. (D) GEN2.2 cells were untreated or stimulated for 2 hours in the absence or presence of anti–IFN-α/β and anti–IFN-α/βR neutralizing antibodies. After washing, cells were placed in 24-well plates; untreated cells were added in transwells. After 4 hours of culture, TRAIL expression was evaluated by flow cytometry on cells in wells and transwells. Data from 1 representative experiment are shown.

Close modal

Extracellular factors could also contribute to the induction of expression of IFN-inducible genes. To rule out their involvement, TRAIL expression was measured on untreated GEN2.2 cells to which supernatants from TLR7-activated GEN2.2 cells were applied. No induction of TRAIL expression was observed in cells exposed to supernatants from CL097-activated cells (Figure 4C). The expression of TRAIL induced by the supernatant from Flu-activated cells was the result of the presence of secreted IFN-α in this supernatant. This expression could be blocked with the use of neutralizing anti–IFN-α/β antibodies (Figure 4D). These latter results show that expression of IFN-inducible genes after TLR7 triggering is caused by a previously undescribed intracellular signaling pathway.

Phosphorylation of STAT1 in response to TLR7 triggering depends on the PI3K-p38MAPK pathway

Expression of IFN-inducible genes is regulated by the transcription factor IFN-stimulated gene factor 3.23  IFN-stimulated gene factor 3 is in fact a complex of proteins, including STAT1, STAT2, and IRF9. We next examined whether this complex was activated in response to TLR7 triggering. Activation of GEN2.2 cells by both Flu and CL097 induced phosphorylation of STAT1 within 2 hours (Figure 5A-B) at levels comparable to the classic type I IFN activation (Figure 5B). Low-level phosphorylation of STAT2 was also observed (Figure 5B). In these analyses, no variation in total STAT protein quantity was observed (data not shown). These results were further confirmed by flow cytometric analysis. Results showing that STAT1 was phosphorylated in one-third of the GEN2.2 cells within 2 hours after TLR7 activation are presented in Figure 5C. The presence of neutralizing anti–IFN-α/β and anti–IFN-α/βR antibodies, which prevented IFN-α–induced STAT1 activation, had no effect on the level of STAT1 phosphorylation induced subsequent to TLR7 triggering (Figure 5D). Thus, STAT1 activation could also be induced via TLR7 signaling in a type I IFN–independent manner.

Figure 5

STAT1 is phosphorylated independently of type I IFN after TLR7 triggering with either ligand. GEN2.2 cells were untreated or stimulated with Flu, CL097, or IFN-α for 30 minutes, 2 hours, and 3 hours. Whole-cell protein extracts were prepared. (A) Phospho-STAT1 (pY701) was quantified in the protein extracts by CBA. Data shown are representative of 2 independent experiments. (B) Western blot analysis of phospho-STAT1 (pY701) and phospho-STAT2 (pY690) after activation of GEN2.2 cells. Data shown are representative of 2 independent experiments (C) After 2 hours of stimulation, cells were fixed and permeabilized for phospho-STAT1 (pY701) analysis by flow cytometry. Representative dot plots of 4 independent experiments are shown. (D) Activation of GEN2.2 cells in the absence or presence of anti–IFN-α/β and anti–IFN-α/βR neutralizing antibodies for 2 hours. Phospho-STAT1 (pY701) was analyzed by flow cytometry. Data shown are the means and SDs from duplicate values of 2 independent experiments.

Figure 5

STAT1 is phosphorylated independently of type I IFN after TLR7 triggering with either ligand. GEN2.2 cells were untreated or stimulated with Flu, CL097, or IFN-α for 30 minutes, 2 hours, and 3 hours. Whole-cell protein extracts were prepared. (A) Phospho-STAT1 (pY701) was quantified in the protein extracts by CBA. Data shown are representative of 2 independent experiments. (B) Western blot analysis of phospho-STAT1 (pY701) and phospho-STAT2 (pY690) after activation of GEN2.2 cells. Data shown are representative of 2 independent experiments (C) After 2 hours of stimulation, cells were fixed and permeabilized for phospho-STAT1 (pY701) analysis by flow cytometry. Representative dot plots of 4 independent experiments are shown. (D) Activation of GEN2.2 cells in the absence or presence of anti–IFN-α/β and anti–IFN-α/βR neutralizing antibodies for 2 hours. Phospho-STAT1 (pY701) was analyzed by flow cytometry. Data shown are the means and SDs from duplicate values of 2 independent experiments.

Close modal

Other groups have suggested that STAT1 phosphorylation in response to TLR9 triggering depends on p38MAPK activation.24  To test this hypothesis in the TLR7 model, we analyzed the activation of p38MAPK in stimulated GEN2.2 cells. Phosphorylation of p38MAPK was observed within 30 minutes after stimulation of GEN2.2 cells by either Flu or CL097, but not after activation by type I IFN, as was expected (Figure 6A). Thus, phosphorylation of p38MAPK is induced by TLR7 stimulation. We next investigated the possible pathway linking TLR7 via p38MAPK to STAT1. We found PI3K, which has previously been linked to the TLR7 pathway,25  to have a role in this pathway. The PI3K inhibitor LY294002, in addition to altering the p38MAPK phosphorylation profile in response to TLR7 triggering (Figure 6B), also impaired STAT1 phosphorylation and TRAIL expression by GEN2.2 cells. A similar effect was observed in the presence of the specific26  p38MAPK inhibitor SB203580 (Figure 6C-D). These results were confirmed on blood-isolated pDCs after stimulation of cells with Flu or CL097 whereby the presence of LY or SB inhibited TRAIL expression (Figure 6E) and CXCL10 production (Figure 6F). Taken together, these data show that both STAT1 activation and the expression of IFN-inducible genes after TLR7 triggering depended on the PI3K-p38MAPK pathway.

Figure 6

STAT1 phosphorylation and expression of IFN-inducible genes after TLR7 triggering depends on the PI3K-p38MAPK pathway. GEN2.2 cells were untreated or stimulated with Flu, CL097, or IFN-α for 30 minutes or 3 hours. (A) Phospho-p38MAPK (pT180/pY182) was quantified on whole-cell lysates by CBA. Data shown are representative of 2 independent experiments. (B) GEN2.2 cells were untreated or stimulated with Flu, CL097, or IFN-α for 30 minutes in the absence or presence of the PI3K inhibitor LY. Cells were fixed and permeabilized for phospho-p38MAPK (pT180/pY182) analysis by flow cytometry. The mean percentages and SDs from duplicate values of 2 independent experiments are shown. (C) Phospho-STAT1 (pY701) and (D) TRAIL expression were analyzed by flow cytometry after 2 hours or 4 hours of stimulation of GEN2.2 cells with Flu, CL097, or IFN-α in the absence or presence of LY and the specific p38MAPK inhibitor SB203580 (SB). The mean percentages and SDs from duplicate values of 3 independent experiments are shown. Blood-isolated pDCs were unstimulated or stimulated with Flu or CL097 for 4 hours in the absence or presence of LY and SB. (E) Expression of TRAIL was evaluated by flow cytometry. Dot plots representative of 2 experiments are shown. The percentage of TRAIL-positive cells is indicated on each plot. (F) CXCL10 production was measured in cell culture supernatants by CBA. The mean and SD from duplicate values of 2 experiments performed with 2 different donors are shown.

Figure 6

STAT1 phosphorylation and expression of IFN-inducible genes after TLR7 triggering depends on the PI3K-p38MAPK pathway. GEN2.2 cells were untreated or stimulated with Flu, CL097, or IFN-α for 30 minutes or 3 hours. (A) Phospho-p38MAPK (pT180/pY182) was quantified on whole-cell lysates by CBA. Data shown are representative of 2 independent experiments. (B) GEN2.2 cells were untreated or stimulated with Flu, CL097, or IFN-α for 30 minutes in the absence or presence of the PI3K inhibitor LY. Cells were fixed and permeabilized for phospho-p38MAPK (pT180/pY182) analysis by flow cytometry. The mean percentages and SDs from duplicate values of 2 independent experiments are shown. (C) Phospho-STAT1 (pY701) and (D) TRAIL expression were analyzed by flow cytometry after 2 hours or 4 hours of stimulation of GEN2.2 cells with Flu, CL097, or IFN-α in the absence or presence of LY and the specific p38MAPK inhibitor SB203580 (SB). The mean percentages and SDs from duplicate values of 3 independent experiments are shown. Blood-isolated pDCs were unstimulated or stimulated with Flu or CL097 for 4 hours in the absence or presence of LY and SB. (E) Expression of TRAIL was evaluated by flow cytometry. Dot plots representative of 2 experiments are shown. The percentage of TRAIL-positive cells is indicated on each plot. (F) CXCL10 production was measured in cell culture supernatants by CBA. The mean and SD from duplicate values of 2 experiments performed with 2 different donors are shown.

Close modal

Many studies have suggested that the effects triggered by TLR7 ligands as adjuvants in cancer immunotherapy could be attributed to the activation of pDCs and their subsequent type I IFN production.7,27,28  To understand how agonists for the same receptor lead to different activated pDC phenotypes, we initiated a study of the signaling pathways induced in pDCs by 2 different TLR7 ligands. In this study we show that, in contrast to Flu activation, CL097 stimulation of pDCs does not induce production of high levels of IFN-α. However, we were able to show that both TLR7 ligands led to rapid expression of IFN-inducible genes via a type I IFN–independent pathway.

The data presented here show that Flu and CL097 both trigger human TLR7 but induce differential cytokine production. With the use of stably shRNA-transfected cells, GENshTLR7, we could show that human pDC activation by inactivated influenza virus required TLR7, as had been previously described in a mouse model.17  Type I IFN production after CL097 activation was found to be null or extremely low compared with the massive amounts of IFN-α produced in the presence of Flu. This result corroborated those concerning IRF7 translocation into the nucleus of pDCs after stimulation with the different ligands. This differential type I IFN production is reminiscent of results obtained with TLR9 ligands.29  Kerkmann et al29  have shown that TLR9 triggering with CpG A ODNs leads to a high production of type I IFN, whereas CpG B ODNs, although they bind TLR9, are unable to induce IFN secretion. The mechanism responsible for the differential behavior of pDCs after TLR9 triggering has been suggested to be the result of the spatiotemporal location of the ligand within the cell and its higher order structure.30  Indeed, multimeric CpG A ODNs are retained within endosomes, whereas monomeric CpG B ODNs traffic to lysosomal compartments where they trigger the activation of different signaling factors.31,32  Similarly, Heil et al33  observed that synthetic TLR7 ligands induce higher IFN-α production by PBMCs when complexed to the cationic lipid DOTAP.33  Thus, Flu's RNAs may follow the same cell trafficking as multimeric CpG A ODNs, whereas CL097 may rather stay in a monomeric structure such as CpG B, leading to a lysosomal location and absence of type I IFN production.

MxA, CXCL10, and TRAIL, through their role in the inhibition of virus replication, in the chemotaxis of immune cells, and in the apoptosis of transformed and infected cells, are key players in countering infection and cancers.34  Here, we have shown that these genes, which have been described to be tightly regulated by IFNs in various hematopoietic cells,23,35  are rapidly expressed in TLR7-activated pDCs in the absence of type I IFN production, even when type I IFN–receptor signaling was blocked with neutralizing anti–IFN-α/β and anti–IFN-α/βR antibodies. Although expression of the “IFN-inducible” genes was independent of IFN, it remained dependent on STAT1 phosphorylation on Tyr-701. This phosphorylation was found to be p38MAPK dependent in our model, a similar signaling pathway was also described in CpG A–activated pDCs.24  However, because p38MAPK is a serine kinase, known to phosphorylate STAT1 on Ser-727,36  this tyrosine phosphorylation of STAT1 must be mediated by an intermediate tyrosine kinase whose activation depends on p38MAPK. Both the Janus kinases 1 and 2 and c-Src are able to phosphorylate STAT1 on Tyr-701,23,37  but their activation is generally associated with cytokine receptors.38  In our model, we have shown that no soluble factors were involved. Therefore, the intermediate kinase responsible for STAT1 phosphorylation in response to TLR7 stimulation remains to be identified.

Here, we have shown that PI3K is involved in the activation of p38MAPK, whereas other groups have shown its implication in IRF7 translocation.25,39  Moreover, the NF-κB–dependent expression of CD80 and cytokine production were also impaired in the presence of LY after TLR7 triggering (data not shown), suggesting that PI3K is a key early signaling factor in the TLR7 pathway.

Altogether, exhaustive characterization of the signaling pathway downstream of TLR7 in pDCs led us to describe the expression of IFN-inducible genes in the absence of cytokine receptor engagement. This originality may confer on pDCs the capacity to respond more rapidly to viral infection, reinforcing their role in the early phases of the immune response. Finally, the work described here may help to define new therapeutic targets that could favor pDC activation in tumor and viral immunotherapy or to dampen it in pDC-driven autoimmunity.

The online version of this article contains a data supplement.

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

This work was supported by 2 grants from the Institut National du Cancer and the Cancéropôle Lyon- Auvergne Rhône-Alpes (ACI-63-04 and Cancéropôle 2004-5). J.D.D.'s PhD was financed by the Ministère de l'Education Nationale de la Recherche et de la Technologie. A.B. was the recipient of a grant from EFS. M.G.-G. received a Young Researcher's grant from Inserm.

Contribution: J.D.D. performed the experiments, analyzed the results, made the figures, and wrote the paper; A.B. and J.-P.M. performed part of the experiments; M.G.-G., L.C., and J.P. proofread the manuscript; and J.D.D., L.C., and J.P. designed the research.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Laurence Chaperot, R&D Laboratory, EFS Rhônes-Alpes, 29 Av Maquis du Gresivaudan, BP35 La Tronche, 38701 France; e-mail: laurence.chaperot@efs.sante.fr.

1
Akira
S
Takeda
K
Toll-like receptor signalling.
Nat Rev Immunol
2004
4
7
499
511
2
Ito
T
Wang
YH
Liu
YJ
Plasmacytoid dendritic cell precursors/type I interferon-producing cells sense viral infection by Toll-like receptor (TLR) 7 and TLR9.
Springer Semin Immunopathol
2005
26
3
221
229
3
Gibson
SJ
Lindh
JM
Riter
TR
et al
Plasmacytoid dendritic cells produce cytokines and mature in response to the TLR7 agonists imiquimod and resiquimod.
Cell Immunol
2002
218
1–2
74
86
4
Asselin-Paturel
C
Trinchieri
G
Production of type I interferons: plasmacytoid dendritic cells and beyond.
J Exp Med
2005
202
4
461
465
5
Fonteneau
JF
Gilliet
M
Larsson
M
et al
Activation of influenza virus-specific CD4+ and CD8+ T cells: a new role for plasmacytoid dendritic cells in adaptive immunity.
Blood
2003
101
9
3520
3526
6
Chaperot
L
Blum
A
Manches
O
et al
Virus or TLR agonists induce TRAIL-mediated cytotoxic activity of plasmacytoid dendritic cells.
J Immunol
2006
176
1
248
255
7
Stary
G
Bangert
C
Tauber
M
Strohal
R
Kopp
T
Stingl
G
Tumoricidal activity of TLR7/8-activated inflammatory dendritic cells.
J Exp Med
2007
204
6
1441
1451
8
Garland
SM
Waddell
R
Mindel
A
Denham
IM
McCloskey
JC
An open-label phase II pilot study investigating the optimal duration of imiquimod 5% cream for the treatment of external genital warts in women.
Int J STD AIDS
2006
17
7
448
452
9
Gollnick
H
Barona
CG
Frank
RG
et al
Recurrence rate of superficial basal cell carcinoma following treatment with imiquimod 5% cream: conclusion of a 5-year long-term follow-up study in Europe.
Eur J Dermatol
2008
18
6
677
682
10
Gilliet
M
Cao
W
Liu
YJ
Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases.
Nat Rev Immunol
2008
8
8
594
606
11
Birmachu
W
Gleason
RM
Bulbulian
BJ
et al
Transcriptional networks in plasmacytoid dendritic cells stimulated with synthetic TLR 7 agonists.
BMC Immunol
2007
8
26
12
Gautier
G
Humbert
M
Deauvieau
F
et al
A type I interferon autocrine-paracrine loop is involved in Toll-like receptor-induced interleukin-12p70 secretion by dendritic cells.
J Exp Med
2005
201
9
1435
1446
13
Chaperot
L
Bendriss
N
Manches
O
et al
Identification of a leukemic counterpart of the plasmacytoid dendritic cells.
Blood
2001
97
10
3210
3217
14
Chaperot
L
Perrot
I
Jacob
MC
et al
Leukemic plasmacytoid dendritic cells share phenotypic and functional features with their normal counterparts.
Eur J Immunol
2004
34
2
418
426
15
Jurk
M
Heil
F
Vollmer
J
et al
Human TLR7 or TLR8 independently confer responsiveness to the antiviral compound R-848 [letter].
Nat Immunol
2002
3
6
499
16
Lee
J
Chuang
TH
Redecke
V
et al
Molecular basis for the immunostimulatory activity of guanine nucleoside analogs: activation of Toll-like receptor 7.
Proc Natl Acad Sci U S A
2003
100
11
6646
6651
17
Diebold
SS
Kaisho
T
Hemmi
H
Akira
S
Reis e Sousa
C
Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA.
Science
2004
303
5663
1529
1531
18
Gurney
KB
Colantonio
AD
Blom
B
Spits
H
Uittenbogaart
CH
Endogenous IFN-alpha production by plasmacytoid dendritic cells exerts an antiviral effect on thymic HIV-1 infection.
J Immunol
2004
173
12
7269
7276
19
Megjugorac
NJ
Young
HA
Amrute
SB
Olshalsky
SL
Fitzgerald-Bocarsly
P
Virally stimulated plasmacytoid dendritic cells produce chemokines and induce migration of T and NK cells.
J Leukoc Biol
2004
75
3
504
514
20
Kemp
TJ
Elzey
BD
Griffith
TS
Plasmacytoid dendritic cell-derived IFN-alpha induces TNF-related apoptosis-inducing ligand/Apo-2L-mediated antitumor activity by human monocytes following CpG oligodeoxynucleotide stimulation.
J Immunol
2003
171
1
212
218
21
Pierce
JW
Schoenleber
R
Jesmok
G
et al
Novel inhibitors of cytokine-induced IkappaBalpha phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo.
J Biol Chem
1997
272
34
21096
21103
22
Burke
JR
Pattoli
MA
Gregor
KR
et al
BMS-345541 is a highly selective inhibitor of I kappa B kinase that binds at an allosteric site of the enzyme and blocks NF-kappa B-dependent transcription in mice.
J Biol Chem
2003
278
3
1450
1456
23
Stark
GR
How cells respond to interferons revisited: from early history to current complexity.
Cytokine Growth Factor Rev
2007
18
5–6
419
423
24
Takauji
R
Iho
S
Takatsuka
H
et al
CpG-DNA-induced IFN-alpha production involves p38 MAPK-dependent STAT1 phosphorylation in human plasmacytoid dendritic cell precursors.
J Leukoc Biol
2002
72
5
1011
1019
25
Guiducci
C
Ghirelli
C
Marloie-Provost
MA
et al
PI3K is critical for the nuclear translocation of IRF-7 and type I IFN production by human plasmacytoid predendritic cells in response to TLR activation.
J Exp Med
2008
205
2
315
322
26
Davies
SP
Reddy
H
Caivano
M
Cohen
P
Specificity and mechanism of action of some commonly used protein kinase inhibitors.
Biochem J
2000
35
pt 1
95
105
27
Inglefield
JR
Dumitru
CD
Alkan
SS
et al
TLR7 agonist 852A inhibition of tumor cell proliferation is dependent on plasmacytoid dendritic cells and type I IFN.
J Interferon Cytokine Res
2008
28
4
253
263
28
Smits
EL
Ponsaerts
P
Berneman
ZN
Van Tendeloo
VF
The use of TLR7 and TLR8 ligands for the enhancement of cancer immunotherapy.
Oncologist
2008
13
8
859
875
29
Kerkmann
M
Rothenfusser
S
Hornung
V
et al
Activation with CpG-A and CpG-B oligonucleotides reveals two distinct regulatory pathways of type I IFN synthesis in human plasmacytoid dendritic cells.
J Immunol
2003
170
9
4465
4474
30
Honda
K
Ohba
Y
Yanai
H
et al
Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction.
Nature
2005
434
7036
1035
1040
31
Guiducci
C
Ott
G
Chan
JH
et al
Properties regulating the nature of the plasmacytoid dendritic cell response to Toll-like receptor 9 activation.
J Exp Med
2006
203
8
1999
2008
32
Haas
T
Schmitz
F
Heit
A
Wagner
H
Sequence independent interferon-alpha induction by multimerized phosphodiester DNA depends on spatial regulation of Toll-like receptor-9 activation in plasmacytoid dendritic cells.
Immunology
2009
126
2
290
298
33
Heil
F
Hemmi
H
Hochrein
H
et al
Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8.
Science
2004
303
5663
1526
1529
34
Chawla-Sarkar
M
Lindner
DJ
Liu
YF
et al
Apoptosis and interferons: role of interferon-stimulated genes as mediators of apoptosis.
Apoptosis
2003
8
3
237
249
35
van Boxel-Dezaire
AH
Rani
MR
Stark
GR
Complex modulation of cell type-specific signaling in response to type I interferons.
Immunity
2006
25
3
361
372
36
Kovarik
P
Stoiber
D
Eyers
PA
et al
Stress-induced phosphorylation of STAT1 at Ser727 requires p38 mitogen-activated protein kinase whereas IFN-gamma uses a different signaling pathway.
Proc Natl Acad Sci U S A
1999
96
24
13956
13961
37
Chang
YJ
Holtzman
MJ
Chen
CC
Differential role of Janus family kinases (JAKs) in interferon-gamma-induced lung epithelial ICAM-1 expression: involving protein interactions between JAKs, phospholipase Cgamma, c-Src, and STAT1.
Mol Pharmacol
2004
65
3
589
598
38
Baker
SJ
Rane
SG
Reddy
EP
Hematopoietic cytokine receptor signaling.
Oncogene
2007
26
47
6724
6737
39
Cao
W
Manicassamy
S
Tang
H
et al
Toll-like receptor-mediated induction of type I interferon in plasmacytoid dendritic cells requires the rapamycin-sensitive PI(3)K-mTOR-p70S6K pathway.
Nat Immunol
2008
9
10
1157
1164

Supplemental data

Sign in via your Institution