Plasmacytoid dendritic cells (pDCs) produce large amounts of type I IFN in response to TLR7/9 ligands. This conveys antiviral effects, activates other immune cells (NK cells, conventional DCs, B, and T cells), and causes the induction and expansion of a strong inflammatory response. pDCs are key players in various type I IFN–driven autoimmune diseases such as systemic lupus erythematosus or psoriasis, but pDCs are also involved in (anti-)tumor immunity. The sphingolipid sphingosine-1-phosphate (S1P) signals through five G-protein–coupled receptors (S1PR1–5) to regulate, among other activities, immune cell migration and activation. The present study shows that S1P stimulation of human, primary pDCs substantially decreases IFN-α production after TLR7/9 activation with different types of CpG oligodeoxynucleotides or tick-borne encephalitis vaccine, which occurred in an S1PR4-dependent manner. Mechanistically, S1PR4 activation preserves the surface expression of the human pDC-specific inhibitory receptor Ig-like transcript 7. We provide novel information that Ig-like transcript 7 is rapidly internalized upon receptor-mediated endocytosis of TLR7/9 ligands to allow high IFN-α production. This is antagonized by S1PR4 signaling, thus decreasing TLR-induced IFN-α secretion. At a functional level, attenuated IFN-α production failed to alter Ag-driven T cell proliferation in pDC-dependent T cell activation assays, but shifted cytokine production of T cells from a Th1 (IFN-γ) to a regulatory (IL-10) profile. In conclusion, S1PR4 agonists block human pDC activation and may therefore be a promising tool to restrict pathogenic IFN-α production.

Plasmacytoid dendritic cells (pDCs) are a rare DC subpopulation, characterized by their potent ability to produce large amounts of type I IFN (IFN-α/β; IFN-I), a plasma cell-like morphology, and a unique set of cell-surface markers. IFN-α plays a crucial role in antiviral immunity by augmenting the expression of a broad repertoire of antiviral molecules that dampen viral spread and promote apoptosis of infected cells (reviewed in Ref. 1). Furthermore, IFN-α was shown to inhibit the growth of cells undergoing malignant transformation (reviewed in Ref. 2). In pDCs, IFN-α is produced mainly via the TLR7/9–MyD88 pathway and the rapid production of high levels IFN-α is due to the constitutive expression of the transcription factor IFN regulatory factor 7 (IRF7) (3). TLR7 and -9 sense viral ssRNA and unmethylated CpG oligodeoxynucleotides (ODNs), respectively, and are located within endosomes. The subcellular localization of TLR7/9 prevents the host to respond to self-DNA, which might induce autoreactive immune responses. The uptake of physiological TLR7/9 ligands and their shuttling to the endosomal compartment is a controlled, receptor-mediated process. However, detailed mechanisms of TLR7/9 ligand uptake are currently unclear and require further investigation (4).

Controlling pDC activation is indispensable to avoid an overshooting immune response that can harm the host. Therefore, pDCs express a set of regulatory surface receptors that regulate IFN-α production. Among these, Ig-like transcript 7 (ILT7), a surface marker exclusively expressed on human pDCs, was shown to associate with the adapter protein FcεRIγ. Activation of this receptor complex causes ITAM-mediated signaling to restrict IFN-α production (57). The only known ligand for ILT7 is bone marrow stromal Ag 2 (BST2), which is expressed on immune cells such as pDCs, but also on cancer cells, including melanoma and breast cancer (8). The current model of ILT7/BST2 interaction suggests that BST2 expression is upregulated in an inflammatory environment to interact with ILT7, serving as a negative-feedback loop to prevent an uncontrolled and overshooting IFN-α response. In contrast, constitutive expression of BST2 by various cancer types inhibits IFN-α production of tumor-infiltrating pDCs. As IFN-α has antiangiogenic and proapoptotic effects on cancer cells, this mechanism allows tumor-associated immunosuppression. However, leukemic pDCs tend to downregulate ILT7 although the underlying reason is unclear (9). pDCs were shown to infiltrate human solid tumors without secreting adequate levels of IFN-α after TLR9 activation (10). Tumors orchestrate an immunosuppressive microenvironment, which is characterized by specific mediators (e.g., sphingosine-1-phosphate [S1P], PGE2, IL-10, vascular endothelial growth factor, and TGF-β), released from tumor-associated immune or cancer cells, favoring regulatory immune cell phenotypes (reviewed in Ref. 11). Furthermore, pDCs, like all DCs, are able to present Ags to T cells and therefore induce, depending on the microenvironment, variable adaptive immune responses. There is evidence that pDCs promote regulatory T cell (Treg) expansion within the tumor by presentation of self-Ags to naive T cells. However, other studies show that fully activated pDCs can have a cytotoxic, tumoricidal function (12).

S1P is a bioactive sphingolipid mediator that is produced by various (immune) cell types (e.g., RBCs, platelets, macrophages, and DCs) (reviewed in Ref. 13). S1P acts as a specific ligand for five receptors (S1PR1–5), which differ in tissue distribution. For instance, S1PR1 is ubiquitously expressed, whereas S1PR4 is largely restricted to lymphoid tissue (14). Due to the fact that S1PRs couple to diverse G-proteins, S1P is able to affect multiple cellular and physiological functions (e.g., immune cell trafficking, angiogenesis, vascular maturation, cardiac development, and immune cell activation) (reviewed in Ref. 15). Currently, S1PR4 signaling is ill defined. However, its activation might alter immune cell phenotypes and activation (1619).

In the current study, we investigated the influence of S1P on pDC activation and function. Administration of S1P decreases IFN-α production via S1PR4. Mechanistically, S1PR4 signaling preserves ILT7 surface expression that normally decreases upon pDC activation. This enables ILT7 to bind to its ligand and to decrease IFN-α production. As a consequence of reduced IFN-α production, S1PR4 signaling in pDCs shifts cytokine production of cocultured T cells from a Th1 (IFN-γ) to a regulatory (IL-10) profile. S1PR4 signaling inhibits human pDC activation and might therefore be a promising target to restrict pathogenic IFN-α production in several IFN-I–induced autoimmune diseases or to increase IFN-I production in tumors.

PBMCs were obtained from Buffy Coats (DRK-Blutspendedienst Baden-Württemberg-Hessen, Frankfurt, Germany) using Ficoll–Isopaque (PAA Laboratories, Cölbe, Germany) gradient centrifugation; RBCs were removed by KCl lysis. PBMCs were cultured in six-well plates in RPMI 1640 (PAA Laboratories) containing 2% FCS. Primary pDCs were magnetically purified from PBMCs using a pDC purification kit (negative selection), according to the manufacturer’s instructions, and the autoMACS Separator (Miltenyi Biotec, Bergisch Gladbach, Germany). pDCs were characterized as blood DC Ag 4+, CD123+, MHC class II (MHC II)int, and CD11c. The purity was ≥93%. Cells were cultured in 96-well plates in X-Vivo (Lonza, Verviers, Belgium) supplemented with 2.5% human serum containing 20 ng/ml IL-3 (Immunotools, Friesoythe, Germany). T cells were isolated from PBMCs using the Pan T cell isolation kit, according to the manufacturer’s instructions, and the autoMACS Separator (Miltenyi Biotec) and cultured in RPMI 1640 (PAA Laboratories) supplemented with 10% heat-inactivated FCS, 1% nonessential amino acids, 1% essential amino acids, 1% sodium pyruvate, 1% HEPES buffer (all from PAA Laboratories), 2-ME, and 10 ng/ml IL-2 (Immunotools).

S1P and VPC23019 (both 1 μmol; Avanti Polar Lipids, Alabaster, AL) were dissolved following the manufacturers’ instructions. JTE-013 (100 nmol; Biomol, Hamburg, Germany), Cym50358 (200 nmol), and Cym50138 (200 nmol) were dissolved in DMSO. CpG-A (2336) ODN (5 μg/ml), CpG-A (2336) FITC, CpG-B (2006) ODN (1 μmol), imiquimod (IMQ; 2.5 μg/ml) (all from Invivogen, San Diego, CA), 10% v/v FSME-Immun (Baxter, Unterschleißheim, Germany), Lyn peptide inhibitor (LPI; 10 μmol), RhoA inhibitor Rhosin (10 μmol), and Rho-associated protein kinase (ROCK) inhibitor Y-27632 (10 μmol) (Tocris Bioscience, Bristol, U.K.) were dissolved following the manufacturers’ instructions.

RNA from PBMCs was isolated using peqGold RNA Pure (Peqlab Biotechnologie, Erlangen, Germany) followed by cDNA transcription with the iScript cDNA synthesis kit (Bio-Rad, München, Germany). pDC RNA (≤1 × 106 cells/sample) was isolated using the RNeasy Micro kit (Qiagen, Hilden, Germany) and quantitated using the Bioanalyzer (Agilent Technologies, Böblingen, Germany) followed by transcription with sensiscript RT kits (Qiagen). Real-time quantitative PCR (qPCR) was performed using the MyIQ real-time PCR system (Bio-Rad) and Absolute Blue qPCR SYBR Green fluorescein mix (Thermo Scientific, Karlsruhe, Germany). Primers against all S1P receptors (S1PR1–5), IFN-α2, and IRF7 were from Qiagen. Additional primers (Biomers, Ulm, Germany) were: Mx1 sense, 5′-CACCGTGACACTGGGATTC-3′ and antisense, 5′-ATAGCGAGGAGGTGCTGAAG-3′; and dsRNA-activated protein kinase sense, 5′-CTAATTTGGCTGCGGCATT-3′ and antisense, 5′-CCGTCAGAAGCAGGGAGTAG-3′. Actin expression was used for normalization. RT-qPCR results were quantified using Gene Expression Macro (Bio-Rad).

S1PR4 silencing in primary human pDCs was performed using the Accell siRNA system (Dharmacon, Lafayette, CO) essentially following the manufacturer’s instructions. Briefly, 1 × 105 pDCs were incubated with 1 μmol pooled small interfering RNA (siRNA; S1PR4 or nontargeting) in 100 μl serum-free medium containing IL-3 for 48 h, followed by addition of 100 μl medium containing human serum for another 24 h.

CCL5, CXCL10, IL-2, IL-6, IL-8, IL-10, IFN-α, INF-γ, or TNF-α in cell-culture supernatants were quantified using a cytometric bead array (CBA) Th1/Th2/Th17 Kit or CBA Flex Sets (BD Biosciences, Heidelberg, Germany). Samples were acquired by flow cytometry and processed with FCAP software V1.0.1 (BD Biosciences).

For quantification of PGE2 levels in pDC supernatants, a commercial ELISA (Biomol) was used as described (20).

For detection of surface markers, cells were harvested and washed in PBS, followed by incubation with Fc-blocking reagent (Miltenyi Biotec) in 0.5% BSA/PBS for 15 min and Ab staining for 30 min on ice. The following Abs were used: anti-CD123–allophycocyanin or anti-CD123–PE-Cy5, anti- blood DC Ag 4–allophycocyanin, anti-ILT7–biotin (all from Miltenyi Biotec); anti–MHC II–PE-Cy7, anti-ILT7–PE, anti-CD86–FITC, streptavidin-PE–CF594 (all from BD Biosciences); anti-CD11c–FITC (Immunotools), anti-CD40–FITC, anti-CD80–allophycocyanin, anti-CD83–PE, anti-OX40L–Alexa Fluor647 (all from BioLegend, San Diego, CA); and anti-CD3–eFluor 605NC (eBioscience, San Diego, CA), or anti-TLR9–PE Ab (clone 26C593.2; Novus Biologicals, Wiesbaden, Germany). To detect intracellular protein levels, pDCs were resuspended in Fix/Perm solution (BD Biosciences) and incubated for 20 min on ice. Cells were washed twice in Perm/Wash solution (BD Biosciences), followed by anti-S1PR4–FITC (Biozol, Eching, Germany), anti–IFN-α–PE (BD Biosciences), or anti-granzyme B (GrB)–PE Ab (Immunotools) staining for 30 min on ice. To analyze accumulation of pDCs in cell clusters, PBMCs were stimulated as indicated and immediately fixed in paraformaldehyde (1% in PBS), followed by Ab staining. Samples were analyzed on an LSR II/Fortessa flow cytometer (BD Biosciences).

Human primary pDCs were isolated as described above and plated in 96-well plates with IL-3 (20 ng/ml) overnight. pDCs were treated with FITC-labeled CpG-A (10 μg/ml) for 5 min and prestimulated with an S1PR4 agonist (Cym50138; 200 nmol) for 30 min. Cells were then harvested and centrifuged on glass slides using cytospin. pDCs were fixed with 1% paraformaldehyde for 30 min at room temperature followed by permeabilization in 2% Triton X-100/PBS for 10 min. For analyzing ILT7 internalization, biotinylated ILT7 Abs (Miltenyi Biotec) and streptavidin-PE–CF594 (BD Biosciences) were added for 1 h each. Nuclei were counterstained with DAPI (1 μg/ml) for 20 min at room temperature. Cover slips were mounted on microscopy slides using Vectashield H 1400 mounting medium (Vector Laboratories, Burlingame, CA) and analyzed with the AxioVert 200M fluorescence microscope and the AxioVision software (all from Carl Zeiss Microimaging, Jena, Germany).

For direct coculture assays, T cells and pDCs were isolated from human blood of the same donor. A total of 5 × 105 T cells/ml was seeded in a 96-well plate and cultured with IL-2 (10 ng/ml) for 24 h. A total of 1 × 105 pDCs/ml was seeded in a 96-well plate and cultured with IL-3 (20 ng/ml) and incubated with tetanus toxoid (TTX; 2 μg/ml) for 24 h. To investigate the influence of pDC S1PR4 signaling on T cell proliferation and cytokine production, pDCs were stimulated with Cym50138 (200 nml) for 30 min. Afterwards, pDCs were washed with PBS and resuspended in T cell medium containing IL-3 (20 ng/ml) and IL-2 (10 ng/ml) with or without the addition of CpG-A (5 μg/ml). T cells were stained with the proliferation dye eFluor 670 (5 mmol; eBioscience), washed twice in PBS, and mixed with pDCs at a ratio of 1:5 (pDC/T cells). For indirect coculture assays, primary human T cells were stained with eFluor670 and preactivated using ImmunoCult Human CD3/CD28/CD2 T Cell Activator (Stemcell Technologies, Cologne, Germany), seeded in 96-well plates (5 × 105 T cells in 100 μl T cell medium), and daily pulsed with 50 μl primary pDC supernatants for 5 d. Cells and supernatants were harvested after 5 d of culturing. Supernatants were measured for T cell cytokine secretion as described above. Cells were stained for CD3 expression, and proliferation of CD3+ T cells was measured using flow cytometry by analyzing eFluor 670 dilution.

Data were analyzed using GraphPad Prism 5.0 (GraphPad Software, San Diego, CA). The p values were calculated using one-sample t test, Student t test, or ANOVA with Bonferroni correction as indicated in the figure legends.

Taking immunomodulatory effects of S1P on various cell types into account, we asked whether stimulation with S1P affects pDC activation. We stimulated human primary pDCs with the TLR9 agonist CpG-A to induce IFN-α secretion. Supplying S1P, followed by CpG-A addition for additional 16 h, concentration-dependently reduced IFN-α secretion (Fig. 1A). Next, we asked for the S1P receptor suppressing IFN-α secretion. We first analyzed the S1PR expression profile on human primary pDCs before and after activation with CpG-A by RT-qPCR. Unstimulated human pDCs dominantly expressed S1PR1 as well as S1PR4 and, to a lesser extent, S1PR5 (Fig. 1B). Interestingly, S1PR4 was strongly downregulated 16 h after pDC activation both on mRNA (Fig. 1B) and protein level (Fig. 1C), suggesting a regulatory role in pDC activation. To identify the S1PR transmitting the S1P-dependent IFN-α inhibition, we prestimulated human primary pDCs with S1P in combination with antagonists of different S1PRs for 30 min, followed by TLR9 activation with CpG-A for an additional 16 h. We used VPC23019, a selective antagonist of S1PR1/3 (21), JTE-013, a S1PR2 inhibitor when used at 100 nmol (18), and Cym50358, a potent and selective S1PR4 antagonist (22). Neither VPC23019 nor JTE-013 prevented the S1P-triggered decrease in IFN-α, indicating that S1PR1/2/3 signaling were dispensable for IFN-α suppression by S1P. However, blocking S1PR4 with Cym50358 significantly abolished the S1P-dependent IFN-α decrease (Fig. 1D). Thus, S1P inhibits IFN-α secretion exclusively via S1PR4. Because IFN-α production by pDCs is amplified in a positive feed-forward loop, we asked whether the S1P-triggered reduction was due to a blockade of IFN-α/β receptor signaling or whether it affected early IFN-α production. We used the selective pharmacological S1PR4 agonist Cym50138 (23) to mimic S1P effects. S1PR4-dependent reduction of TLR9-induced IFN-α secretion was obvious 4 h after TLR9 activation (Fig. 1E). Therefore, S1PR4 signaling influences early IFN-α production by pDCs. To validate the specificity of the S1PR4 agonist, we performed siRNA-mediated knockdown of S1PR4 in primary human pDC and analyzed IFN-α2 expression by qPCR after activation with CpG-A with or without addition of Cym50138. In control siRNA–transfected pDCs, the S1PR4 agonist reduced CpG-A–dependent IFN-α expression, which was reversed in S1PR4 knockdown pDCs (Fig. 1F, 1G). The effect of S1PR4 signaling on soluble mediator production seemed to be rather specific for IFN-α, as CpG-A–dependent expression of the immune mediators IL-6, IL-8, TNF-α, CXCL10, CCL5, and PGE2 was not affected by preincubation with the S1PR4 agonist (Fig. 1H–J). In conclusion, S1P selectively reduced IFN-α expression by primary human pDC via S1PR4.

FIGURE 1.

S1PR4-dependent reduction of IFN-α production in human pDCs. (AJ) Human pDCs were isolated from buffy coats and cultured in X-Vivo medium containing IL-3 (20 ng/ml). (A) pDCs were controls or activated with the TLR9 agonist CpG-A (5 μg/ml) for 16 h and prestimulated with 0.1–1 μmol S1P for 30 min. IFN-α secretion was measured by CBA. Data are means ± SEM of five independent experiments. (B) S1PR expression on freshly isolated human pDCs and 4 or 16 h after activation with CpG-A was determined by qPCR. Data are means ± SEM of five independent experiments. (C) pDCs were controls or activated with the TLR9 agonist CpG-A (5 μg/ml) for 16 h. Expression of S1PR4 and IFN-α were measured by intracellular Ab staining and FACS. Representative FACS plots of three independent experiments are shown. (D) pDCs were activated with CpG-A (CpG2336) for 16 h with or without 30 min S1P (1 μmol) prestimulation and/or the S1PR antagonists JTE-013 (S1PR2/4 antagonist; 100 nmol), VPC23019 (S1PR1/3 antagonist, 1 μmol), and Cym50358 (S1PR4 antagonist; 200 nmol). The CpG-A–activated group was set to 1. Data are means ± SEM of three independent experiments. (E) Cells were activated with CpG-A for 4 or 16 h, respectively, with or without preaddition of the specific S1PR4 agonist Cym50138 (200 nmol) for 30 min. IFN-α secretion was measured by CBA. The control group was set to 1. Data are means ± SEM of four independent experiments. pDCs were transfected with nontargeting siRNA (ctrl siRNA) or S1PR4 siRNA for 72 h, activated with CpG-A for 16 h with or without preaddition of the specific S1PR4 agonist Cym50138 (200 nmol) for 30 min, and expression of IFN-α (F) and S1PR4 (G) compared with actin was quantified by quantitative RT-PCR. Data are means ± SEM of five independent experiments. pDCs were activated with CpG-A for 16 h with or without preaddition of the specific S1PR4 agonist Cym50138 (200 nmol) for 30 min, and IL-6, IL-8, and TNF-α (H), CXCL10 and CCL5 (I), and PGE2 (J) levels in supernatants were quantified by CBA (H and I) or ELISA (J). Data are means ± SEM of six independent experiments. p values were calculated using one-sample t test (G), Student t test (D and F), one-way ANOVA with Bonferroni correction (A and E), or two-way ANOVA with Bonferroni correction (B). *p ≤ 0.05, **p ≤ 0.01.

FIGURE 1.

S1PR4-dependent reduction of IFN-α production in human pDCs. (AJ) Human pDCs were isolated from buffy coats and cultured in X-Vivo medium containing IL-3 (20 ng/ml). (A) pDCs were controls or activated with the TLR9 agonist CpG-A (5 μg/ml) for 16 h and prestimulated with 0.1–1 μmol S1P for 30 min. IFN-α secretion was measured by CBA. Data are means ± SEM of five independent experiments. (B) S1PR expression on freshly isolated human pDCs and 4 or 16 h after activation with CpG-A was determined by qPCR. Data are means ± SEM of five independent experiments. (C) pDCs were controls or activated with the TLR9 agonist CpG-A (5 μg/ml) for 16 h. Expression of S1PR4 and IFN-α were measured by intracellular Ab staining and FACS. Representative FACS plots of three independent experiments are shown. (D) pDCs were activated with CpG-A (CpG2336) for 16 h with or without 30 min S1P (1 μmol) prestimulation and/or the S1PR antagonists JTE-013 (S1PR2/4 antagonist; 100 nmol), VPC23019 (S1PR1/3 antagonist, 1 μmol), and Cym50358 (S1PR4 antagonist; 200 nmol). The CpG-A–activated group was set to 1. Data are means ± SEM of three independent experiments. (E) Cells were activated with CpG-A for 4 or 16 h, respectively, with or without preaddition of the specific S1PR4 agonist Cym50138 (200 nmol) for 30 min. IFN-α secretion was measured by CBA. The control group was set to 1. Data are means ± SEM of four independent experiments. pDCs were transfected with nontargeting siRNA (ctrl siRNA) or S1PR4 siRNA for 72 h, activated with CpG-A for 16 h with or without preaddition of the specific S1PR4 agonist Cym50138 (200 nmol) for 30 min, and expression of IFN-α (F) and S1PR4 (G) compared with actin was quantified by quantitative RT-PCR. Data are means ± SEM of five independent experiments. pDCs were activated with CpG-A for 16 h with or without preaddition of the specific S1PR4 agonist Cym50138 (200 nmol) for 30 min, and IL-6, IL-8, and TNF-α (H), CXCL10 and CCL5 (I), and PGE2 (J) levels in supernatants were quantified by CBA (H and I) or ELISA (J). Data are means ± SEM of six independent experiments. p values were calculated using one-sample t test (G), Student t test (D and F), one-way ANOVA with Bonferroni correction (A and E), or two-way ANOVA with Bonferroni correction (B). *p ≤ 0.05, **p ≤ 0.01.

Close modal

Next, we asked for consequences of S1PR4-dependent IFN-α suppression. PDC-derived IFN-α mediates its antiviral and growth inhibitory effects through expression of IFN-I–stimulated genes (ISGs) (reviewed in Ref. 24). To investigate the relevance of the reduced IFN-α levels after S1PR4 activation on antiviral properties of pDCs, whole PBMC cultures were prestimulated with Cym50138 and activated with CpG-A, followed by IFN-α measurement and elucidation of specific ISGs at mRNA level. As PBMCs depleted of pDCs only produce minor amounts of IFN-α (<2 pg/ml) in response to CpG-A compared with whole PBMC cultures containing pDCs (>10,000 pg/ml) (Fig. 2A), we presume that pDCs are the main source of IFN-α. TLR9-induced cytokine secretion was also significantly reduced in whole PBMC cultures upon Cym50138 addition (Fig. 2B). Next, we analyzed three ISGs for their expression after CpG-A treatment, with or without Cym50138 prestimulation. The expression of the ISGs Mx1, IFN-induced dsRNA-activated protein kinase, and IRF7 were upregulated after CpG-A addition. Preadministration of Cym50138 did not reduce their expression, rather inducing a slight but insignificant upregulation (Fig. 2C). Therefore, we conclude that reduced IFN-α levels did not translate into reduced antiviral activity in our experimental setup.

FIGURE 2.

Attenuated IFN-α secretion does not influence the expression of IFN-inducible genes. (A) PBMCs with (− pDCs) or without (+ pDCs) pDC depletion were activated with CpG-A for 16 h, and IFN-α secretion was measured using CBA Flex Sets. Data are means ± SEM of seven independent experiments. (B) Whole PBMC cultures were controls or treated with CpG-A (2.5 μg/ml) for 16 h with or without prestimulation with Cym50318 for 30 min. IFN-α secretion in all samples was measured using CBA Flex Sets. Data are means ± SEM of 13 independent experiments. (C) Expression of IFN-α–inducible genes and actin were quantified by quantitative RT-PCR. Data are means ± SEM of seven independent experiments. p values were calculated using one-sample t test (B and C) or Student t test (A). *p ≤ 0.05, **p ≤ 0.01.

FIGURE 2.

Attenuated IFN-α secretion does not influence the expression of IFN-inducible genes. (A) PBMCs with (− pDCs) or without (+ pDCs) pDC depletion were activated with CpG-A for 16 h, and IFN-α secretion was measured using CBA Flex Sets. Data are means ± SEM of seven independent experiments. (B) Whole PBMC cultures were controls or treated with CpG-A (2.5 μg/ml) for 16 h with or without prestimulation with Cym50318 for 30 min. IFN-α secretion in all samples was measured using CBA Flex Sets. Data are means ± SEM of 13 independent experiments. (C) Expression of IFN-α–inducible genes and actin were quantified by quantitative RT-PCR. Data are means ± SEM of seven independent experiments. p values were calculated using one-sample t test (B and C) or Student t test (A). *p ≤ 0.05, **p ≤ 0.01.

Close modal

Besides conventional DCs (cDCs), also pDCs are considered as APCs that can induce T cell proliferation/activation (12, 25). The quality of the resulting T cell response depends on the pDC activation state and microenvironment (12, 2527). We asked whether S1P alters surface expression of costimulatory molecules on pDCs and concomitant T cell activation. Activation of human primary pDCs with CpG-A for 16 h increased MHC class I (MHC I) but not MHC II surface expression (Fig. 3A). Neither MHC I nor MHC II expression was altered upon S1P treatment (Fig. 3A). Furthermore, we analyzed costimulatory molecules on pDCs (Fig. 3B). The unaltered expression of CD80, CD83, CD86, CD40, and OX40L after S1P treatment suggested that S1P did not affect APC potential of pDCs. GrB production by pDCs is downregulated upon TLR9 activation to support T cell proliferation (28, 29). Freshly isolated pDCs did not express GrB (not shown), whereas IL-3 induced strong GrB expression (Fig. 3C). Administration of the S1PR4 agonist together with IL-3 induced GrB expression slightly but not significantly. Supplying CpG-A to IL-3 and Cym50138-treated cells failed to alter intracellular GrB expression compared with the corresponding control (Fig. 3C). These data suggested that S1P, via S1PR4, did not influence the Ag-presenting capacity of pDCs upon CpG-A treatment. CpG-A strongly induces IFN-α production by pDCs, but has only minor effects on costimulatory molecule expression, which is strongly induced by CpG-B oligonucleotides (30). Therefore, we analyzed whether S1PR4 signaling would affect the APC potential of CpG-B–activated pDCs. As expected, CpG-B induced strong upregulation of CD80, CD83 and CD86 compared with CpG-A (Fig. 3D, 3E). However, pretreatment with the S1PR4 agonist Cym50138 had no impact on CpG-B–induced expression of these molecules (Fig. 3E). Taken together, S1P signaling through S1PR4 unlikely alters the APC capacity of pDCs.

FIGURE 3.

S1PR4 signaling has no impact on APC properties of pDCs. (AE) Human pDCs were isolated from buffy coats and cultured in X-Vivo medium containing IL-3 (20 ng/ml). (A and B) Human pDCs were controls or cultured with CpG-A (5 μg/ml) for 16 h, with or without Cym50138 (1 μmol) prestimulation for 30 min. Surface expression of the indicated proteins was measured by FACS. CpG-A–treated group set to 1. Data are means ± SEM of three independent experiments for each protein. (C) Expression of GrB by pDCs was measured by intracellular Ab staining and FACS. The IL-3–treated (20 ng/ml) group was used as control and set to 1. Data are means ± SEM of four independent experiments. (D and E) Human pDCs were controls or cultured with CpG-B (1 μmol) for 16 h, with or without Cym50138 (200 nmol) prestimulation for 30 min. Surface expression of the indicated proteins was measured by FACS. (D) Representative FACS plots of resting versus CpG-B–activated pDCs are shown. (E) Quantification of FACS data. CpG-B–treated group set to 1. Data are means ± SEM of six independent experiments. The p values were calculated using one-sample t test. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. MFI, mean fluorescence intensity.

FIGURE 3.

S1PR4 signaling has no impact on APC properties of pDCs. (AE) Human pDCs were isolated from buffy coats and cultured in X-Vivo medium containing IL-3 (20 ng/ml). (A and B) Human pDCs were controls or cultured with CpG-A (5 μg/ml) for 16 h, with or without Cym50138 (1 μmol) prestimulation for 30 min. Surface expression of the indicated proteins was measured by FACS. CpG-A–treated group set to 1. Data are means ± SEM of three independent experiments for each protein. (C) Expression of GrB by pDCs was measured by intracellular Ab staining and FACS. The IL-3–treated (20 ng/ml) group was used as control and set to 1. Data are means ± SEM of four independent experiments. (D and E) Human pDCs were controls or cultured with CpG-B (1 μmol) for 16 h, with or without Cym50138 (200 nmol) prestimulation for 30 min. Surface expression of the indicated proteins was measured by FACS. (D) Representative FACS plots of resting versus CpG-B–activated pDCs are shown. (E) Quantification of FACS data. CpG-B–treated group set to 1. Data are means ± SEM of six independent experiments. The p values were calculated using one-sample t test. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. MFI, mean fluorescence intensity.

Close modal

Cytokines act in synergy with presented Ags and costimulatory molecules to support T cell activation and proliferation. IFN-α directly affects CD8+ T cell responses by maintaining their proliferation (31). Despite the fact that S1PR4 signaling had no influence on costimulatory molecule expression on pDCs, we asked if altered IFN-α levels affected T cell activation. We used a T cell activation and proliferation assay in which cocultured pDCs were pulsed with TTX. Cytokine secretion (IFN-α) of pDCs was induced by CpG-A and modulated by the addition of Cym50138. TTX served as a natural Ag, presented by pDCs via MHCs to induce T cell proliferation. As pDCs can induce T cell anergy in coculture systems, which can be reversed by adding IL-2 (32), we supplemented adequate IL-2 levels. pDCs were washed before adding them to T cells to prohibit a direct contact of T cells with the Ag or Cym50138. Proliferation of CD3+ T cells was measured 5 d after coculture by monitoring eFluor670 proliferation dye dilution by FACS (Fig. 4A, 4B). Compared to the negative control, activated and Ag-loaded pDCs induced moderate T cell proliferation, but Cym50138 prestimulation of pDCs failed to mediate a significant change in T cell proliferation. In general, APCs are able to prime naive cells into different effector cells. For instance, IFN-α induces Th1 responses, which are characterized by IFN-γ secretion (3335). Moreover, priming of naive T cells into IL-10 producing Tregs by alternatively activated pDCs is discussed. Based on this discrepancy we asked, if altered IFN-α levels in pDC/T cell cocultures may alter the differentiation of cocultured T cells. To address this question, we measured cytokine levels (IL-2, IL-10, and IFN-γ) in coculture supernatants. Besides IL-2, as a marker for T cell proliferation (which we supplied), all other cytokines were induced in cocultures with activated and Ag-loaded pDCs (Fig. 4C). Interestingly, IL-10 was strongly induced in the Cym50138-treated group, whereas IFN-γ levels were diminished compared with CpG-A only–treated cells. To gain further evidence that IFN-α production rather than Ag presentation or costimulation were involved in altered T cell differentiation by S1PR4-stimulated pDCs, we employed an approach in which isolated T cells were preactivated by stimulating CD2, CD3, and CD28 and subsequently pulsed daily with pDC supernatants for 5 d. Also in this setting, supernatants of Cym50138 plus CpG-A–stimulated pDCs resulted in increased IL-10, but decreased IFN-γ production by T cells in comparison with supernatants of only CpG-A stimulated pDCs (Fig. 4D). Importantly, readdition of IFN-α largely prevented these alterations in cytokine production (Fig. 4D). Taken together, S1PR4 signaling in pDCs translated into altered T cell differentiation, which was largely dependent on reduced IFN-α production.

FIGURE 4.

Influence of pDC S1PR4 signaling on T cell proliferation and activation. (AC) T cells and pDCs were isolated from buffy coats of the same donor. T cells were cultured overnight in media containing IL-2 (10 ng/ml). Untreated pDCs were controls, or pDCs were pulsed for 24 h with TTX (2 μg/ml). After pulsing and prestimulation with Cym50138 for 30 min, pDCs were washed with PBS and resuspended in T cell media containing IL-3 and IL-2, with or without the addition of CpG-A (5 μg/ml). (A and B) To measure T cell proliferation, T cells were prestained with a proliferation dye (eFluor 670; 5 μmol) and seeded on pDCs in a ratio of 1:5 (DC/T) for 5 d. Gray bars are controls; black bar is Cym50138-pretreated cells. Proliferation (eFluor 670 dilution) was measured by flow cytometry. (A) The gating strategy is displayed. Single and clustered lymphocytes were analyzed for CD3 (T cells) versus CD123 (pDCs, usually absent after 5 d coculture) expression. Dilution of eFluor 670 indicating proliferation of CD3+ T cells was analyzed to identify cells that had undergone at least one cycle of proliferation. (B) Quantification of proliferating T cells. Data are means ± SEM of five independent experiments. (C) Cocultures supernatants were harvested at day 5, and cytokine secretion was measured using CBA. Data are means ± SEM of five independent experiments. (D) Primary human T cells were stained with eFluor 670 and preactivated using human CD3/CD28/CD2 T Cell Activator, seeded in 96-well plates (5 × 105 T cells in 100 μl T cell medium), and pulsed daily with 50 μl primary pDC supernatants for 5 d. Supernatants were harvested at day 5, and cytokine secretion was measured using CBA. Data are means ± SEM of six independent experiments. For all experiments, the CpG-A–activated group was set to 1. The p values were calculated using one-sample t test (B–D). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.

FIGURE 4.

Influence of pDC S1PR4 signaling on T cell proliferation and activation. (AC) T cells and pDCs were isolated from buffy coats of the same donor. T cells were cultured overnight in media containing IL-2 (10 ng/ml). Untreated pDCs were controls, or pDCs were pulsed for 24 h with TTX (2 μg/ml). After pulsing and prestimulation with Cym50138 for 30 min, pDCs were washed with PBS and resuspended in T cell media containing IL-3 and IL-2, with or without the addition of CpG-A (5 μg/ml). (A and B) To measure T cell proliferation, T cells were prestained with a proliferation dye (eFluor 670; 5 μmol) and seeded on pDCs in a ratio of 1:5 (DC/T) for 5 d. Gray bars are controls; black bar is Cym50138-pretreated cells. Proliferation (eFluor 670 dilution) was measured by flow cytometry. (A) The gating strategy is displayed. Single and clustered lymphocytes were analyzed for CD3 (T cells) versus CD123 (pDCs, usually absent after 5 d coculture) expression. Dilution of eFluor 670 indicating proliferation of CD3+ T cells was analyzed to identify cells that had undergone at least one cycle of proliferation. (B) Quantification of proliferating T cells. Data are means ± SEM of five independent experiments. (C) Cocultures supernatants were harvested at day 5, and cytokine secretion was measured using CBA. Data are means ± SEM of five independent experiments. (D) Primary human T cells were stained with eFluor 670 and preactivated using human CD3/CD28/CD2 T Cell Activator, seeded in 96-well plates (5 × 105 T cells in 100 μl T cell medium), and pulsed daily with 50 μl primary pDC supernatants for 5 d. Supernatants were harvested at day 5, and cytokine secretion was measured using CBA. Data are means ± SEM of six independent experiments. For all experiments, the CpG-A–activated group was set to 1. The p values were calculated using one-sample t test (B–D). *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001.

Close modal

Next, we were interested in the mechanism by which S1P, via S1PR4, blocks IFN-α production in pDCs. To address this question, we first analyzed the binding and uptake of FITC-labeled CpG-A by human primary pDCs. The first steps of endosomal TLR7/9 activation are the recognition and uptake of the ligand, followed by intracellular trafficking to early endosomes. Because only one CpG uptake receptor (DEC-205) has been described (4), we analyzed its surface expression on human primary pDCs. As shown in Fig. 5A, human blood pDCs express DEC-205. However, activation with CpG-A for different time points did not result in receptor internalization (Fig. 5B). Recently, it was shown that CpG ODNs enter the cell rapidly, and colocalization of CpG ODNs with TLR9 within early endosomes takes place 5 min after treatment (36). Time kinetics of CpG-A-FITC binding/uptake in human primary pDCs by FACS showed that CpG-A-FITC binds to pDCs during the first 5 min of activation (Fig. 5C). However, there was no difference between the control group (Fig. 5C, black line) and Cym50138 prestimulated cells (Fig. 5C, red line). Moreover, we also failed to detect differences in intracellular accumulation of FITC-labeled CpG-A by immunofluorescence analysis (Fig. 5D). A next option was the availability of the receptor for CpG-A. Recently, it was shown that TLR9 traffics from the Golgi apparatus to the plasma membrane, followed by shuttling to the endosomes, where ligand attachment takes place (37). Thus, enhanced TLR9 accumulation at the plasma membrane might be a marker of cells responding weaker to TLR9 ligands. However, S1PR4 signaling did not induce TLR9 expression at the plasma membrane, which was observed upon CpG-A administration, indicating recycling of the activated receptor. Rather, S1PR4 activation reduced TLR9 accumulation, thus being another marker (besides IFN-α production) of delayed/reduced cell activation in the presence of an S1PR4 agonist (Supplemental Fig. 1, black bar). Primary pDCs were reported to acquire a more mature phenotype after isolation upon overnight culture in vitro, which affects IFN-α production (38). Thus, we wondered whether S1PR4 signaling affects human pDC maturation. After overnight culture indeed two pDC subpopulations could be distinguished based on their light scatter profile as reported before (Supplemental Fig. 2A) (38). Interestingly, CpG-A uptake was higher in the reported mature (forward light scatterlowside scatterhigh) pDC subpopulation (Supplemental Fig. 2B). Adding CpG-A to freshly isolated pDCs also increased the forward light scatterlowside scatterhigh pDC population compared with control cells. However, S1PR4 signaling slightly, but not significantly, decreased this population (Supplemental Fig. 2C, 2D), pointing to an S1PR4-independent in vitro maturation and arguing against maturation as the underlying principle of S1PR4-dependent IFN-α suppression.

FIGURE 5.

Influence of S1PR4 signaling on CpG uptake. (AD) Human pDCs were isolated from buffy coats and cultured in X-Vivo medium containing IL-3 (20 ng/ml). (A) Expression of the CpG uptake receptor DEC-205 on human primary pDCs was analyzed by FACS. Data are representative of four independent experiments. (B) DEC-205 surface expression on primary pDCs was analyzed after different time points of CpG-A treatment. Untreated cells were set to 1. Data are means ± SEM of five independent experiments. (C) Human primary pDCs were controls or prestimulated with Cym50138 for 30 min, followed by activation for different time points with FITC-labeled CpG-A and FACS analysis. Data are means ± SEM of five independent experiments. (D) Immunofluorescence staining of human primary pDCs. Cells were incubated for 5 min with FITC-labeled CpG-A (green) with or without Cym50138 prestimulation, followed by fixation, DAPI (blue) staining, and immunofluorescence imaging (original magnification ×640). Data are representative of three independent experiments. MFI, mean fluorescence intensity.

FIGURE 5.

Influence of S1PR4 signaling on CpG uptake. (AD) Human pDCs were isolated from buffy coats and cultured in X-Vivo medium containing IL-3 (20 ng/ml). (A) Expression of the CpG uptake receptor DEC-205 on human primary pDCs was analyzed by FACS. Data are representative of four independent experiments. (B) DEC-205 surface expression on primary pDCs was analyzed after different time points of CpG-A treatment. Untreated cells were set to 1. Data are means ± SEM of five independent experiments. (C) Human primary pDCs were controls or prestimulated with Cym50138 for 30 min, followed by activation for different time points with FITC-labeled CpG-A and FACS analysis. Data are means ± SEM of five independent experiments. (D) Immunofluorescence staining of human primary pDCs. Cells were incubated for 5 min with FITC-labeled CpG-A (green) with or without Cym50138 prestimulation, followed by fixation, DAPI (blue) staining, and immunofluorescence imaging (original magnification ×640). Data are representative of three independent experiments. MFI, mean fluorescence intensity.

Close modal

To avoid an overshooting IFN-I response, IFN-α production by pDCs is highly regulated. Various regulatory surface receptors on pDCs signal via ITAM/ITIM pathways that involve members of the Src kinase family as downstream signaling modules (6, 39). Some of these receptors undergo internalization after ligand binding (reviewed in Ref. 1). To elucidate the mechanism behind the decreased IFN-α production downstream of S1PR4 activation, we analyzed, among others, the surface expression of the inhibitory receptor ILT7 on primary blood pDCs by FACS (Fig 6A). Unexpectedly, ILT7 surface expression rapidly (5 min) decreased after CpG-A stimulation (Fig. 6B, black line) and reappeared after additional 5 min. In contrast, pDCs prestimulated with Cym50138 and activated with CpG-A did not show any changes in ILT7 surface expression during uptake of the TLR ligand (Fig. 6B, red line, and 6C, 6D). These findings were confirmed by immunofluorescence microscopy (Fig. 6E). Although a decrease in ILT7 mRNA after pDC activation was observed before (5), long-term activation (16 h) of pDCs with CpG-A had no effect on ILT7 surface expression (Supplemental Fig. 3A). Because the ligand for ILT7, BST2, is expressed on human blood pDCs, we investigated if the expression of BST2 was affected in our experimental setup. BST2 is an IFN-I–induced gene. Hence, surface expression was upregulated after long-time CpG-A stimulation (16 h) (Supplemental Fig. 3B). Although S1PR4 signaling (30 min) slightly increased BST2 surface expression, activation of S1PR4 in combination with TLR9 activation failed to alter BST2 expression (Supplemental Fig. 3B), indicating that alterations in BST2 expression did not contribute to S1PR4-dependent IFN-α suppression. We therefore hypothesized that the internalization of ILT7 was required for CpG-A–dependent IFN-α production and that inhibition of this process was the primary mechanism by which S1PR4 attenuated IFN-α production. A requirement for such a mechanism would be that altered ILT7 expression in the observed short time frame translated into differences in ILT7 receptor engagement. We approached this question by asking whether accumulation of pDC in cell clusters, indicative of enhanced receptor/ligand interaction, was altered after short-term CpG-A treatment with or without S1PR4 activation. Human PBMCs were therefore prestimulated with Cym50138 followed by activation with CpG-A for 5 min, fixation, and flow cytometric analysis (Fig. 6F). Indeed, we observed a small but very consistent decrease in pDC accumulation in cell clusters at this early time point after CpG-A treatment, which was prevented by S1PR4 activation (Fig. 6G, 6H). These data may reflect ILT7 internalization.

FIGURE 6.

CpG-A–induced ILT7 internalization is counteracted by S1PR4. (A) Gating strategy to determine ILT7 expression on pDCs in whole PBMCs (as shown) or after negative selection from buffy coats and culture in X-Vivo medium containing IL-3 (20 ng/ml). Light scatter gating focuses roughly on the region between lymphocyte and monocyte population where DCs reside. pDCs are gated based on CD123 expression, but lack of CD11c expression. Finally, ILT7-expressing pDCs among CD123+ cells are identified. (B) Time kinetics of ILT7 surface expression on human primary pDCs with or without Cym50138 prestimulation for 30 min followed by CpG-A (5 μg/ml) treatment, determined by FACS, are shown. CpG-A–untreated groups (control [ctrl] and Cym50138 treated) were set to 1. Data are means ± SEM of five independent experiments. (C and D) Quantification of ILT7 surface expression by FACS 5 min after CpG-A (5 μg/ml) activation, with or without Cym50138 prestimulation for 30 min. (C) Representative FACS histograms are shown. (D) Quantification of ILT7 surface expression. The untreated group was set to 1. Data are means ± SEM of six independent experiments. (E) Immunofluorescence staining of ILT7 in human primary pDCs. Cells were controls or incubated for 5 min with FITC-labeled CpG-A (green) with or without Cym50138 prestimulation for 30 min, followed by fixation, ILT7 (red) and DAPI (blue) staining, and immunofluorescence imaging. Data are representative of three independent experiments. (FH) Human PBMCs were fixed 5 min after CpG-A (5 μg/ml) activation, with or without Cym50138 prestimulation for 30 min to determine pDC accumulation in cell clusters by flow cytometry. (F) Gating strategy is displayed. Doublets were determined by light scatter analysis. CD123-expressing CD11c cells were quantified. (G) Representative FACS plots of CD123+ cell accumulation in doublets are shown. (H) Quantification of CD123+ cell accumulation in doublets. The CpG-A–treated group was set to 1. Data are means ± SEM of six independent experiments. The p values were calculated using one-sample t test (H), Student t test (D), or two-way ANOVA with Bonferroni correction (B). *p > 0.05, **p > 0.01. MFI, mean fluorescence intensity.

FIGURE 6.

CpG-A–induced ILT7 internalization is counteracted by S1PR4. (A) Gating strategy to determine ILT7 expression on pDCs in whole PBMCs (as shown) or after negative selection from buffy coats and culture in X-Vivo medium containing IL-3 (20 ng/ml). Light scatter gating focuses roughly on the region between lymphocyte and monocyte population where DCs reside. pDCs are gated based on CD123 expression, but lack of CD11c expression. Finally, ILT7-expressing pDCs among CD123+ cells are identified. (B) Time kinetics of ILT7 surface expression on human primary pDCs with or without Cym50138 prestimulation for 30 min followed by CpG-A (5 μg/ml) treatment, determined by FACS, are shown. CpG-A–untreated groups (control [ctrl] and Cym50138 treated) were set to 1. Data are means ± SEM of five independent experiments. (C and D) Quantification of ILT7 surface expression by FACS 5 min after CpG-A (5 μg/ml) activation, with or without Cym50138 prestimulation for 30 min. (C) Representative FACS histograms are shown. (D) Quantification of ILT7 surface expression. The untreated group was set to 1. Data are means ± SEM of six independent experiments. (E) Immunofluorescence staining of ILT7 in human primary pDCs. Cells were controls or incubated for 5 min with FITC-labeled CpG-A (green) with or without Cym50138 prestimulation for 30 min, followed by fixation, ILT7 (red) and DAPI (blue) staining, and immunofluorescence imaging. Data are representative of three independent experiments. (FH) Human PBMCs were fixed 5 min after CpG-A (5 μg/ml) activation, with or without Cym50138 prestimulation for 30 min to determine pDC accumulation in cell clusters by flow cytometry. (F) Gating strategy is displayed. Doublets were determined by light scatter analysis. CD123-expressing CD11c cells were quantified. (G) Representative FACS plots of CD123+ cell accumulation in doublets are shown. (H) Quantification of CD123+ cell accumulation in doublets. The CpG-A–treated group was set to 1. Data are means ± SEM of six independent experiments. The p values were calculated using one-sample t test (H), Student t test (D), or two-way ANOVA with Bonferroni correction (B). *p > 0.05, **p > 0.01. MFI, mean fluorescence intensity.

Close modal

Our hypothesis gained further indirect support by the observation that not all TLR ligands altered ILT7 surface expression on human blood pDCs. Activation of whole PBMCs with another TLR9 ligand, CpG-B (CpG2006), provoked ILT7 internalization on pDCs, whereas TLR7 activation with IMQ did not (Fig. 7A). Interestingly, a more physiological TLR7 ligand, the tick-borne encephalitis vaccine (FSME), promoted a rapid and prolonged ILT7 internalization (Fig. 7B, 7C). ILT7 internalization by CpG-B or FSME was reversed by S1PR4 activation, whereas ILT7 levels on IMQ-treated cells were not altered by S1PR4 signaling (Fig. 7A–C). Importantly, for all used TLR ligands, ILT7 internalization was inversely correlated with IFN-α production. Both CpG-A and CpG-B, as well as the tick-borne encephalitis vaccine FSME, induced robust amounts of extracellular IFN-α, whereas IMQ administration elicited low IFN-α secretion (Fig. 7D). IFN-α levels induced by CpG-B or FSME were decreased by Cym50138. In contrast, pDCs prestimulated with Cym50138 and activated with IMQ produced equal amounts of IFN-α compared with only IMQ-treated cells (Fig. 7E). These data suggest that unaltered ILT7 expression after IMQ stimulation might be responsible for low IFN-α production compared with TLR ligands that induce ILT7 internalization. In contrast, S1PR4 signaling only prevents IFN-α secretion triggered by ligands that induce ILT7 depletion from the cell surface. To support this hypothesis, we blocked ILT7 downstream signaling by inhibiting of the tyrosine-protein kinase Lyn. As shown in Fig. 7F, Cym50138-pretreated cells decreased CpG-A–induced IFN-α production compared with control pDCs. However, preadministration of an LPI for 30 min blocked the effect of S1PR4 signaling on IFN-α secretion. Lyn is not exclusively activated downstream of ILT7, but also other inhibitory pDC receptors such as CD303 (40). Therefore, extensive future studies will be required to link these molecules in the context of our setting. Finally, we approached signaling pathways downstream of S1PR4 preactivation (Supplemental Figure 4) that are involved in blocking ILT7 internalization following pDC activation. S1PR4 couples to Gα12/13, which triggers activation of the RhoA/ROCK pathway that is, among others, involved in vesicle trafficking and membrane dynamics. Blocking of RhoA as well as ROCK with pharmacological inhibitors in combination with Cym50138 indeed restored ILT7 internalization after pDC activation with CpG-A (Fig. 7G). Unfortunately, RhoA/ROCK inhibition prevented CpG-A–induced IFN-α secretion. Therefore, these ILT7 expression data could not be correlated with IFN-α levels.

FIGURE 7.

S1PR4-dependent regulation of ILT7 surface expression is linked to IFN-α regulation. (A) pDCs were untreated or prestimulated with Cym50138 for 30 min and activated with the TLR9 ligand CpG-B (1 μmol) or the TLR7 ligand IMQ (2.5 μg/ml) for 5 min. Untreated cells were controls. ILT7 surface expression was measured by FACS. Data are means ± SEM of five independent experiments for each TLR ligand. (B) ILT7 surface expression on pDCs was measured at different time points following activation with an FSME vaccine (10% v/v; signals through TLR7). Data are means ± SEM of six independent experiments. (C) pDCs were untreated or prestimulated with Cym50138 for 30 min and activated with FSME (10% v/v) for 5 min. Untreated cells were controls. ILT7 surface expression was measured by FACS. Data are means ± SEM of six independent experiments. (D) IFN-α levels in pDC supernatants after activation with different TLR ligands (IMQ, 2.5 μg/ml; CpG-A, 5 μg/ml; CpG-B, 1 μmol; and FSME, 10% v/v) were measured using CBA Flex sets. Data are means ± SEM of six independent experiments. (E) pDCs were activated with the TLR9 ligand CpG-B (1 μmol) and the TLR7 ligands FSME (10% v/v) or IMQ (2.5 μg/ml), with or without Cym50138 (200 nmol) prestimulation for 30 min. IFN-α levels were measured by CBA. The TLR ligand–treated group was set to 1 each. Data are means ± SEM of at least three independent experiments for each TLR ligand. (F) Human primary pDCs were prestimulated with LPI (10 μmol) to block ILT7 downstream signaling in combination with Cym50138 for 30 min and activated overnight (16 h) with CpG-A. IFN-α levels were measured using CBA. (G) Quantification of ILT7 surface expression on human pDCs by FACS analysis 5 min after CpG-A (5 μg/ml) activation, with or without prestimulation with Cym50138 (200 nmol), the RhoA inhibitor Rhosin (10 μmol), or the ROCK inhibitor Y-27632 (10 μmol) for 30 min. Data are means ± SEM of six independent experiments. Data are means ± SEM of four independent experiments. The p values were calculated using one-sample t test (E), Student t test (A, C, F, and G), or one-way ANOVA with Bonferroni correction (B). *p > 0.05, **p > 0.01. MFI, mean fluorescence intensity.

FIGURE 7.

S1PR4-dependent regulation of ILT7 surface expression is linked to IFN-α regulation. (A) pDCs were untreated or prestimulated with Cym50138 for 30 min and activated with the TLR9 ligand CpG-B (1 μmol) or the TLR7 ligand IMQ (2.5 μg/ml) for 5 min. Untreated cells were controls. ILT7 surface expression was measured by FACS. Data are means ± SEM of five independent experiments for each TLR ligand. (B) ILT7 surface expression on pDCs was measured at different time points following activation with an FSME vaccine (10% v/v; signals through TLR7). Data are means ± SEM of six independent experiments. (C) pDCs were untreated or prestimulated with Cym50138 for 30 min and activated with FSME (10% v/v) for 5 min. Untreated cells were controls. ILT7 surface expression was measured by FACS. Data are means ± SEM of six independent experiments. (D) IFN-α levels in pDC supernatants after activation with different TLR ligands (IMQ, 2.5 μg/ml; CpG-A, 5 μg/ml; CpG-B, 1 μmol; and FSME, 10% v/v) were measured using CBA Flex sets. Data are means ± SEM of six independent experiments. (E) pDCs were activated with the TLR9 ligand CpG-B (1 μmol) and the TLR7 ligands FSME (10% v/v) or IMQ (2.5 μg/ml), with or without Cym50138 (200 nmol) prestimulation for 30 min. IFN-α levels were measured by CBA. The TLR ligand–treated group was set to 1 each. Data are means ± SEM of at least three independent experiments for each TLR ligand. (F) Human primary pDCs were prestimulated with LPI (10 μmol) to block ILT7 downstream signaling in combination with Cym50138 for 30 min and activated overnight (16 h) with CpG-A. IFN-α levels were measured using CBA. (G) Quantification of ILT7 surface expression on human pDCs by FACS analysis 5 min after CpG-A (5 μg/ml) activation, with or without prestimulation with Cym50138 (200 nmol), the RhoA inhibitor Rhosin (10 μmol), or the ROCK inhibitor Y-27632 (10 μmol) for 30 min. Data are means ± SEM of six independent experiments. Data are means ± SEM of four independent experiments. The p values were calculated using one-sample t test (E), Student t test (A, C, F, and G), or one-way ANOVA with Bonferroni correction (B). *p > 0.05, **p > 0.01. MFI, mean fluorescence intensity.

Close modal

Together, our data suggest a mechanism of S1PR4-dependent IFN-α suppression (Fig. 8). S1PR4 signaling through the RhoA/ROCK pathway preserves surface expression of the inhibitory receptor ILT7. Increased ILT7 signaling consequently limits IFN-α production.

FIGURE 8.

Proposed mechanism of ILT7 regulation during pDC activation and the impact of S1PR4 signaling. (1) IMQ enters the cells via diffusion, reaches endosomes, and triggers IFN-α production via TLR7. This results in moderate cytokine levels, due to ILT7 surface expression, concomitant ligand (e.g., BST2) engagement, and a negative signaling input via Lyn kinase. (2) TLR ligands (TLR-L), such as CpGs or viral particles, are taken up by receptor-mediated endocytosis, thereby inducing ILT7 internalization, resulting in massive IFN-α production. (3) S1PR4 activation stabilizes ILT7 at the cell surface. ILT7 binds its ligand on bystander cells, thereby reducing IFN-α levels.

FIGURE 8.

Proposed mechanism of ILT7 regulation during pDC activation and the impact of S1PR4 signaling. (1) IMQ enters the cells via diffusion, reaches endosomes, and triggers IFN-α production via TLR7. This results in moderate cytokine levels, due to ILT7 surface expression, concomitant ligand (e.g., BST2) engagement, and a negative signaling input via Lyn kinase. (2) TLR ligands (TLR-L), such as CpGs or viral particles, are taken up by receptor-mediated endocytosis, thereby inducing ILT7 internalization, resulting in massive IFN-α production. (3) S1PR4 activation stabilizes ILT7 at the cell surface. ILT7 binds its ligand on bystander cells, thereby reducing IFN-α levels.

Close modal

The sphingolipid S1P signals via G-protein–coupled receptors to affect inflammation. Whereas S1PR1 and 2 mainly regulate immune cell migration, the role of the immune cell-specific S1PR4 is ill defined (14). There is evidence that S1PR4 signaling modulates immune cell phenotypes, because a lack of S1PR4 in mouse cDCs reduces Th17 and increases Th2 immune responses (17). Moreover, S1PR4 activation on human monocyte-derived DCs induced IL-27 production to increase Treg-dependent suppression of cytotoxicity against human tumor cells (19). The present study suggests that S1PR4 signaling also regulates critical functions of human pDCs, by blocking IFN-I secretion.

Upon in vitro activation, pDCs rapidly produce significant amounts of IFN-α, which initiates a feed-forward loop that further increases IFN-α production at later time points (41, 42). The S1PR4-triggered decrease in IFN-α secretion was apparent already 4 h after TLR9 activation, pointing to a regulatory impact on the initial steps following TLR ligand uptake/recognition rather than on feed-forward signaling. This notion is supported by experiments with a reversed experimental setup (i.e., TLR9 activation followed by S1PR4 agonist stimulation), in which S1PR4 signaling failed to diminish IFN-α secretion (Supplemental Fig. 4). The first step leading to endosomal TLR activation is, for a variety of agonists, receptor-dependent uptake. For instance, viruses and bacteria typically enter the cell by receptor-mediated endocytosis or phagocytic pathways, followed by their degradation and the release of immunogenic nucleic acids. The uptake of synthetic TLR ligands such as CpG is also receptor mediated, although the underlying mechanisms are largely unknown. DEC-205, an Ag delivery receptor expressed by murine CD8+ DCs and different human leukocytes including pDCs (4345), was recently identified as an uptake receptor for CpG ODNs (4). However, DEC-205 was irrelevant at least for CpG-A uptake in human primary pDCs, because the receptor failed to internalize upon CpG-A administration in our experimental setup. Moreover, uptake of FITC-labeled CpG by the whole pDC population was not affected by S1PR4 signaling, indicating a regulatory impact downstream or independent of ligand uptake/recognition.

We observed that CpG was taken up at higher levels by a subpopulation of primary human pDCs, which was, to our knowledge, not reported before. This subpopulation corresponds to pDCs showing increased maturation marker expression, which are generated upon in vitro culture of primary human pDCs isolated from blood and further expanded following TLR9 ligation (38) (Supplemental Fig. 2). An S1PR4-dependent alteration of this pDC subset was not apparent (Supplemental Fig. 2C). However, we noticed an impact of S1PR4 signaling on ILT7 expression, which was proposed as a maturation marker of human pDC in vitro (38). We observed rapid ILT7 internalization after CpG-A administration, which was prevented by S1PR4 signaling and coupled to IFN-α production. By using different TLR ligands, we confirmed the impact of S1PR4 on ILT7 trafficking and concomitant IFN-α regulation. Strikingly, only stimulation with TLR ligands that are taken up by receptor-mediated endocytosis (FSME vaccine or CpG ODNs) triggered ILT7 internalization and were responsive to S1PR4-dependent IFN-α suppression. In contrast, IMQ, which enters the cell via diffusion through the plasma membrane, failed to alter ILT7 surface expression, and IMQ-dependent IFN-α production was not regulated by S1PR4. These data allow proposing a model of ILT7-dependent IFN-I regulation. ILT7, when expressed at the cell surface, binds its ligand BST2 (e.g., on bystander pDCs), thereby limiting IFN-α production. Full-blown pDC activation requires ILT7 internalization, which is coupled to endocytosis of TLR7/9 ligands via an unknown mechanism, to prevent ILT7 interaction with its ligand and downstream inhibitory signaling (Fig. 8). Interestingly, ILT7 internalization was prolonged when using an immobilized virus instead of synthetic TLR ligands, thus strengthening the physiological relevance of this mechanism during pDC activation.

S1PR4 signaling was previously coupled to receptor trafficking. We observed that production of inflammatory cytokines by tumor-associated macrophages required shuttling of the nerve growth factor receptor TrKA to the plasma membrane, which was initiated by S1P and S1PR4 (18). Membrane trafficking of cell-surface receptors involves alterations of actin dynamics. Accordingly, S1PR4 predominantly couples to G12/13 that activates Rho and regulates cell shape and stress fiber formation (46). Indeed, inhibiting Rho and downstream ROCK activity prevented the impact of S1PR4 on ILT7 trafficking. Future studies will provide further mechanistic insight as to how S1PR4 signals locally prevent ILT7 internalization and whether it involves the activation of known immunomodulatory mediator systems that are known to affect IFN-α production by pDC. Recently, it was shown that other G-protein–coupled receptor ligands, such as PGE2, can reduce IFN-α production by pDCs (4749). PGE2 in immune cells signals mostly via EP2 or EP4, which couple to Gs to elevate cAMP. Interestingly, G12/13 are also capable of increasing cAMP via adenylate cyclase 7 that is highly expressed in human primary pDCs (50). It will be interesting to determine whether cAMP signaling contributes to inhibiting IFN-α production downstream of S1PR4.

The defense against viral infection is a hallmark of pDC function (51). Hence, production of IFN-I in response to viral components via TLR7/9 is indispensable to ensure upregulation of antiviral IFN-stimulated genes. In our setup, decreased IFN-α levels after S1PR4 activation were not translated into decreased mRNA levels of different ISGs, which might be due to the relatively high remaining IFN-α levels (mean >1000 pg/ml) and the high affinity of the IFN-α/β receptor 1/2 heterodimer for IFN-α. However, reduced IFN-α production was coupled to T cell polarization. The induction of adequate T cell responses requires the ability of APCs to process Ags, followed by precise presentation through the MHCs, the expression of costimulatory molecules, and, finally, a suitable milieu displayed by specific (inflammatory) cytokines (e.g., IFN-α and IL-12) (52). Human pDCs as APCs were shown to capture and cross-present viral Ags via MHC I to naive T cells, resulting in CD8+ T cell responses (53). Furthermore, human blood pDCs express several costimulatory and T cell–activating molecules, which are required for optimal T cell activation. S1PR4 activation failed to alter the expression of MHCs and costimulatory molecules or others regulators of T cell activation/proliferation such as GrB (29). Taken together, our data do not reveal an impact of S1PR4 on APC functions of human blood pDCs, corroborating a previous observation that S1PR4 did not affect Ag presentation by human cDCs (19). T cells require, besides Ag presentation and costimulation, a third signal to develop strong effector functions (52). IFN-α was shown to maintain proliferation of CD8+ T cells (54, 55). However, activation of S1PR4 signaling in pDCs and thus reducing IFN-α failed to alter T cell proliferation. Beside its role in lymphocyte activation, IFN-α modulates the outcome of T cell responses. It has been described that IFN-α directly affects human naive CD4+ T cells to favor their differentiation into IFN-γ–producing Th1 cells (33). Furthermore, IFN-α suppresses the expansion of regulatory T cells, while increasing IFN-γ–producing effector T cells (56). Triggering S1PR4 on Ag-pulsed and TLR-activated pDCs, followed by cocultivation with autologous T cells, shifted cytokine secretion from IFN-γ to IL-10, corroborating these previous reports. In this regard, S1PR4 signaling enables pDCs to adopt a regulatory phenotype inducing tolerogenic T cell responses.

Our findings illustrate one possible mechanism by which the tumor microenvironment, via enhanced S1P production, avoids tumor eradication by high levels of IFN-I (57, 58). In concordance with this hypothesis, tumor-associated pDCs show a partially activated phenotype and produce very low amounts of IFN-α compared with pDCs isolated from patient’s blood (59). Furthermore, high numbers of infiltrating pDCs in tumors were correlated with poor patient prognosis in ovarian cancer (60). Targeting the S1P–S1PR axis is a promising tool to overcome several diseases, including autoimmune disorders and cancer. There are many studies dealing with the modulation of S1PR1 in different autoimmune diseases (e.g., multiple sclerosis and rheumatoid arthritis), in which the therapeutic benefit is attributed to inhibition of immune cell trafficking (reviewed in Ref. 61). Interestingly, S1PR1 signaling did not affect pDC trafficking into diseased areas in a murine MS model, which proved beneficial for disease outcome (62). This was attributed to a low S1PR1 expression level in murine pDC, which selectively express S1PR4 (63). Whether the immunosuppressive effect of pDCs accumulating in the brain of experimental animals in the multiple sclerosis model is at least partially transmitted through S1PR4 remains to be determined. However, the current study and previous studies (1719) reveal a role for S1PR4 in modulating immune cell activation that is relevant in an inflammatory context. S1PR4 might be a promising therapeutic target to modulate pathogenic IFN-α production in IFN-I–driven diseases or to relieve a block of IFN-I production in tumors.

We thank Praveen Mathoor and Margarethe Mijatovic for excellent technical assistance.

This work is supported by the Else Kröner-Fresenius Foundation Research Training Group Translational Research Innovation–Pharma, the Sander Foundation (2013.036.01), the Deutsche Krebshilfe (110637), and by the Deutsche Forschungsgemeinschaft (SFB 1039 TP B04 and SFB 1039 TP B06).

The online version of this article contains supplemental material.

Abbreviations used in this article:

BST2

bone marrow stromal Ag 2

CBA

cytometric bead array

cDC

conventional DC

DC

dendritic cell

FSME

tick-borne encephalitis vaccine

GrB

granzyme B

IFN-I

type I IFN

IMQ

imiquimod

IRF7

IFN regulatory factor 7

ISG

IFN-I–stimulated gene

LPI

Lyn peptide inhibitor

MHC I

MHC class I

MHC II

MHC class II

ODN

oligodeoxynucleotide

pDC

plasmacytoid dendritic cell

qPCR

quantitative PCR

ROCK

Rho-associated protein kinase

siRNA

small interfering RNA

S1P

sphingosine-1-phosphate

S1PR

sphingosine-1-phosphate receptor

Treg

regulatory T cell

TTX

tetanus toxoid.

1
Swiecki
M.
,
Colonna
M.
.
2010
.
Unraveling the functions of plasmacytoid dendritic cells during viral infections, autoimmunity, and tolerance.
Immunol. Rev.
234
:
142
162
.
2
George
P. M.
,
Badiger
R.
,
Alazawi
W.
,
Foster
G. R.
,
Mitchell
J. A.
.
2012
.
Pharmacology and therapeutic potential of interferons.
Pharmacol. Ther.
135
:
44
53
.
3
Ito
T.
,
Kanzler
H.
,
Duramad
O.
,
Cao
W.
,
Liu
Y. J.
.
2006
.
Specialization, kinetics, and repertoire of type 1 interferon responses by human plasmacytoid predendritic cells.
Blood
107
:
2423
2431
.
4
Caminschi
I.
,
Meuter
S.
,
Heath
W. R.
.
2013
.
DEC-205 is a cell surface receptor for CpG oligonucleotides.
OncoImmunology
2
:
e23128
.
5
Ju
X. S.
,
Hacker
C.
,
Scherer
B.
,
Redecke
V.
,
Berger
T.
,
Schuler
G.
,
Wagner
H.
,
Lipford
G. B.
,
Zenke
M.
.
2004
.
Immunoglobulin-like transcripts ILT2, ILT3 and ILT7 are expressed by human dendritic cells and down-regulated following activation.
Gene
331
:
159
164
.
6
Cao
W.
,
Rosen
D. B.
,
Ito
T.
,
Bover
L.
,
Bao
M.
,
Watanabe
G.
,
Yao
Z.
,
Zhang
L.
,
Lanier
L. L.
,
Liu
Y. J.
.
2006
.
Plasmacytoid dendritic cell-specific receptor ILT7-Fc epsilonRI gamma inhibits Toll-like receptor-induced interferon production.
J. Exp. Med.
203
:
1399
1405
.
7
Tavano
B.
,
Boasso
A.
.
2014
.
Effect of immunoglobin-like transcript 7 cross-linking on plasmacytoid dendritic cells differentiation into antigen-presenting cells.
PLoS One
9
:
e89414
.
8
Cao
W.
,
Bover
L.
,
Cho
M.
,
Wen
X.
,
Hanabuchi
S.
,
Bao
M.
,
Rosen
D. B.
,
Wang
Y. H.
,
Shaw
J. L.
,
Du
Q.
, et al
.
2009
.
Regulation of TLR7/9 responses in plasmacytoid dendritic cells by BST2 and ILT7 receptor interaction.
J. Exp. Med.
206
:
1603
1614
.
9
Angelot-Delettre
F.
,
Biichle
S.
,
Ferrand
C.
,
Seilles
E.
,
Gaugler
B.
,
Harrivel
V.
,
Rosenthal-Allieri
M. A.
,
Deconinck
E.
,
Saas
P.
,
Garnache-Ottou
F.
.
2012
.
Intracytoplasmic detection of TCL1--but not ILT7-by flow cytometry is useful for blastic plasmacytoid dendritic cell leukemia diagnosis.
Cytometry A
81
:
718
724
.
10
Hartmann
E.
,
Wollenberg
B.
,
Rothenfusser
S.
,
Wagner
M.
,
Wellisch
D.
,
Mack
B.
,
Giese
T.
,
Gires
O.
,
Endres
S.
,
Hartmann
G.
.
2003
.
Identification and functional analysis of tumor-infiltrating plasmacytoid dendritic cells in head and neck cancer.
Cancer Res.
63
:
6478
6487
.
11
Zou
W.
.
2005
.
Immunosuppressive networks in the tumour environment and their therapeutic relevance.
Nat. Rev. Cancer
5
:
263
274
.
12
Tel
J.
,
Smits
E. L.
,
Anguille
S.
,
Joshi
R. N.
,
Figdor
C. G.
,
de Vries
I. J.
.
2012
.
Human plasmacytoid dendritic cells are equipped with antigen-presenting and tumoricidal capacities.
Blood
120
:
3936
3944
.
13
Rivera
J.
,
Proia
R. L.
,
Olivera
A.
.
2008
.
The alliance of sphingosine-1-phosphate and its receptors in immunity.
Nat. Rev. Immunol.
8
:
753
763
.
14
Gräler
M. H.
,
Bernhardt
G.
,
Lipp
M.
.
1998
.
EDG6, a novel G-protein-coupled receptor related to receptors for bioactive lysophospholipids, is specifically expressed in lymphoid tissue.
Genomics
53
:
164
169
.
15
Spiegel
S.
,
Milstien
S.
.
2003
.
Sphingosine-1-phosphate: an enigmatic signalling lipid.
Nat. Rev. Mol. Cell Biol.
4
:
397
407
.
16
Wang
W.
,
Graeler
M. H.
,
Goetzl
E. J.
.
2005
.
Type 4 sphingosine 1-phosphate G protein-coupled receptor (S1P4) transduces S1P effects on T cell proliferation and cytokine secretion without signaling migration.
FASEB J.
19
:
1731
1733
.
17
Schulze
T.
,
Golfier
S.
,
Tabeling
C.
,
Räbel
K.
,
Gräler
M. H.
,
Witzenrath
M.
,
Lipp
M.
.
2011
.
Sphingosine-1-phospate receptor 4 (S1P4) deficiency profoundly affects dendritic cell function and TH17-cell differentiation in a murine model.
FASEB J.
25
:
4024
4036
.
18
Ley
S.
,
Weigert
A.
,
Weichand
B.
,
Henke
N.
,
Mille-Baker
B.
,
Janssen
R. A.
,
Brüne
B.
.
2013
.
The role of TRKA signaling in IL-10 production by apoptotic tumor cell-activated macrophages.
Oncogene
32
:
631
640
.
19
Sekar
D.
,
Hahn
C.
,
Brüne
B.
,
Roberts
E.
,
Weigert
A.
.
2012
.
Apoptotic tumor cells induce IL-27 release from human DCs to activate Treg cells that express CD69 and attenuate cytotoxicity.
Eur. J. Immunol.
42
:
1585
1598
.
20
Sha
W.
,
Olesch
C.
,
Hanaka
H.
,
Rådmark
O.
,
Weigert
A.
,
Brüne
B.
.
2013
.
Necrosis in DU145 prostate cancer spheroids induces COX-2/mPGES-1-derived PGE2 to promote tumor growth and to inhibit T cell activation.
Int. J. Cancer
133
:
1578
1588
.
21
Davis
M. D.
,
Clemens
J. J.
,
Macdonald
T. L.
,
Lynch
K. R.
.
2005
.
Sphingosine 1-phosphate analogs as receptor antagonists.
J. Biol. Chem.
280
:
9833
9841
.
22
Guerrero
M.
,
Urbano
M.
,
Velaparthi
S.
,
Zhao
J.
,
Schaeffer
M. T.
,
Brown
S.
,
Rosen
H.
,
Roberts
E.
.
2011
.
Discovery, design and synthesis of the first reported potent and selective sphingosine-1-phosphate 4 (S1P4) receptor antagonists.
Bioorg. Med. Chem. Lett.
21
:
3632
3636
.
23
Guerrero
M.
,
Urbano
M.
,
Zhao
J.
,
Crisp
M.
,
Chase
P.
,
Hodder
P.
,
Schaeffer
M. T.
,
Brown
S.
,
Rosen
H.
,
Roberts
E.
.
2012
.
Discovery, design and synthesis of novel potent and selective sphingosine-1-phosphate 4 receptor (S1P4-R) agonists.
Bioorg. Med. Chem. Lett.
22
:
537
542
.
24
Platanias
L. C.
.
2005
.
Mechanisms of type-I- and type-II-interferon-mediated signalling.
Nat. Rev. Immunol.
5
:
375
386
.
25
Sage
A. P.
,
Murphy
D.
,
Maffia
P.
,
Masters
L. M.
,
Sabir
S. R.
,
Baker
L. L.
,
Cambrook
H.
,
Finigan
A. J.
,
Ait-Oufella
H.
,
Grassia
G.
, et al
.
2014
.
MHC Class II-restricted antigen presentation by plasmacytoid dendritic cells drives proatherogenic T cell immunity.
Circulation
130
:
1363
1373
.
26
Le Mercier
I.
,
Poujol
D.
,
Sanlaville
A.
,
Sisirak
V.
,
Gobert
M.
,
Durand
I.
,
Dubois
B.
,
Treilleux
I.
,
Marvel
J.
,
Vlach
J.
, et al
.
2013
.
Tumor promotion by intratumoral plasmacytoid dendritic cells is reversed by TLR7 ligand treatment.
Cancer Res.
73
:
4629
4640
.
27
Pallotta
M. T.
,
Orabona
C.
,
Volpi
C.
,
Vacca
C.
,
Belladonna
M. L.
,
Bianchi
R.
,
Servillo
G.
,
Brunacci
C.
,
Calvitti
M.
,
Bicciato
S.
, et al
.
2011
.
Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells.
Nat. Immunol.
12
:
870
878
.
28
Fabricius
D.
,
Nußbaum
B.
,
Busch
D.
,
Panitz
V.
,
Mandel
B.
,
Vollmer
A.
,
Westhoff
M. A.
,
Kaltenmeier
C.
,
Lunov
O.
,
Tron
K.
, et al
.
2013
.
Antiviral vaccines license T cell responses by suppressing granzyme B levels in human plasmacytoid dendritic cells.
J. Immunol.
191
:
1144
1153
.
29
Jahrsdörfer
B.
,
Vollmer
A.
,
Blackwell
S. E.
,
Maier
J.
,
Sontheimer
K.
,
Beyer
T.
,
Mandel
B.
,
Lunov
O.
,
Tron
K.
,
Nienhaus
G. U.
, et al
.
2010
.
Granzyme B produced by human plasmacytoid dendritic cells suppresses T-cell expansion.
Blood
115
:
1156
1165
.
30
Jähn
P. S.
,
Zänker
K. S.
,
Schmitz
J.
,
Dzionek
A.
.
2010
.
BDCA-2 signaling inhibits TLR-9-agonist-induced plasmacytoid dendritic cell activation and antigen presentation.
Cell. Immunol.
265
:
15
22
.
31
Hervas-Stubbs
S.
,
Mancheño
U.
,
Riezu-Boj
J. I.
,
Larraga
A.
,
Ochoa
M. C.
,
Alignani
D.
,
Alfaro
C.
,
Morales-Kastresana
A.
,
Gonzalez
I.
,
Larrea
E.
, et al
.
2012
.
CD8 T cell priming in the presence of IFN-α renders CTLs with improved responsiveness to homeostatic cytokines and recall antigens: important traits for adoptive T cell therapy.
J. Immunol.
189
:
3299
3310
.
32
Kuwana
M.
2002
.
Induction of anergic and regulatory T cells by plasmacytoid dendritic cells and other dendritic cell subsets.
Hum. Immunol.
63
:
1156
1163
.
33
Brinkmann
V.
,
Geiger
T.
,
Alkan
S.
,
Heusser
C. H.
.
1993
.
Interferon alpha increases the frequency of interferon gamma-producing human CD4+ T cells.
J. Exp. Med.
178
:
1655
1663
.
34
Xia
C. Q.
,
Peng
R.
,
Chernatynskaya
A. V.
,
Yuan
L.
,
Carter
C.
,
Valentine
J.
,
Sobel
E.
,
Atkinson
M. A.
,
Clare-Salzler
M. J.
.
2014
.
Increased IFN-α-producing plasmacytoid dendritic cells (pDCs) in human Th1-mediated type 1 diabetes: pDCs augment Th1 responses through IFN-α production.
J. Immunol.
193
:
1024
1034
.
35
Curtsinger
J. M.
,
Valenzuela
J. O.
,
Agarwal
P.
,
Lins
D.
,
Mescher
M. F.
.
2005
.
Type I IFNs provide a third signal to CD8 T cells to stimulate clonal expansion and differentiation.
J. Immunol.
174
:
4465
4469
.
36
Julian
M. W.
,
Shao
G.
,
Bao
S.
,
Knoell
D. L.
,
Papenfuss
T. L.
,
VanGundy
Z. C.
,
Crouser
E. D.
.
2012
.
Mitochondrial transcription factor A serves as a danger signal by augmenting plasmacytoid dendritic cell responses to DNA.
J. Immunol.
189
:
433
443
.
37
Lee
B. L.
,
Moon
J. E.
,
Shu
J. H.
,
Yuan
L.
,
Newman
Z. R.
,
Schekman
R.
,
Barton
G. M.
.
2013
.
UNC93B1 mediates differential trafficking of endosomal TLRs.
eLife
2
:
e00291
.
38
Tavano
B.
,
Galao
R. P.
,
Graham
D. R.
,
Neil
S. J.
,
Aquino
V. N.
,
Fuchs
D.
,
Boasso
A.
.
2013
.
Ig-like transcript 7, but not bone marrow stromal cell antigen 2 (also known as HM1.24, tetherin, or CD317), modulates plasmacytoid dendritic cell function in primary human blood leukocytes.
J. Immunol.
190
:
2622
2630
.
39
Bao
M.
,
Liu
Y. J.
.
2013
.
Regulation of TLR7/9 signaling in plasmacytoid dendritic cells.
Protein Cell
4
:
40
52
.
40
Röck
J.
,
Schneider
E.
,
Grün
J. R.
,
Grützkau
A.
,
Küppers
R.
,
Schmitz
J.
,
Winkels
G.
.
2007
.
CD303 (BDCA-2) signals in plasmacytoid dendritic cells via a BCR-like signalosome involving Syk, Slp65 and PLCgamma2.
Eur. J. Immunol.
37
:
3564
3575
.
41
Kerkmann
M.
,
Rothenfusser
S.
,
Hornung
V.
,
Towarowski
A.
,
Wagner
M.
,
Sarris
A.
,
Giese
T.
,
Endres
S.
,
Hartmann
G.
.
2003
.
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.
170
:
4465
4474
.
42
Liao
A. P.
,
Salajegheh
M.
,
Morehouse
C.
,
Nazareno
R.
,
Jubin
R. G.
,
Jallal
B.
,
Yao
Y.
,
Greenberg
S. A.
.
2010
.
Human plasmacytoid dendritic cell accumulation amplifies their type 1 interferon production.
Clin. Immunol.
136
:
130
138
.
43
Mahnke
K.
,
Guo
M.
,
Lee
S.
,
Sepulveda
H.
,
Swain
S. L.
,
Nussenzweig
M.
,
Steinman
R. M.
.
2000
.
The dendritic cell receptor for endocytosis, DEC-205, can recycle and enhance antigen presentation via major histocompatibility complex class II-positive lysosomal compartments.
J. Cell Biol.
151
:
673
684
.
44
Kato
M.
,
McDonald
K. J.
,
Khan
S.
,
Ross
I. L.
,
Vuckovic
S.
,
Chen
K.
,
Munster
D.
,
MacDonald
K. P.
,
Hart
D. N.
.
2006
.
Expression of human DEC-205 (CD205) multilectin receptor on leukocytes.
Int. Immunol.
18
:
857
869
.
45
Tel
J.
,
Benitez-Ribas
D.
,
Hoosemans
S.
,
Cambi
A.
,
Adema
G. J.
,
Figdor
C. G.
,
Tacken
P. J.
,
de Vries
I. J.
.
2011
.
DEC-205 mediates antigen uptake and presentation by both resting and activated human plasmacytoid dendritic cells.
Eur. J. Immunol.
41
:
1014
1023
.
46
Gräler
M. H.
,
Grosse
R.
,
Kusch
A.
,
Kremmer
E.
,
Gudermann
T.
,
Lipp
M.
.
2003
.
The sphingosine 1-phosphate receptor S1P4 regulates cell shape and motility via coupling to Gi and G12/13.
J. Cell. Biochem.
89
:
507
519
.
47
Fabricius
D.
,
Neubauer
M.
,
Mandel
B.
,
Schütz
C.
,
Viardot
A.
,
Vollmer
A.
,
Jahrsdörfer
B.
,
Debatin
K. M.
.
2010
.
Prostaglandin E2 inhibits IFN-alpha secretion and Th1 costimulation by human plasmacytoid dendritic cells via E-prostanoid 2 and E-prostanoid 4 receptor engagement.
J. Immunol.
184
:
677
684
.
48
Contractor
N.
,
Louten
J.
,
Kim
L.
,
Biron
C. A.
,
Kelsall
B. L.
.
2007
.
Cutting edge: Peyer’s patch plasmacytoid dendritic cells (pDCs) produce low levels of type I interferons: possible role for IL-10, TGFbeta, and prostaglandin E2 in conditioning a unique mucosal pDC phenotype.
J. Immunol.
179
:
2690
2694
.
49
Bekeredjian-Ding
I.
,
Schäfer
M.
,
Hartmann
E.
,
Pries
R.
,
Parcina
M.
,
Schneider
P.
,
Giese
T.
,
Endres
S.
,
Wollenberg
B.
,
Hartmann
G.
.
2009
.
Tumour-derived prostaglandin E and transforming growth factor-beta synergize to inhibit plasmacytoid dendritic cell-derived interferon-alpha.
Immunology
128
:
439
450
.
50
Jiang
L. I.
,
Collins
J.
,
Davis
R.
,
Fraser
I. D.
,
Sternweis
P. C.
.
2008
.
Regulation of cAMP responses by the G12/13 pathway converges on adenylyl cyclase VII.
J. Biol. Chem.
283
:
23429
23439
.
51
Haeryfar
S. M.
.
2005
.
The importance of being a pDC in antiviral immunity: the IFN mission versus Ag presentation?
Trends Immunol.
26
:
311
317
.
52
Curtsinger
J. M.
,
Mescher
M. F.
.
2010
.
Inflammatory cytokines as a third signal for T cell activation.
Curr. Opin. Immunol.
22
:
333
340
.
53
Lui
G.
,
Manches
O.
,
Angel
J.
,
Molens
J. P.
,
Chaperot
L.
,
Plumas
J.
.
2009
.
Plasmacytoid dendritic cells capture and cross-present viral antigens from influenza-virus exposed cells.
PLoS One
4
:
e7111
.
54
Sikora
A. G.
,
Jaffarzad
N.
,
Hailemichael
Y.
,
Gelbard
A.
,
Stonier
S. W.
,
Schluns
K. S.
,
Frasca
L.
,
Lou
Y.
,
Liu
C.
,
Andersson
H. A.
, et al
.
2009
.
IFN-alpha enhances peptide vaccine-induced CD8+ T cell numbers, effector function, and antitumor activity.
J. Immunol.
182
:
7398
7407
.
55
Starbeck-Miller
G. R.
,
Xue
H. H.
,
Harty
J. T.
.
2014
.
IL-12 and type I interferon prolong the division of activated CD8 T cells by maintaining high-affinity IL-2 signaling in vivo.
J. Exp. Med.
211
:
105
120
.
56
Golding
A.
,
Rosen
A.
,
Petri
M.
,
Akhter
E.
,
Andrade
F.
.
2010
.
Interferon-alpha regulates the dynamic balance between human activated regulatory and effector T cells: implications for antiviral and autoimmune responses.
Immunology
131
:
107
117
.
57
Fuertes
M. B.
,
Kacha
A. K.
,
Kline
J.
,
Woo
S. R.
,
Kranz
D. M.
,
Murphy
K. M.
,
Gajewski
T. F.
.
2011
.
Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8alpha+ dendritic cells.
J. Exp. Med.
208
:
2005
2016
.
58
Diamond
M. S.
,
Kinder
M.
,
Matsushita
H.
,
Mashayekhi
M.
,
Dunn
G. P.
,
Archambault
J. M.
,
Lee
H.
,
Arthur
C. D.
,
White
J. M.
,
Kalinke
U.
, et al
.
2011
.
Type I interferon is selectively required by dendritic cells for immune rejection of tumors.
J. Exp. Med.
208
:
1989
2003
.
59
Sisirak
V.
,
Faget
J.
,
Gobert
M.
,
Goutagny
N.
,
Vey
N.
,
Treilleux
I.
,
Renaudineau
S.
,
Poyet
G.
,
Labidi-Galy
S. I.
,
Goddard-Leon
S.
, et al
.
2012
.
Impaired IFN-α production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression.
Cancer Res.
72
:
5188
5197
.
60
Labidi-Galy
S. I.
,
Treilleux
I.
,
Goddard-Leon
S.
,
Combes
J. D.
,
Blay
J. Y.
,
Ray-Coquard
I.
,
Caux
C.
,
Bendriss-Vermare
N.
.
2012
.
Plasmacytoid dendritic cells infiltrating ovarian cancer are associated with poor prognosis.
OncoImmunology
1
:
380
382
.
61
Kunkel
G. T.
,
Maceyka
M.
,
Milstien
S.
,
Spiegel
S.
.
2013
.
Targeting the sphingosine-1-phosphate axis in cancer, inflammation and beyond.
Nat. Rev. Drug Discov.
12
:
688
702
.
62
Galicia-Rosas
G.
,
Pikor
N.
,
Schwartz
J. A.
,
Rojas
O.
,
Jian
A.
,
Summers-Deluca
L.
,
Ostrowski
M.
,
Nuesslein-Hildesheim
B.
,
Gommerman
J. L.
.
2012
.
A sphingosine-1-phosphate receptor 1-directed agonist reduces central nervous system inflammation in a plasmacytoid dendritic cell-dependent manner.
J. Immunol.
189
:
3700
3706
.
63
Gao
Y.
,
Majchrzak-Kita
B.
,
Fish
E. N.
,
Gommerman
J. L.
.
2009
.
Dynamic accumulation of plasmacytoid dendritic cells in lymph nodes is regulated by interferon-beta.
Blood
114
:
2623
2631
.

The authors have no financial conflicts of interest.

Supplementary data