The type-III interferon (IFN) family is composed of 3 molecules in humans: IFN-λ1 (interleukin-29 [IL-29]), IFN-λ2 (IL-28A), and IFN-λ3 (IL-28B), each of which signals through the same receptor complex. Plasmacytoid dendritic cells (pDCs) are major IFN-λ producers among peripheral lymphocytes. Recently, it has been shown that IFN-λ1 exerts a powerful inhibitory effect over the T-helper 2 (Th2) response by antagonizing the effect of IL-4 on CD4+ T cells and inhibiting the production of Th2-associated cytokines. Here, we asked whether Th2 cytokines exert reciprocal control over IFN-λ production. IL-4 treatment during stimulation of human peripheral lymphocytes significantly elevated IFN-λ1 transcription and secretion. However, pDCs were not directly responsive to IL-4. Using depletion and reconstitution experiments, we showed that IL-4–responsive monocytes are an intermediary cell, responding to IL-4 by elevating their secretion of IL-1 receptor antagonist (IL-Ra); this IL-1Ra acts on pDCs to elevate their IFN-λ1 output. Thus, our experiments revealed a novel mechanism for regulation of both IFN-λ1 production and pDC function, and suggests an expanded immunomodulatory role for Th2-associated cytokines.

The 3 members of the interferon-λ (IFN-λ) family (λ1, λ2, and λ3) were originally described in the context of their similarity to both IFN-α/β and the IL-10 family of ligands.1,2  A recent report has confirmed that while IFN-λ exhibits IFN-like activity,3  it is structurally related to members of the IL-10 cytokine family, particularly IL-22.4  Early characterization of type-III IFN activity revealed that signaling is mediated through a heterodimeric receptor complex composed of the signaling subunit, IL-28Rα, and the nonsignaling partner IL-10Rβ; all 3 ligands signal through this receptor. Ligation of the IFN-λ receptor leads to the induction of signature molecules typically associated with antiviral immunity, such as 2′ to 5′ OAS and MxA.1,2  Although there is some overlap in the functions of type-I and type-III IFNs, there are clear differences in their biologic activities. The antiviral effects of IFN-λ are less robust compared with those of IFN-α,5,,8  but the IFN-λ molecules remain able to induce antiviral activity both in vivo and in vitro,5,9,10  causing, for example, reduction of hepatitis B and C replication in hepatocytes, and of HSV-2 in the vaginal mucosa. These and other studies have highlighted the particularly powerful effect of IFN-λ1 on epithelial cells,11  which is mediated by the relatively high expression of IL-28Rα.12 

In addition to their antiviral effects, type-III IFNs have been shown to play a critical role in regulating the adaptive immune response by acting directly on T cells to inhibit T-helper 2 (Th2) polarization and cytokine production.13,15  Furthermore, treatment of peripheral blood mononuclear cells (PBMCs),16  and more specifically naive and memory CD4+ T cells,17  with IFN-λ1 results in marked reductions of IL-4, IL-13, and IL-5, and a down-regulation in both GATA3 and IL-4Rα expression.

An imbalance of Th1/Th2 regulation is a hallmark component of the pathophysiology of asthma and allergic inflammation. The chronic up-regulation of Th2 cytokines in patients with asthma drives inflammation and lymphocyte recruitment to the airway; high levels of IL-4 promote IgE class switching and mast cell activity, and excessive IL-13 contributes to airway remodeling.18,19  Recently, it was shown that patients with asthma are markedly deficient in IFN-λ1 production by both epithelial and brochoalveolar lavage cells.20  Many acute asthma exacerbations are a consequence of infection by respiratory viruses, most commonly rhinovirus. Primary bronchial epithelial cells from normal donors respond to rhinovirus (RV) stimulation by producing high levels of IFN-λ1, IFN-λ2, IFN-λ3, and IFN-β, and undergo rapid apoptosis, which prevents viral replication. However, patients with asthma do not produce sufficient amounts of type-I or type-III IFNs and thus yield high titers of infectious virus. The mechanisms by which deficient IFN-λ production supports asthma development are unknown (either through reduced ability to stifle viral replication or loss of control over elevated Th2 responses), but it is evident that IFN-λ is a pivotal element in this process.

Dendritic cells (DCs) are known to be important producers of IFN-λ. IFN-λ mRNA is induced in monocyte-derived DCs (MDDCs) in response to influenza, poly I:C, and lipopolysaccharide (LPS),21,22  and in human plasmacytoid dendritic cells (pDCs) by influenza.21  IFN-λ protein production has been observed in MDDCs upon stimulation with LPS or CD40L plus IFN-γ,23  and recently in our own work, in pDCs in responses to HSV, CpG, and imiquimod.24  This is in line with robust type-I IFN responses typically observed in pDCs.25 

Given the direct antagonism of IL-4 by IFN-λ in both T cells and myeloid/classical DCs (mDCs), we were prompted to ask whether Th2 cytokines act in a reciprocal fashion to regulate IFN-λ production. Our data show that in the presence of specific viral stimuli, IL-4 acts to enhance IFN-λ1 production from the PBMCs of healthy donors. Stepwise depletion and reconstitution experiments revealed that production of IL-1 receptor antagonist (IL-1Ra) from monocytes mediated this Th2-enhanced production of IFN-λ1 by human pDCs.

Isolation of human PBMCs

Buffy coats were obtained from the New Jersey Blood Bank, and PBMCs were isolated by density gradient separation over Ficoll-Paque (Sigma-Aldrich). Cells were then washed in RPMI 1640 (Invitrogen), resuspended in medium supplemented with 10% (vol/vol) fetal bovine Serum (FBS; Thermo Scientific), and enumerated by hemacytometer using trypan blue (30% vol/vol; Sigma-Aldrich). Cells were resuspended at a density of 1 × 106/mL and cocultured as indicated with HSV-1 (multiplicity of infection [MOI] = 1; originally a gift from Dr John Blaho, Medical Diagnostic Laboratories, Hamilton, NJ), in the presence or absence of IL-4 (50 ng/mL; R&D Systems).

IFN-λ1 quantitation

Levels of IFN-λ1 protein in culture supernatants were measured using the “Duoset” human IL-29 enzyme-linked immunosorbent assay (ELISA) kit from R&D Systems according to the manufacturer's protocol. End point optical densities at 450 nm were determined using a VERSAmax spectrophotometer and compiled using SoftMax software (both Molecular Devices).

pDC isolation

pDCs were isolated from fresh PBMCs using the Human Plasmacytoid DC Negative Isolation Kit (StemCell Technologies) according to the manufacturer's protocol. The enriched cells were assessed for more than 95% purity using the following antibodies: anti-CD123–APC and anti–BDCA-2–FITC (Miltenyi Biotec) and anti–lineage cocktail–FITC and anti–HLA-DR–PerCP (Becton Dickinson–Pharmingen). Cells were blocked with 5% (vol/vol) heat-inactivated human AB serum and incubated with the appropriate antibodies in the dark at 4°C for 30 minutes, then washed twice in phosphate-buffered saline (PBS) and fixed in 1% paraformaldehyde in PBS before being acquired using a FACSCalibur flow cytometer (BD) and analyzed using CellQuest Pro (BD) and FlowJo (TreeStar) software packages.

PBMC depletions

PBMCs depleted of target subsets were generated from 1 × 108 PBMCs using the following positive isolation kits from StemCell Technologies according to the manufacturer's instructions: T cells, CD3+ EasySep positive selection kit; B cells, CD19+ EasySep positive selection kit; natural killer (NK) cells, CD56+ positive selection kit; and monocytes, CD14+ positive selection kit. pDCs were labeled as described using BDCA-2–PE and depleted using the PE Depletion kit. Both positively and negatively isolated populations were assayed for percentage of pDCs by flow cytometry, as well as for purity and recovery using the following antibodies: anti-CD3–FITC, anti-CD4–PeCy5, anti-CD14–PE, anti-CD16–FITC, anti-CD20–FITC, anti-CD56–PeCy5 (eBioscience), anti-CD14–PE, and anti-CD8–PeCy5 (BD Pharmingen).

Monocyte enrichment

Negatively isolated monocytes were selected using the Monocyte Enrichment kit; positively selected monocytes were obtained using the CD14+ Positive Selection kit, each from StemCell Technologies. The selected cells were harvested and washed twice in RPMI 1640. The resulting cell populations were assessed for purity and the presence of contaminating pDCs via flow cytometry. Neutralizing antibody to IL-1Ra (R&D Systems) was used at 10 μg/mL where indicated.

pDCs are the principal IFN-λ1–secreting cells upon stimulation with HSV

Human pDCs have been shown to produce IFN-λ1 in response to a variety of stimuli, including HSV. To verify whether pDCs are the principal IFN-λ1–secreting population in response to HSV, PBMCs were partially depleted of pDCs (mean reduction 42.5%) and cultured overnight in the presence of stimulatory poly I:C, LPS, PMA/ionomycin, or HSV. Compared with PBMCs, only HSV elicited a substantial production of IFN-λ1 (Figure 1A) that was decreased by approximately 60% in the pDC-depleted cells. Depleting pDCs did not affect secretion in response to any other stimulus. These findings confirmed previous observations that pDCs are the major PBMC population to produce IFN-λ1 in response to HSV,24  which is known to stimulate pDCs at least in part through Toll-like receptor 9 (TLR9)26,28  and potentially through TLR7.29  Unlike myeloid/classical DCs (mDCs) or MDDCs,23  highly enriched pDCs did not produce appreciable amounts of IFN-λ1 in response to poly I:C or LPS, as has been previously observed.24  This is in agreement with their selective expression of TLR7 and TLR9, and paucity of expression and/or response to TLR3 (poly I:C) or TLR4 (LPS) agonists.30,31  Nonetheless, these stimuli elicit production from mDCs, which are present in PBMCs at nearly the same low frequency of pDCs (0.2%-0.5%).32 

Figure 1

IL-4 enhances IFN-λ1 by PBMCs, but not purified pDCs. (A) Peripheral blood mononuclear cells (PBMCs) were partially depleted of plasmacytoid dendritic cells (pDCs) using magnetic beads. PBMCs or pDC-depleted PBMCs were incubated overnight with the indicated stimuli. Supernatants (SNs) were harvested and IFN-λ1 was measured by enzyme-linked immunosorbent assay (ELISA; n = 3; mean ± SD is shown). The effect of interleukin-4 (IL-4) on IFN-λ1 production by PBMCs (B) or pDCs (C) was determined by stimulating either population overnight with HSV in the presence or absence of IL-4. SNs were harvested and assayed as in panel A. IL-4 was shown to significantly increased IFN-λ1 production in HSV-stimulated PBMCs (n = 18; P < .001) but not pDCs (n = 7; P = .35) as determined by the Wilcoxon signed-rank test.

Figure 1

IL-4 enhances IFN-λ1 by PBMCs, but not purified pDCs. (A) Peripheral blood mononuclear cells (PBMCs) were partially depleted of plasmacytoid dendritic cells (pDCs) using magnetic beads. PBMCs or pDC-depleted PBMCs were incubated overnight with the indicated stimuli. Supernatants (SNs) were harvested and IFN-λ1 was measured by enzyme-linked immunosorbent assay (ELISA; n = 3; mean ± SD is shown). The effect of interleukin-4 (IL-4) on IFN-λ1 production by PBMCs (B) or pDCs (C) was determined by stimulating either population overnight with HSV in the presence or absence of IL-4. SNs were harvested and assayed as in panel A. IL-4 was shown to significantly increased IFN-λ1 production in HSV-stimulated PBMCs (n = 18; P < .001) but not pDCs (n = 7; P = .35) as determined by the Wilcoxon signed-rank test.

Close modal

IL-4 modulates IFN-λ1 responses of PBMCs but not purified pDCs

Recent work in our laboratory has shown that IFN-λ1 exerts a significant negative regulatory effect on the production of Th2 cytokines by T cells. We hypothesized that type-III IFNs were a critical determinant during T-cell polarization and sought to understand what reciprocal effect, if any, Th2 cytokines had on IFN-λ1 production by PBMCs and/or pDCs. To that end, PBMCs were stimulated with HSV in the presence or absence of the Th2-associated cytokine IL-4 and analyzed for secretion of IFN-λ1.

A marked increase in IFN-λ1 production was observed when IL-4 was present during HSV stimulation (56.53%, SD ± 21.8; Figure 1B; P < .001). This effect was observed in 18 of 18 donors tested. To determine whether or not this was a direct effect of IL-4 on the principal IFN-λ1–producing cells, pDCs were isolated and stimulated in the same manner. However, IL-4 had no such effect on purified pDCs (Figure 1C; 0 of 7 donors tested). These data suggested that either a second cell type actively secreted IFN-λ1 in response to IL-4 and HSV (but to neither alone), or that an unidentified cell type was being stimulated to secrete one or more cytokines or additional factor(s) that in turn increased IFN-λ1 production by pDCs.

IL-4–responsive monocytes are permissive for enhanced IFN-λ1 production

To identify additional subpopulations that may contribute to IL-4–enhanced IFN-λ1 production, PBMCs were depleted of T cells, B cells, NK cells, or monocytes using antibody-conjugated magnetic beads. To assess the efficiency of each depletion, resultant populations were evaluated for the successful removal of target cells using flow cytometry (Figure 2A top panels; percentages shown indicate effective depletion of each subpopulation). The proportion of pDCs remaining in each subset-depleted population was also determined (Figure 2A bottom panels). A slight enrichment of pDCs can be observed in the T cell–depleted cells, due to the removal of the population that comprises a high percentage (∼ 75%) of PBMCs. B cells, NK cells, and monocytes combine to make up the majority of remaining portion, so their deletion did not result in the same elevation in pDC frequency.

Figure 2

Monocytes mediate IL-4–enhanced IFN-λ1 production. PBMCs were depleted of T cells, B cells, NK cells, or monocytes by magnetic bead separation. (A) Successful depletion of the indicated populations was verified using flow cytometry (percentage of remaining cells indicated; top panels). The percentage of pDCs present in each depleted population was also measured by flow cytometry (percentages indicated; bottom panels). Depleted populations were then stimulated with HSV in the presence or absence of IL-4 to determine the contribution of each cell type to enhanced IFN-λ1 production. (B) SNs were harvested after 24 hours and assayed for presence of IFN-λ1 by ELISAs (n = 4). The graph shows percentage change in HSV-induced IFN-λ1 production upon addition of IL-4. Means ± SE for the 4 donors are shown. *P < .05, determined using the Student t test.

Figure 2

Monocytes mediate IL-4–enhanced IFN-λ1 production. PBMCs were depleted of T cells, B cells, NK cells, or monocytes by magnetic bead separation. (A) Successful depletion of the indicated populations was verified using flow cytometry (percentage of remaining cells indicated; top panels). The percentage of pDCs present in each depleted population was also measured by flow cytometry (percentages indicated; bottom panels). Depleted populations were then stimulated with HSV in the presence or absence of IL-4 to determine the contribution of each cell type to enhanced IFN-λ1 production. (B) SNs were harvested after 24 hours and assayed for presence of IFN-λ1 by ELISAs (n = 4). The graph shows percentage change in HSV-induced IFN-λ1 production upon addition of IL-4. Means ± SE for the 4 donors are shown. *P < .05, determined using the Student t test.

Close modal

Whole PBMCs and each of the depleted populations were then stimulated with IL-4, HSV, or both. IL-4–enhanced IFN-λ1 secretion continued to be observed after depletion of either T, B, or NK cells. In contrast, monocyte depletion prevented the expected IL-4–mediated enhancement of IFN-λ1 secretion (Figure 2B).

pDCs, not monocytes, secrete enhanced levels of IFN-λ1 in response to IL-4

The results obtained from the depletion experiments suggested that monocytes are a critical component of IL-4–enhanced IFN-λ1 production. To determine whether pDCs work in concert with monocytes to achieve this, purified pDCs were cocultured with autologous monocytes at a ratio of 9:1 (pDC/monocyte). Supernatants (SNs) were harvested from these cultures after overnight incubation with HSV in the presence or absence of IL-4 (Figure 3A), and secreted IFN-λ1 was measured. As shown, the addition of monocytes to pDCs conferred the ability to produce elevated levels of IFN-λ1 upon viral stimulation, which does not occur in pDCs alone (Figure 1C).

Figure 3

pDCs are the cellular source of IL-4–enhanced IFN-λ1. (A) Purified pDCs were cocultured with autologous monocytes at a ratio of 9:1 (pDC/monocyte). SNs were harvested from these cultures after overnight incubation with HSV in the presence or absence of IL-4; IFN-λ1 levels were determined by ELISA. (B) Monocytes were stimulated with IL-4 (50 ng/mL) for 1 hour then washed. Cells were then cocultured with autologous pDCs and stimulated overnight with HSV in the presence or absence of IL-4 as shown. One representative experiment of 3 is shown; mean of triplicate wells ± SD.

Figure 3

pDCs are the cellular source of IL-4–enhanced IFN-λ1. (A) Purified pDCs were cocultured with autologous monocytes at a ratio of 9:1 (pDC/monocyte). SNs were harvested from these cultures after overnight incubation with HSV in the presence or absence of IL-4; IFN-λ1 levels were determined by ELISA. (B) Monocytes were stimulated with IL-4 (50 ng/mL) for 1 hour then washed. Cells were then cocultured with autologous pDCs and stimulated overnight with HSV in the presence or absence of IL-4 as shown. One representative experiment of 3 is shown; mean of triplicate wells ± SD.

Close modal

Negatively isolated (ie, untouched) T cells, B cells, NK cells, and monocytes were also each assayed for their ability to produce IFN-λ1 in response to HSV. IL-4–enhanced IFN-λ1 production could not be detected in any of these isolated populations (data not shown). From these data, we hypothesized that in response to IL-4, monocytes induce pDCs to produce greater amounts of IFN-λ, either via secretion of a soluble factor or in a contact-dependent manner. To address this question, monocytes were treated with IL-4 for 1 hour, washed, then cocultured with pDCs. Monocytes alone or monocyte-pDC cocultures were then stimulated overnight with HSV (Figure 3B) and assessed for IFN-λ1 production by ELISA. IL-4–treated monocytes themselves did not produce IFN-λ1 (with or without viral stimulation). pDCs, however, once again exhibited increased IFN-λ1 production, dependent on IL-4–treated monocytes.

IL-4–treated monocytes up-regulate IFN-λ1 production by stimulated pDCs via IL-1Ra

To identify factors which are secreted by monocytes and may mediate this phenomenon, positively selected monocytes were stimulated for 1 hour with IL-4, washed twice, and incubated overnight. Supernatants from IL-4–treated monocytes were analyzed using Cytokine Antibody Arrays from RayBiotech. This analysis identified the presence of IL-1Ra (which has been reported previously33 ). IL-1Ra is known to function as a soluble IL-1 antagonist by binding to the IL-1R with virtually equal avidity as IL-1, but failing to trigger receptor activation. To assess its effect on IFN-λ1 production, purified pDCs were exposed to HSV overnight in the presence of IL-4, IL-1β, or IL-1Ra (Figure 4A); supernatants were analyzed by ELISA. As observed previously, IL-4 did not alter the levels of IFN-λ1 secretion. IL-1β exerted an inhibitory effect on IFN-λ1 production, reducing expression by 26.6%. However, IFN-λ1 production was enhanced up to 57.5% (± 32.0%) upon addition of IL-1Ra, in a dose-responsive manner. To confirm that IL-1Ra was the agent mediating IL-4–enhanced, monocyte-dependent IFN-λ1 secretion, monocytes were cultured with IL-4 for 1 hour, washed twice, and incubated overnight; culture SNs were then harvested. pDCs were cultured in these supernatants and stimulated with HSV in the presence or absence of neutralizing antibody to IL-1Ra (Figure 4B). Supernatants from IL-4–treated monocytes were sufficient to mediate enhanced IFN-λ1 production in virally stimulated pDCs, demonstrating that the phenomenon is indeed contact-independent. Addition of neutralizing antibody to IL-1Ra blocked the enhanced IFN-λ1 secretion, thereby confirming the responsiveness of pDCs to IL-1Ra and its ability to modulate cytokine production.

Figure 4

IL-1Ra augments IFN-λ1 production by pDCs. (A) pDCs were stimulated overnight with HSV in the presence of IL-4 (50 ng/mL), IL-1 (100 ng/mL), or IL-1Ra as indicated; SNs were assayed by ELISA for IFN-λ1 (means ± SD from 3 independent experiments are shown). (B) Monocytes from 2 separate donors were incubated for 1 hour with IL-4, washed twice, and then cultured overnight. SNs were collected and used as culture medium for purified allogeneic pDCs, isolated from 2 independent donors. pDCs were incubated overnight in medium or SNs from IL-4–treated monocytes with or without neutralizing Ab to IL-1Ra, and stimulated with HSV. IFN-λ1 levels were measured by ELISA. A total of 2 independent experiments are shown (means ± SD). *P < .05 as determined by the Student t test.

Figure 4

IL-1Ra augments IFN-λ1 production by pDCs. (A) pDCs were stimulated overnight with HSV in the presence of IL-4 (50 ng/mL), IL-1 (100 ng/mL), or IL-1Ra as indicated; SNs were assayed by ELISA for IFN-λ1 (means ± SD from 3 independent experiments are shown). (B) Monocytes from 2 separate donors were incubated for 1 hour with IL-4, washed twice, and then cultured overnight. SNs were collected and used as culture medium for purified allogeneic pDCs, isolated from 2 independent donors. pDCs were incubated overnight in medium or SNs from IL-4–treated monocytes with or without neutralizing Ab to IL-1Ra, and stimulated with HSV. IFN-λ1 levels were measured by ELISA. A total of 2 independent experiments are shown (means ± SD). *P < .05 as determined by the Student t test.

Close modal

The Th1/Th2 paradigm was originally defined by Mosmann, Coffman, and colleagues.34  Recently, we have shown that IFN-λ1, a novel cytokine produced by pDCs24  and other immune cells,23  is a powerful inhibitor of human Th2-cell development.13,16,17 

Here, we have shown that the Th2-related cytokine IL-4, through its activity on monocytes, serves to regulate the production of IFN-λ1 by human pDCs. This effect is specific to monocytes, as neither T, B, or NK cells responded to IL-4 in this way. Monocytes do not directly produce IFN-λ in response to IL-4, but rather enhance production by pDCs, through secretion of IL-1Ra; thus, we have defined a novel feedback loop between pDCs and Th2 cells. Although we have described the role of IL-4 here, it should be noted that we observed similar effects with IL-13; these effects were consistent and of a lesser magnitude than those observed with IL-4. This may be due to differences in expression of the IL-4/13 type-I/type-II receptors35  on pDCs, which we did not investigate here.

As we have shown, increased production of IFN-λ1 by pDCs is mediated principally by IL-1Ra, which attenuates the effect of IL-1 by competitively binding its receptor without activating the IL-1–signaling pathway.36,37  IL-1Ra was already known to be produced by monocytes in response to IL-4,33  and is shown here to enhance HSV-induced IFN-λ1 production by pDCs. Elevated serum levels of IL-1Ra have previously been observed in patients undergoing IL-4 cancer immunotherapy33 ; similarly, monocytes isolated from healthy individuals and treated with IL-4 in vitro also yield higher IL-1Ra levels.38  In studies reported here, we used increasing amounts of IL-1Ra (∼ 3- to 15-fold above systemic serum levels in patients) to show dose responsiveness on pDCs. For IL-1Ra to function as an enhancer of type-III cytokine production by pDCs, it would appear to counterbalance a repressive signal from IL-1, which our data also suggest. However, only sparse information exists about the response of pDCs to IL-1. Upon ligation of IL-1R by IL-1, the adaptor molecule myeloid differentiation factor 88 (MyD88) permits interaction with IL-1R–associated kinases (IRAKs) in a signaling cascade that results in activation of NF-κB.39,41  TLR7 and TLR9, the predominant Toll-like receptors present in pDCs, both signal through MyD88- and IRAK-dependent pathways. Interestingly, in our experiments, agonists for each of these receptors (imiquimod and CpG, respectively) did not elicit the same high levels of IL-1Ra–enhanced IFN-λ1 production as HSV (data not shown). This may be because HSV can trigger a broad range of TLR-mediated pathways. There is some conjecture about the nature of HSV recognition by pDCs. Receptor binding to the mannose receptor and subsequent endocytosis were shown to be critical,42  and more recent studies suggest that not only TLR9,26  but also TLR729  and potentially TLR2,43  combine additively in signaling the detection and response to HSV by pDCs.

The contribution of IL-4 (and IL-13) to enhanced production of IFN-λ in this system is of specific interest in asthma, especially since IFN-λ1 functions as a potent inhibitor of Th2 cytokine production in both PBMCs and T cells.13,14,16,17  In T cells, this was preceded by a decrease in GATA3,17  the master regulator of Th2 polarization.44,46  A major consequence of GATA3 deficiency is the loss of IL-4Rα from the cell surface. Left unable to respond to IL-4, developing T cells that have been treated with IFN-λ1 cannot sustain effective polarization toward a Th2 phenotype. In the present report, we show that the complementary result is also true, and define a novel feedback loop between pDCs and Th2 cells; Th2 cytokines, acting indirectly via monocytes, augment IFN-λ1 production in stimulated pDCs. In this way, IL-4 and IFN-λ1 comprise a feedback loop whereby Th2 cytokines induce their own repressor (Figure 5). Thus, IFN-λ is shown to not only inhibit the production of IL-4 and IL-13 by Th2-polarized cells, but also to suppress the differentiation toward a Th2 phenotype. In light of studies describing deficient viral-responsive IFN-λ production in patients with asthma, who display chronically elevated levels of IL-4 and IL-13,20  this feedback loop represents a natural checkpoint for control of Th2 cytokines. Whereas respiratory virus infection in healthy individuals triggers effective type-III IFN production, which may then act to dampen secretion of Th2 cytokines, asthmatics exhibit an impaired ability to secrete IFN-λ and may therefore be unable to repress these same cytokines.

Figure 5

Reciprocal control of IFN-λ1 and Th2-associated cytokines. An illustration representing our current thinking of the feedback loop between pDCs, Th2 cells, and monocytes is shown.

Figure 5

Reciprocal control of IFN-λ1 and Th2-associated cytokines. An illustration representing our current thinking of the feedback loop between pDCs, Th2 cells, and monocytes is shown.

Close modal

Whether or not IFN-λ contributes to inhibition of inflammation in these patients is unknown. However, IFN-λ1 has been shown to increase production of IL-6, IL-8, and IL-10 by PBMCs, with no concomitant increase in IL-1 or TNF, suggesting it may not directly engender local tissue destruction, but could contribute to the inflammatory process.14  Irrespective of the pathways that govern this phenomenon, the reciprocal regulation of IFN-λ and IL-4 or IL-13 represents a previously undescribed channel through which a developing immune response may be regulated.

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 funded intramurally by HUMIGEN LLC.

Contribution: N.J.M. designed and executed experiments, analyzed data, and wrote and revised the manuscript; G.E.G. contributed to experimental design, executed experiments, analyzed data, and proofread the manuscript; and G.G. designed experiments and analyzed data, and wrote, revised, and finalized the manuscript.

Conflict-of-interest statement: The authors are employees of HUMIGEN LLC, but otherwise declare no competing financial interests.

Correspondence: Grant Gallagher, HUMIGEN LLC, The Institute for Genetic Immunology, 2439 Kuser Rd, Hamilton, NJ 08690; e-mail: g.gallagher@humigen.org.

1
Kotenko
SV
Gallagher
G
Baurin
VV
et al
IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex.
Nat Immunol
2003
4
1
69
77
2
Sheppard
P
Kindsvogel
W
Xu
W
et al
IL-28, IL-29 and their class II cytokine receptor IL-28R.
Nat Immunol
2003
4
1
63
68
3
Dellgren
C
Gad
HH
Hamming
OJ
Melchjorsen
J
Hartmann
R
Human interferon-lambda3 is a potent member of the type III interferon family.
Genes Immun
2009
10
2
125
131
4
Gad
HH
Dellgren
C
Hamming
OJ
Vends
S
Paludan
SR
Hartmann
R
Interferon-lambda is functionally an interferon but structurally related to the IL-10 family.
J Biol Chem
2009
284
31
20869
20875
5
Bartlett
NW
Buttigieg
K
Kotenko
SV
Smith
GL
Murine interferon lambdas (type III interferons) exhibit potent antiviral activity in vivo in a poxvirus infection model.
J Gen Virol
2005
86
Pt 6
1589
1596
6
Meager
A
Visvalingam
K
Dilger
P
Bryan
D
Wadhwa
M
Biological activity of interleukins-28 and -29: comparison with type I interferons.
Cytokine
2005
31
2
109
118
7
Ank
N
West
H
Bartholdy
C
Eriksson
K
Thomsen
AR
Paludan
SR
Lambda interferon (IFN-lambda), a type III IFN, is induced by viruses and IFNs and displays potent antiviral activity against select virus infections in vivo.
J Virol
2006
80
9
4501
4509
8
Mordstein
M
Kochs
G
Dumoutier
L
et al
Interferon-lambda contributes to innate immunity of mice against influenza A virus but not against hepatotropic viruses.
PLoS Pathog
2008
4
9
e1000151
9
Robek
MD
Boyd
BS
Chisari
FV
Lambda interferon inhibits hepatitis B and C virus replication.
J Virol
2005
79
6
3851
3854
10
Doyle
SE
Schreckhise
H
Khuu-Duong
K
et al
Interleukin-29 uses a type 1 interferon-like program to promote antiviral responses in human hepatocytes.
Hepatology
2006
44
4
896
906
11
Brand
S
Beigel
F
Olszak
T
et al
IL-28A and IL-29 mediate antiproliferative and antiviral signals in intestinal epithelial cells and murine CMV infection increases colonic IL-28A expression.
Am J Physiol Gastrointest Liver Physiol
2005
289
5
G960
G968
12
Sommereyns
C
Paul
S
Staeheli
P
Michiels
T
IFN-lambda (IFN-lambda) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo.
PLoS Pathog
2008
4
3
e1000017
13
Jordan
WJ
Eskdale
J
Srinivas
S
et al
Human interferon lambda-1 (IFN-lambda1/IL-29) modulates the Th1/Th2 response.
Genes Immun
2007
8
3
254
261
14
Jordan
WJ
Eskdale
J
Boniotto
M
Rodia
M
Kellner
D
Gallagher
G
Modulation of the human cytokine response by interferon lambda-1 (IFN-lambda1/IL-29).
Genes Immun
2007
8
1
13
20
15
Pekarek
V
Srinivas
S
Eskdale
J
Gallagher
G
Interferon lambda-1 (IFN-lambda1/IL-29) induces ELR(-) CXC chemokine mRNA in human peripheral blood mononuclear cells, in an IFN-gamma-independent manner.
Genes Immun
2007
8
2
177
180
16
Srinivas
S
Dai
J
Eskdale
J
Gallagher
GE
Megjugorac
NJ
Gallagher
G
Interferon-lambda1 (interleukin-29) preferentially down-regulates interleukin-13 over other T helper type 2 cytokine responses in vitro.
Immunology
2008
125
4
492
502
17
Dai
J
Megjugorac
NJ
Gallagher
GE
Yu
RY
Gallagher
G
IFN-lambda1 (IL-29) inhibits GATA3 expression and suppresses Th2 responses in human naive and memory T cells.
Blood
2009
113
23
5829
5838
18
Azzawi
M
Bradley
B
Jeffery
PK
et al
Identification of activated T lymphocytes and eosinophils in bronchial biopsies in stable atopic asthma.
Am Rev Respir Dis
1990
142
6 Pt 1
1407
1413
19
Broide
DH
Gleich
GJ
Cuomo
AJ
et al
Evidence of ongoing mast cell and eosinophil degranulation in symptomatic asthma airway.
J Allergy Clin Immunol
1991
88
4
637
648
20
Contoli
M
Message
SD
Laza-Stanca
V
et al
Role of deficient type III interferon-lambda production in asthma exacerbations.
Nat Med
2006
12
9
1023
1026
21
Coccia
EM
Severa
M
Giacomini
E
et al
Viral infection and Toll-like receptor agonists induce a differential expression of type I and lambda interferons in human plasmacytoid and monocyte-derived dendritic cells.
Eur J Immunol
2004
34
3
796
805
22
Osterlund
P
Veckman
V
Siren
J
et al
Gene expression and antiviral activity of alpha/beta interferons and interleukin-29 in virus-infected human myeloid dendritic cells.
J Virol
2005
79
15
9608
9617
23
Wolk
K
Witte
K
Witte
E
et al
Maturing dendritic cells are an important source of IL-29 and IL-20 that may cooperatively increase the innate immunity of keratinocytes.
J Leukoc Biol
2008
83
5
1181
1193
24
Megjugorac
NJ
Gallagher
GE
Gallagher
G
Modulation of human plasmacytoid DC function by IFN-γ1 (IL-29).
J Leukoc Biol
2009
86
6
1359
1363
25
Fitzgerald-Bocarsly
P
Dai
J
Singh
S
Plasmacytoid dendritic cells and type I IFN: 50 years of convergent history.
Cytokine Growth Factor Rev
2008
19
1
3
19
26
Lund
J
Sato
A
Akira
S
Medzhitov
R
Iwasaki
A
Toll-like receptor 9-mediated recognition of Herpes simplex virus-2 by plasmacytoid dendritic cells.
J Exp Med
2003
198
3
513
520
27
Hochrein
H
Schlatter
B
O'Keeffe
M
et al
Herpes simplex virus type-1 induces IFN-alpha production via Toll-like receptor 9-dependent and -independent pathways.
Proc Natl Acad Sci U S A
2004
101
31
11416
11421
28
Krug
A
French
AR
Barchet
W
et al
TLR9-dependent recognition of MCMV by IPC and DC generates coordinated cytokine responses that activate antiviral NK cell function.
Immunity
2004
21
1
107
119
29
Zucchini
N
Bessou
G
Traub
S
et al
Cutting edge: overlapping functions of TLR7 and TLR9 for innate defense against a herpesvirus infection.
J Immunol
2008
180
9
5799
5803
30
Jarrossay
D
Napolitani
G
Colonna
M
Sallusto
F
Lanzavecchia
A
Specialization and complementarity in microbial molecule recognition by human myeloid and plasmacytoid dendritic cells.
Eur J Immunol
2001
31
11
3388
3393
31
Kadowaki
N
Ho
S
Antonenko
S
et al
Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens.
J Exp Med
2001
194
6
863
869
32
Olweus
J
BitMansour
A
Warnke
R
et al
Dendritic cell ontogeny: a human dendritic cell lineage of myeloid origin.
Proc Natl Acad Sci U S A
1997
94
23
12551
12556
33
Wong
HL
Costa
GL
Lotze
MT
Wahl
SM
Interleukin (IL) 4 differentially regulates monocyte IL-1 family gene expression and synthesis in vitro and in vivo.
J Exp Med
1993
177
3
775
781
34
Mosmann
TR
Cherwinski
H
Bond
MW
Giedlin
MA
Coffman
RL
Two types of murine helper T cell clone, I: definition according to profiles of lymphokine activities and secreted proteins.
J Immunol
1986
136
7
2348
2357
35
Nelms
K
Keegan
AD
Zamorano
J
Ryan
JJ
Paul
WE
The IL-4 receptor: signaling mechanisms and biologic functions.
Annu Rev Immunol
1999
17
701
738
36
Hannum
CH
Wilcox
CJ
Arend
WP
et al
Interleukin-1 receptor antagonist activity of a human interleukin-1 inhibitor.
Nature
1990
343
6256
336
340
37
Arend
WP
Malyak
M
Guthridge
CJ
Gabay
C
Interleukin-1 receptor antagonist: role in biology.
Annu Rev Immunol
1998
16
27
55
38
Fenton
MJ
Buras
JA
Donnelly
RP
IL-4 reciprocally regulates IL-1 and IL-1 receptor antagonist expression in human monocytes.
J Immunol
1992
149
4
1283
1288
39
Burns
K
Martinon
F
Esslinger
C
et al
MyD88, an adapter protein involved in interleukin-1 signaling.
J Biol Chem
1998
273
20
12203
12209
40
Wesche
H
Henzel
WJ
Shillinglaw
W
Li
S
Cao
Z
MyD88: an adapter that recruits IRAK to the IL-1 receptor complex.
Immunity
1997
7
6
837
847
41
Janssens
S
Beyaert
R
A universal role for MyD88 in TLR/IL-1R-mediated signaling.
Trends Biochem Sci
2002
27
9
474
482
42
Milone
MC
Fitzgerald-Bocarsly
P
The mannose receptor mediates induction of IFN-alpha in peripheral blood dendritic cells by enveloped RNA and DNA viruses.
J Immunol
1998
161
5
2391
2399
43
Sato
A
Linehan
MM
Iwasaki
A
Dual recognition of herpes simplex viruses by TLR2 and TLR9 in dendritic cells.
Proc Natl Acad Sci U S A
2006
103
46
17343
17348
44
Ho
IC
Vorhees
P
Marin
N
et al
Human GATA-3: a lineage-restricted transcription factor that regulates the expression of the T cell receptor alpha gene.
EMBO J
1991
10
5
1187
1192
45
Oosterwegel
M
Timmerman
J
Leiden
J
Clevers
H
Expression of GATA-3 during lymphocyte differentiation and mouse embryogenesis.
Dev Immunol
1992
3
1
1
11
46
Samson
SI
Richard
O
Tavian
M
et al
GATA-3 promotes maturation, IFN-gamma production, and liver-specific homing of NK cells.
Immunity
2003
19
5
701
711
Sign in via your Institution